You are on page 1of 1977

The Scene of Action 1

Amino Acids, Peptides, and Proteins 39


Determining Structures and Analyzing Cells 95
Sugars, Polysaccharides, and Glycoproteins 161
The Nucleic Acids 199
Thermodynamics and Biochemical Equilibria 281
How Macromolecules Associate 325
Lipids, Membranes, and Cell Coats 379
Enzymes: The Catalysts of Cells 455
An Introduction to Metabolism 505
The Regulation of Enzymatic Activity and Metabolism 535
Transferring Groups by Displacement Reactions 589
Enzymatic Addition, Elimination, Condensation,
and Isomerization: Roles for Enolate and Carbocation
Intermediates 677
Coenzymes: Natures Special Reagents 719
Coenzymes of Oxidation Reduction Reactions 765
Transition Metals in Catalysis and Electron Transport 837
Chapter 1.
Chapter 2.
Chapter 3.
Chapter 4.
Chapter 5.
Chapter 6.
Chapter 7.
Chapter 8.
Chapter 9.
Chapter 10.
Chapter 11.
Chapter 12.
Chapter 13.
Chapter 14.
Chapter 15.
Chapter 16.
Table of Contents
Volume 1
Chapter 17.
Chapter 18.
Chapter 19.
Chapter 20.
Chapter 21.
Chapter 22.
Chapter 23.
Chapter 24.
Chapter 25.
Chapter 26.
Chapter 27.
Chapter 28.
Chapter 29.
Chapter 30.
Chapter 31.
Chapter 32.
The Organization of Metabolism 938
Electron Transport, Oxidative Phosphorylation,
and Hydroxylation 1012
The Chemistry of Movement 1088
Some Pathways of Carbohydrate Metabolism 1128
Specific Aspects of Lipid Metabolism 1180
Polyprenyl (Isoprenoid) Compounds 1226
Light and Life 1272
The Metabolism of Nitrogen and Amino Acids 1358
Metabolism of Aromatic Compounds and Nucleic
Acid Bases 1420
Biochemical Genetics 1472
Organization, Replication, Transposition, and Repair
of DNA 1528
The Transcription of Genes 1602
Ribosomes and the Synthesis of Proteins 1668
Chemical Communication Between Cells 1740
Biochemical Defense Mechanisms 1830
Growth and Development 1878
Table of Contents
Volume 2
Front matter Vol 2 2/14/03, 3:03 PM 10
Contents
A. The Simplest Living Things
1. Escherichia coli
2. The Bacterial Genome
3. Ribonucleic Acids (RNA) and the Transcription
and Translation of Genetic Information
4. Membranes and Cell Walls
5. Flagella and Pili
6. Classification and Evolution of Bacteria
7. Nutrition and Growth of Bacteria
8. Photosynthetic and Nitrogen-Fixing Prokaryotes
B. Eukaryotic Cells
1. The Nucleus
2. The Plasma Membrane
3. Vacuoles, Endocytosis, and Lysosomes
4. The Endoplasmic Reticulum and Golgi Membranes
5. Mitochondria, Plastids, and Peroxisomes
6. Centrioles, Cilia, Flagella, and Microtubules
7. Cell Coats, Walls, and Shells
C. Inheritance, Metabolic Variation, and Evolution of
Eukaryotes
1. A Changing Genome
2. Genetic Recombination, Sex, and Chromosomes
3. Haploid and Diploid Phases
D. Survey of the Protists
1. Protozoa
2. Fungi
3. Algae
E. The Variety of Animal Forms
1. Major Groups of Multicellular Animals
2. Cell Types and Tissues
Tissues
Blood cells
Cell culture
3. Communication
Cell contacts and junctions
Cell recognition
F. Higher Plants and Plant Tissues
G. The Chemical Composition of Cells
References
Study Questions
Boxes
Box 1-A About Measurements
Box 1-B Relative Molecular Mass, M
r
, and Daltons
Box 1-C In the Beginning
Box 1-D Inherited Metabolic Diseases
Box 1-E Errors, Misconceptions, and Speculation
Box 1-F About the References
Tables
Table 1-1 A Systematic Classification Scheme for
Bacteria
Table 1-2 Approximate Sizes of Some Cells
Table 1-3 Haploid Genome Sizes for Several
Organisms
Table 1-4 Approximate Composition of
Metabolically Active Cells and Tissues
2
4
9
16
25
32
7
11
12
31
2
3
3
5
5
6
6
8
9
11
11
11
12
13
14
15
15
15
17
17
18
18
18
20
20
23
23
25
26
26
26
26
28
29
29
30
34
36
Left: Cells of the pathogenic O157:H7 strain of Escherichia coli attached to the surface epithelium of the
cecum of a neonatal piglet. Electron-dense filaments (presumably polymerized actin) in the host
cytoplasm can be seen subjacent to attached bacteria. The bacteria have effaced most micro-villi but
some remain between the bacterial cells. Courtesy of Evelyn A. Dean-Nystrom, National Animal
Disease Center, USDA, Agricultural Research Service, Ames, IA. Center: Many unicellular organisms
such as these Vorticella inhabit wet and moist environments throughout the biosphere. Invertebrates
have evolved as long as humans and have complex specializations such as the contractile stem of these
protozoa. Courtesy of Ralph Buchsbaum. Right: Although 97% of animals are invertebrates, ~ 3%
of the several million known species have backbones. Giraffe: M. P. Kahl, Photo Researchers
1
This book is about the chemistry of living cells
with special emphasis on the trillions of cells that make
up your own body. Every aspect of life depends upon
the chemical makeup of cells and on the chemical
properties of the remarkable molecules found within
the cells. The information presented here will give
the reader a solid foundation for understanding not
only the chemical basis of life but also the revolution-
ary developments in molecular biology, biochemical
genetics, medicine, and agriculture which dominate
todays scientific news and which will play an increas-
ingly important role in our lives.
The first theme of the book is biomolecular
structure. Well look carefully at the complex struc-
tures of proteins, carbohydrates, RNA, DNA, and
many other substances. Well not only examine in-
depth their molecular architecture but also study the
chemical properties that make life possible.
A second theme is metabolism, the unceasing,
complex network of thousands of chemical reactions
by which cells grow and reproduce, take up foods and
excrete wastes, move, and communicate with each
other. Within cells we have a steady state, a condi-
tion in which the complex chemical constituents of
cells are continuously being synthesized in one series
of reactions and degraded in another. The result is a
marvelous system of self-renewal or turnover of
tissues. Well examine the chemical reactions involved
in these processes as well as the ways in which they
are controlled. We will consider both the reaction
sequences and the techniques such as cloning of genes,
isotopic labeling, X-ray diffraction, and nuclear mag-
netic resonance spectroscopy, which are used today to
study metabolism.
Human beings are surrounded by many other living
creatures whose activities are important to us. Photo-
synthetic organisms obtain energy from sunlight and
synthesize compounds that the human body requires
but cannot make. Microorganisms cause decay of
organic matter and convert it into forms usable by
plants. This book deals with the chemical reactions
occurring in all of these organisms. Well look at
strange and unusual reactions, along with those meta-
bolic sequences common to most living things.
Each one of the thousands of chemical reactions
of metabolism is catalyzed by an enzyme. Most of
these enzymes are proteins, but others are made from
RNA (ribonucleic acid). In both cases enzymes are
very large molecules with precise three-dimensional
structures. The study of the properties of enzymes
and of enzymatic catalysis is a third theme of the
book. Not only are the chemical mechanisms by
which enzymes act of interest but also enzymes are
often targets for useful drugs. Incorrectly formed en-
zymes can result in serious diseases.
The sequences of the amino acids in the chains
from which proteins are constructed are encoded in
the nucleotide sequences of DNA (deoxyribonucleic
acid). The coding sequence for a protein in the DNA
is found in the structural gene for that protein. The
RNA enzymes are also encoded by DNA genes. A
fourth major theme of the book deals with the nature
of the genetic code used in DNA and with the pro-
cesses by which cells read and interpret the code. It
also includes study of the methods by which thousands
of genes have been mapped to specific positions in
chromosomes, isolated, cloned, and sequenced.
A large number of proteins present in the outer
surfaces of cells serve as receptors that receive chemi-
cal messages and other signals from outside the cell.
The receptors, which are sometimes enzymes, respond
by generating internal signals that control metabolism
and cell growth. Such molecular signaling is another
major area of contemporary biochemistry.
The Scene of Action
1
2 Chapter 1. The Scene of Action
BOX 1-A ABOUT MEASUREMENTS
Biologists have described over a million species,
and several millions of others probably exist.
1
Many
of these organisms have very specialized ways of life.
However, they all have much chemistry in common.
The same 20 amino acids can be isolated from proteins
of plants, animals, and microorganisms. Formation of
lactic acid in both bacteria and human muscle requires
the same enzymes. Except for some small variations,
the genetic code is universalthe same for all organ-
isms. Thus, there is a unity of life and we can study
metabolism as the entirety of chemical transformations
going on in all living things. However, the differences
among species are also impressive. Each species has
its own gene for almost every protein.
When the enzyme that catalyzes a particular meta-
bolic reaction is isolated from a number of different
organisms, it is usually found to have similar proper-
ties and a similar mechanism of catalysis, regardless of
the source. However, the exact sequence of amino acids
in the enzyme will be almost unique to the organism
that produced it. When the three-dimensional struc-
tures are compared it is found that differences between
species often affect only the peripheral parts of an
enzyme molecule. The interior structure of the protein,
including the catalytic machinery, is highly conserved.
However, the surface regions, which often interact with
other macromolecules, vary greatly. Such interactions
help to control metabolism and may account for many
differences in the metabolism among living beings.
Variations in protein structures are not limited to
differences between species. Individuals differ from
one another. Serious genetic diseases sometimes result
from the replacement of a single amino acid unit in a
protein by a different amino acid. Genetic deviations
from the normal structure of a protein result from
mutations. Many mutations, whether they occurred
initially in our own cells or in those of our ancestors,
are detrimental.
However, such mutations also account for variation
among individuals of a species and allow for evolution.
The chemical nature and consequences of mutations
and their significance to health, medicine, and agricul-
ture are dealt with throughout the book. We now have
reliable methods for inducing in the laboratory muta-
tions at any specific place in a protein sequence and
also for synthesizing new DNA sequences. These
techniques of genetic engineering have given bio-
chemists the ability to modify protein structures freely,
to create entirely new proteins, and to provide a basis
for the rapidly developing field of genetic therapy.
It should be clear from this introduction that
biochemistry deals with virtually every aspect of life.
The distinguishing feature of the science is that it
approaches biological questions in terms of the under-
lying chemistry. The term molecular biology is often
regarded as synonymous with biochemistry.
However, some scientists use it to imply a more
biological approach. These molecular biologists also
emphasize structure and function but may have a goal
of understanding biological relationships more than
chemical details. Biophysics, a closely related science,
encompasses the application of physical and mathe-
matical tools to the study of life.
A. The Simplest Living Things
The simplest organisms are the bacteria.
25
Their
cells are called prokaryotic (or procaryotic) because
no membrane-enclosed nucleus is present. Cells of all
other organisms contain nuclei separated from the
cytoplasm by membranes. They are called eukaryotic.
While viruses (Chapter 5) are sometimes regarded as
living beings, these amazing parasitic objects are not
complete organisms and have little or no metabolism of
their own. The smallest bacteria are the mycoplasmas.
68
They do not have the rigid cell wall characteristic of
most bacteria. For this reason they are easily deformed
and often pass through filters designed to stop bacteria.
They are nutritionally fussy and are usually, if not
always, parasitic. Some live harmlessly in mucous
membranes of people, but others cause diseases.
In 1960 the International General Conference
on Weights and Measures adopted an improved
form of the metric system, The International
System of Units (SI). The units of mass, length,
and time are the kilogram (kg), meter (m), and
second (s). The following prefixes are used for
fractions and multiples:
10
18
, atto (a) 10
6
, micro () 10
9
, giga (G)
10
15
, femto (f) 10
3
, milli (m) 10
12
, tera (T)
10
12
, pico (p) 10
3
, kilo (k) 10
15
, peta (P)
10
9
, nano (n) 10
6
, mega (M) 10
18
, exa (E)
There is an inconsistency in that the prefixes are
applied to the gram (g) rather than to the basic
unit, the kilogram.
SI units have been used throughout the book
whenever possible. There are no feet, microns,
miles, or tons. Molecular dimensions are given
uniformly in nanometers rather than in angstrom
units (; 1 = 0.1 nm). Likewise the calorie and
kilocalorie have been replaced by the SI unit of
energy, the joule (J; 1 calorie = 4.184 J).
Throughout the book frequent use is made of
the following symbols:
, appr oximately equal to
~, approximately or about
3
For example, Mycoplasma pneumoniae is responsible for
primary atypical pneumonia.
Cells of mycoplasmas sometimes grow as filaments
but are often spherical and as small as 0.3 micrometer
(m) in diameter. Their outer surface consists of a thin
cell membrane about 8 nanometers (nm) thick. This
membrane encloses the cytoplasm, a fluid material
containing many dissolved substances as well as sub-
microscopic particles. At the center of each cell is a
single, highly folded molecule of DNA, which consti-
tutes the bacterial chromosome. Besides the DNA
there may be, in a small spherical mycoplasma, about
1000 particles ~20 nm in diameter, the ribosomes.
These ribosomes are the centers of protein synthesis.
Included in the cytoplasm are many different kinds of
A pilus. Some E. coli are covered with
hundreds of pili of various lengths
Cell membrane, ~8 nm
Cell wall, ~10 nm
E. coli
~0.8 0.8 2.0 m
DNA, 1.4 mm long.
Only 1% of the total
is drawn here
1314 nm diameter
The lengths of the flagella
vary but are often ~4 longer
than the cell proper
Sheathed
flagellum,
28 nm
A small
mycoplasma
Some strains of E. coli
have flagellaas many
as 8, but often fewer
Ribosomes attached
to thread of mRNA
Ribosomes
~20 nm diameter
0.5 m
Bdellovibrio, a parasite that lives
within E. coli [see picture by J.C.
Burnham, T. Hashimoto, and S.F.
Conti, J. Bacteriol. 96, 1366 (1968)]
Figure 1-1 Escherichia coli and some smaller bacteria.
proteins, but there is room for a total of only about
50,000 protein molecules. Several types of RNA as
well as many smaller molecules are also present.
Although we dont know what minimum quantities
of proteins, DNA, and other materials are needed to
make a living cell, it is clear that they must all fit into
the tiny cell of the mycoplasma.
1. Escherichia coli
The biochemists best friend is Escherichia coli, an
ordinarily harmless inhabitant of our intestinal tract.
This bacterium is easy to grow in the laboratory and
has become the best understood organism at the mo-
lecular level.
4,9
It may be regarded as a
typical true bacterium or eubacterium.
The cell of E. coli (Figs. 1-1, 1-2) is a
rod ~2 m long and 0.8 m in diameter
with a volume of ~1 m
3
and a density
of ~1.1 g/cm
3
. The mass is ~1 x 10
12
g,
i.e., 1 picogram (pg) or ~0.7 x 10
12
daltons (Da) (see Box 1-B).
4
It is about
100 times bigger than the smallest
mycoplasma but the internal structure,
as revealed by the electron microscope,
resembles that of a mycoplasma.
Each cell of E. coli contains from
one to four identical DNA molecules,
depending upon how fast the cell is
growing, and ~15,00030,000 ribosomes.
Other particles that are sometimes seen
within bacteria include food stores such
as fat droplets and granules (Fig. 1-3).
The granules often consist of poly--
hydroxybutyric acid
10
accounting
for up to 25% of the weight of Bacillus
megaterium. Polymetaphosphate, a
highly polymerized phosphoric acid,
is sometimes stored in metachromatic
granules. In addition, there may be
droplets of a separate aqueous phase,
known as vacuoles.
2. The Bacterial Genome
The genetic instructions for a cell
are found in the DNA molecules. All
DNA is derived from four different
kinds of monomers, which we call
nucleotides. DNA molecules are
double-stranded: two polymer chains
are coiled together, their nucleotide
units being associated as nucleotide
pairs (see Fig. 5-7). The genetic mes-
sages in the DNA are in the form of
A. The Simplest Living Things
4 Chapter 1. The Scene of Action
sequences of nucleotides. These sequences usually
consist of a series of code words or codons. Each
codon is composed of three successive nucleotides and
specifies which one of the 20 different kinds of amino
acids will be used at a particular location in a protein.
The sequence of codons in the DNA tells a cell how to
order the amino acids for construction of its many
different proteins.
Figure 1-2 Transmission electron micrograph of a dividing
cell of Escherichia coli O157:H7 attached to the intestinal
epithelium of a neonatal calf. These bacteria, which are able
to efface the intestinal microvilli, form characteristic attach-
ments, and secrete shiga toxins, are responsible for ~73,000
illnesses and 60 deaths per year in the U. S.
10a
After embed-
ding, the glutaraldehyde-fixed tissue section was immuno-
stained with goat anti-O157 IgG followed by protein A con-
jugated to 10-nm gold particles. These are seen around the
periphery of the cell bound to the O-antigen (see Fig. 8-28).
Notice the two microvilli of the epithelium. Courtesy of
Evelyn A. Dean-Nystrom, National Animal Disease Center,
USDA, Agricultural Research Service, Ames, IA.
Figure 1-3 A cell of a Spirillum negatively stained with
phosphotungstic acid. Note the tufts of flagella at the ends,
the rough appearance of the outer surface, the dark granules
of poly--hydroxybutyric acid and the light-colored gran-
ules of unknown nature. Courtesy of F. D. Williams, Gail E.
VanderMolen, and C. F. Amstein.
Assume that a typical protein molecule consists of
a folded chain of 400 amino acids. Its structural gene
will therefore be a sequence of 1200 nucleotide pairs.
Allowing a few more nucleotides to form spacer regions
between genes we can take ~1300 as the number of
nucleotide pairs in a typical bacterial gene. However,
some genes may be longer and some may be much
shorter. The genome is the quantity of DNA that carries
a complete set of genetic instructions for an organism.
In bacteria, the genome is a single chromosome con-
sisting of one double-stranded DNA molecule. Myco-
plasma genitalium is the smallest organism for which
the DNA sequence is known.
11
Its genome is a double-
helical DNA circle of 580,070 nucleotide pairs and
appears to contain about 480 genes (an average of ~1200
nucleotides per gene).
The average mass of a nucleotide pair (as the
disodium salt) is 664 Da. It follows that the DNA of
M. genitalium has a mass of ~385 x 10
6
Da. The relative
molecular mass (M
r
) is 0.385 x 10
9
(See Box 1-B for
definitions of dalton and M
r
). The DNA of E. coli is
about seven times larger with a mass of ~2.7 x 10
9
Da.
It contains ~4.2 x 10
6
nucleotide pairs and encodes
over 4000 different proteins (see Table 1-3).
Each nucleotide pair contributes 0.34 nm to the
length of the DNA molecule; thus, the total length of
DNA of an E. coli chromosome is 1.4 mm. This is
about 700 times the length of the cell which contains it.
Clearly, the molecules of DNA are highly folded, a fact
that accounts for their appearance in the electron micro-
scope as dense aggregates called nucleoids, which
occupy about one-fifth of the cell volume (Fig. 1-4).
Atomic and molecular masses are assigned
relative to the mass of the carbon isotope,
12
C,
whose atomic weight is defined as exactly 12.
The actual mass of a single atom of
12
C is defined
as 12 daltons, one dalton being 1.661 x 10
24
g.
The mass of a molecule can be given in daltons
(Da) or kilodaltons (kDa). This molecular mass
in daltons is numerically equivalent to the relative
molecular mass (M
r
) or molecular weight (MW)
a
and also to the molar mass (g/mol). However, it
is not correct to use the dalton for the unitless
quantity M
r
. Masses of structures such as chro-
mosomes, ribosomes, mitochondria, viruses, and
whole cells as well as macromolecules can be
given in daltons.
b
a
The Union of Pure and Applied Chemistry renamed
molecular weight as relative molecular mass with the
symbol M
r
; M
r
= MW.
b
J. T. Edsall (1970) Nature (London) 228, 888.
BOX 1-B RELATIVE MOLECULAR MASS,
M
r
, AND DALTONS
1 m
1 m
5 A. The Simplest Living Things
Each bacterial nucleoid contains a complete set of
genetic blueprints and functions independently.
Each nucleoid is haploid, meaning that it contains
only a single complete set of genes. In addition to
their chromosome, bacteria often contain smaller
DNA molecules known as plasmids. These plasmids
also carry genetic information that may be useful to
bacteria. For example, they often encode proteins that
confer resistance to antibiotics. The ability to acquire
new genes from plasmids is one mechanism that
allows bacteria to adapt readily to new environments.
12
Plasmids are also used in the laboratory in the cloning
of genes and in genetic engineering (Chapter 26).
3. Ribonucleic Acids (RNA) and the
Transcription and Translation of Genetic
Information
The genetic information in the DNA is not utilized
directly by the protein-synthesizing machinery of cells.
Instead, molecules of ribonucleic acid (RNA) are syn-
thesized according to the instructions encoded in the
DNA, a process called transcription. Although they
differ from DNA significantly in their structure, these
RNA molecules carry the same coded information as
is found in a length of DNA that contains one or a few
genes. If DNA is regarded as the master blueprint of
the cell, molecules of RNA are secondary blueprints.
This concept is embodied in the name messenger
RNA (mRNA) which is applied to a small, short-lived
fraction of RNA that carries information specifying
amino acid sequences of proteins. Each molecule of
mRNA carries the genetic message from one or more
genes to the ribosomes where the proteins are made.
Ribosomes are extraordinarily complex little
protein-synthesizing machines. Each ribosome of
E. coli has a mass of 2.7 x 10
6
Da and contains 65% of
a stable ribosomal RNA and ~35% protein. About
50 different kinds of protein molecules are present as
parts of the ribosomal structure. Working together
with a variety of transfer RNA molecules and enzymes,
the ribosomes are able to read the genetic messages
from mRNA and to accurately assemble any kind of
protein molecule that a gene may specify. This process
is called translation of the genetic message.
4. Membranes and Cell Walls
Like the mycoplasma, the E. coli cell is bounded
by an 8-nm membrane which consists of ~50% protein
and 50% lipid. When stained (e.g., with perman-
ganate) for electron microscopy, this single membrane
appears as two very thin (2.0 nm) dark lines separated
by an unstained center band (~3.5 nm) (Fig. 1-4; see
also Fig. 8-4). Single membranes of approximately the
same thickness and staining behavior occur in all cells,
both of bacteria and of eukaryotes.
A cell membrane is much more than just a sack.
It serves to control the passage of small molecules into
and out of the cell. Its outer surface carries receptors
for recognition of various materials. The inside surface
of bacterial membranes contains enzymes that catalyze
most of the oxidative metabolism of the cells. Bacterial
cell membranes are sometimes folded inward to form
internal structures involved in photosynthesis or other
specialized reactions of metabolism such as oxidation
of ammonia to nitrate.
2
In E. coli replication of DNA
seems to occur on certain parts of the membrane sur-
face, probably under the control of membrane-bound
enzymes. The formation of the new membrane which
Figure 1-4 (A) Thin (~60 nm) section of an aquatic gram-
negative bacterium, Aquaspirillum fasciculus. Note the light-
colored DNA, the dark ribosomes, the double membrane
characteristics of gram-negative bacteria (Chapter 8, Section
E), and the cell wall. In addition, an internal polar mem-
brane is seen at the end. It may be involved in some way in
the action of the flagella. (B) A thin section of dividing cell
of Streptococcus, a gram-positive organism. Note the DNA
(light-stranded material). A portion of a mesosome is seen
in the center and septum can be seen forming between the
cells. Micrographs courtesy of F. D. Williams, Gail E.
VanderMolen, and C. F. Amstein.
B
0.25 m
A
6 Chapter 1. The Scene of Action
divides multiplying cells proceeds synchronously with
the synthesis of DNA.
A characteristic of true bacteria (eubacteria) is a
rigid cell wall which surrounds the cell membrane.
The 40-nm-thick wall of E. coli is a complex, layered
structure five times thicker than the cell membrane.
Its chemical makeup is considered in Chapter 8. One
of the layers is often referred to as the outer mem-
brane. In some bacteria the wall may be as much as
80 nm thick and may be further surrounded by a thick
capsule or glycocalyx (slime layer).
13
The main
function of the wall seems to be to prevent osmotic
swelling and bursting of the bacterial cell when the
surrounding medium is hypotonic.
If the osmotic pressure of the medium is not too
low, bacterial cell walls can sometimes be dissolved,
leaving living cells bounded only by their membranes.
Such protoplasts can be produced by action of the
enzyme lysozyme on gram-positive bacteria such as
Bacillus megaterium. Treatment of cells of gram-negative
bacteria with penicillin (Box 20-G) produces sphero-
plasts, cells with partially disrupted walls. Sphero-
plasts and protoplasts are useful in biochemical studies
because substances enter cells more readily when the
cell wall is absent. Strains of bacteria lacking rigid
walls are known as L forms.
5. Flagella and Pili
Many bacteria swim at speeds of 2060 m/s, ten
or more body lengths per second! Very thin thread-
like flagella of diameter 1320 nm coiled into a helical
form are rotated by the worlds smallest electric
motors to provide the motion.
14
While some bacteria
have a single flagellum, the corkscrew-like Spirillum
(Fig. 1-3) synchronously moves tufts of flagella at both
ends. Some strains of E. coli have no flagella, but
others contain as many as eight flagella per cell dis-
tributed over the surface. The flagella stream out
behind in a bundle when the bacterium swims. The
flagella of the helical spirochetes are located inside
the outer membrane.
15,16
In addition to flagella, extremely thin, long,
straight filaments known as pili or fimbriae (Fig. 1-2)
project from the surfaces of many bacteria.
14
The sex
pili (F pili and I pili) of E. coli have a specific role in
sexual conjugation. The similar but more numerous
common pili or fimbriae range in thickness from 3 to
25 nm and in length from 0.2 to 2 m. Pili are involved
in adhesion of bacteria to surrounding materials or to
other bacteria and facilitate bacterial infections.
1719
A typical E. coli cell has 100300 pili.
5
6. Classification and Evolution of Bacteria
Bacteria vary greatly in their chemistry and meta-
bolism, and it is difficult to classify them in a rational
way. In higher organisms species are often defined as
forms that cannot interbreed and produce fertile off-
spring, but such a criterion is meaningless for bacteria
whose reproduction is largely asexual and which are
able readily to accept visiting genes from other
bacteria.
12
The classification into species and genera
is therefore somewhat arbitrary. A currently used
scheme (Table 1-1)
20
classifies the prokaryotes into 35
groups on the basis of many characteristics including
shape, staining behavior, and chemical activities. Table
1-1 also includes genus names of most of the bacteria
discussed in this book.
Bacteria may have the shape of spheres or straight
or curved rods. Some, such as the actinomycetes,
grow in a branching filamentous form. Words used to
describe bacteria often refer to these shapes: a coccus
is a sphere, a bacillus a rod, and a vibrio a curved rod
with a flagellum at one end. A spirillum is screw-
shaped. These same words are frequently used to
name particular genera or families. Other names are
derived from some chemical activity of the bacterium
being described.
The gram stain provides an important criterion
of classification that depends upon differences in the
structure of the cell wall (see Chapter 20). Bacterial
cells are described as gram-positive or gram-negative
according to their ability to retain the basic dye crystal
violet as an iodine complex. This difference distinguishes
two of four large categories of bacteria.
20
Most actino-
mycetes, the spore-forming bacilli, and most cocci are
gram-positive, while E. coli, other enterobacteria, and
pseudomonads are gram-negative. A third category
consists of eubacteria that lack cell walls, e.g. the
mycoplasma.
Comparisons of amino acid sequences of proteins
and the nucleotide sequences of DNA and RNA have
provided a new approach to classification of bacteria.
2128
Although the origins of life are obscure, we can easily
observe that the genome changes with time through
mutation and through the enzyme-catalyzed process
of genetic recombination. The latter gives rise to the
deletion of some nucleotides and the insertion of others
into a DNA chain. When we examine sequences of
closely related species, such as E. coli and Salmonella
typhimurium, we find that the sequences are very similar.
However, they differ greatly from those of many other
bacteria. Consider the 23S ribosomal RNA, a molecule
found in the ribosomes of all bacteria. It contains ~3300
nucleotides in a single highly folded chain. The basic
structure is highly conserved but between any two
species of bacteria there are many nucleotide substitu-
tions caused by mutations as well as deletions and
insertions. By asking what is the minimum number of
7 A. The Simplest Living Things
TABLE 1-1
A Systematic Classification Scheme for Bacteria
a,b
Kingdom Procaryotae
The bacteria are classified according to the following 35
groups. Within these groups many genera are classified into
subgroups or families. A few genera, most of which are
mentioned elsewhere in this book, are listed here by name.
Members of a single subgroup are placed together and are
separated by semicolons from members of other subgroups.
1. The spirochetes (long bacteria, up to 500 m, that are
propelled by the action of filaments wrapped around
the cell between the membrane and wall). Borrelia (B.
burgdorferi, Lyme disease), Leptospira, Treponema (T.
pallidum, syphilis)
2. Aerobic spiral and curved motile gram-negative
bacteria. Bdellovibrio, Campylobacter (C. jejuni, diarrhea),
Helicobacter (H. pylori, gastric ulcers), Spirillum
3. Nonmotile gram-negative curved bacteria
4. Gram-negative, aerobic rods and cocci.
Acetobacter, Agrobacterium, Azotobacter, Brucella
(B. abortus, brucellosis), Flavobacterium, Gluconobacter,
Legionella (L. pneumophila, Legionnaires disease),
Methylomonas, Neisseria (N. gonorrhea, gonorrhea),
Pseudomonas, Rhizobium, Thermus, Xanthomonas,
Rochalimaea (R. henselae, cat scratch disease)
5. Gram-negative, facultatively anaerobic rods.
Enterobacter,
c
Proteus, Yersinia (Y. pestis, plague), Escheri-
chia, Klebsiella, Salmonella (S. typhi, typhoid fever),
Serratia, Shigella (S. dysenteriae, bacterial dysentery),
Haemophilus; Vibrio (V. cholerae, Asiatic cholera);
Zymomonas
6. Gram-negative, anaerobic bacteria. Butyrivibrio
7. Dissimilatory sulfate- or sulfur-reducing bacteria.
Desulfovibrio
8. Anaerobic gram-negative cocci. Veillonella
9. The rickettsias (parasitic bacteria with exacting nutri-
tional requirements and small genome sizes) and
chlamydias. Chlamydia (C. trachomatis, trachoma),
Rickettsia (R. rickettsii, Rocky Mountain spotted fever)
10. Anoxygenic photosynthetic bacteria. Green sulfur
bacteria. Chlorobium, Prosthecochloris; purple nonsulfur
bacteria: Rhodopseudomonas, Rhodospirillum; purple
sulfur bacteria: Chromatium, Thiospirillum
a
From Bergeys Manual of Systematic Bacteriology, 9th ed. J. G. Holt, N. R. Krieg, P. H. A. Sneath, J. T. Staley and S. T. Williams, Eds. (1994)
Williams and Wilkins, Baltimore, Maryland. For another recent list see http://www.ncbi.nlm.nih.gov/
b
The human diseases caused by some species are also listed.
c
Formerly Aerobacter.
11. Oxygenic photosynthetic bacteria. Cyanobacteria (blue-
green algae): Synechocystis; Anabaena, Nostoc; Oscillatoria
12. Aerobic, chemolithotrophic bacteria. Colorless sulfur
bacteria: Thiobacillus; iron or manganese-oxidizing
bacteria, magnetotactic bacteria; nitrifying bacteria:
Nitrobacter, Nitrosomonas
13. Budding and/or appendaged bacteria. Caulobacter
14. Sheathed bacteria
15. Nonphotosynthetic, nonfruiting gliding bacteria.
Beggiatoa (a filamentous bacterium containing sulfur
granules)
16. Fruiting, gliding bacteria. Myxococcus
17. Gram-positive cocci. Leuconostoc, Micrococcus,
Peptococcus, Staphylococcus (S. aureus, boils, infections),
Streptococcus (S. pyogenes, scarlet fever, throat infections,
S. pneumoniae, pneumonia)
18. Endospore-forming gram-positive rods and cocci.
Aerobic: Bacillus (B. anthracis, anthrax), anaerobic:
Clostridium (C. tetani, tetanus; C. botulinum,
botulism)
19. Regular nonsporing gram-positive rods.
Lactobacillus
20. Irregular nonsporing gram-positive rods.
Actinomyces, Bifidobacterium, Corynebacterium
(C. diphtheriae, diphtheria), Propionibacterium
21. Mycobacteria. Mycobacterium (M. tuberculosis,
tuberculosis; M. leprae, leprosy)
22-29. Actinomycetes
30. Mycoplasmas. Acholeplasma, Mycoplasma
31. Methanogens. Methanobacterium; Methanosarcina;
Methanospirillum
32. Archaeal sulfate reducers
33. Halobacteria. Halobacterium
34. Cell wall-less archaeobacteria. Thermoplasma
35. Very thermophilic S
0
-Metabolizers. Sulfolobus;
Thermococcus
mutations that could have converted one 23S RNA
into another and by assuming a more or less constant
rate of mutation over millions of years it is possible to
construct a phylogenetic tree such as that shown in
Fig. 1-5.
One conclusion from these comparisons is that the
methane-producing bacteria, the methanogens,
24
are
only distantly related to most other bacteria. Methano-
gens together with the cell wall-less Thermoplasma,
28
some salt-loving halobacteria, and some thermo-
philic (heat-loving) sulfur bacteria form a fourth major
category. They are often regarded as a separate king-
dom, the archaeobacteria,
25
which together with the
kingdom of the eubacteria form the superkingdom
prokaryota. Certain archaeobacteria have biochemical
characteristics resembling those of eukaryotes and
8 Chapter 1. The Scene of Action
some biologists therefore classify them as archaea and
rank their kingdom as equal to that of the bacteria and
the eukaryotes (Fig. 1-5).
27,29,30,30a,30b
Others disagree.
31
In Table 1-1, the archaeobacteria are found in groups
3135. Most bacteria are very small in size but there are
species large enough to be confused with eukaryotic
protozoa. The record for bacteria seems to be held by
Epulopiscium fishelsoni, a parasite of the surgeonfish
intestinal tract. A single cell measured > 600 m by
80 m diameter, over 10
6
times larger in volume than
a cell of E. coli.
32
The organism is a gram-positive
bacterium as judged by analysis of its cloned ribo-
somal RNA genes.
7. Nutrition and Growth of Bacteria
Autotrophic (self-nourishing) bacteria can synthe-
size all of their organic cell constituents from carbon
dioxide, water, and inorganic forms of nitrogen and
sulfur. The photoautotrophs extract their energy
from sunlight, while the chemoautotrophs obtain
energy from inorganic chemical reactions. For example,
the hydrogen bacteria oxidize H
2
to H
2
O and sulfur
bacteria oxidize H
2
S to H
2
SO
4
. Like the fungi and
animals, most bacteria are chemoheterotrophic; they
obtain energy from the breakdown of organic com-
pounds. Some of these heterotrophic bacteria are
anaerobes which live without O
2
. Many of them
metabolize complex organic substances such as sugars
in the absence of oxygen, a process called fermentation.
Others oxidize organic compounds with an inorganic
oxidant such as nitrite or sulfate. Members of the
genus Clostridium are poisoned by oxygen and are
known as obligate anaerobes. Others, including
E. coli, are facultative anaerobes, able to grow either
Figure 1-5 Universal phylogenetic tree. From Wheelis et
29
Euryarchaeota
Cren-
archaeota
Thermotogales
Flavobacteria
Cyanobacteria
Purple bacteria
Gram positives
Green non-sulfur bacteria
Pyrodictium
Thermoproteus
T. celer
Methano-
coccus
Methanobacterium
Methanosarcina
Halophiles
Entamoebae
Slime
molds
Animals
Fungi
Plants
Ciliates
Flagellates
Trichomonads
Microsporidia
Diplomonads
Bacteria
Archaea
Eucharya
in the presence or in the absence of oxygen. Obligate
aerobes depend for energy upon combustion of or-
ganic compounds with oxygen.
One of the largest groups of strictly aerobic hetero-
trophic bacteria, the pseudomonads (Pseudomonas and
related genera), are of interest to biochemists because
of their ability to oxidize organic compounds, such as
alkanes, aromatic hydrocarbons, and steroids, which
are not attacked by most other bacteria. Often, the
number of oxidative reactions used by any one species
of bacteria is limited. For example, the acetic acid
bacteria that live in wine and beer obtain all of their
energy by oxidation of ethanol to acetic acid:
CH
3
CH
2
OH + O
2
CH
3
COOH + H
2
O
Bacteria can grow incredibly fast. Under some
conditions, it takes a bacterial cell only 1020 min to
double its size and to divide to form two cells.
4
An
animal cell may take 24 h for the same process. Equally
impressive are the rates at which bacteria transform
their foods into other materials. One factor contributing
to the high rate of bacterial metabolism may be the
large surface to volume ratio. For a small spherical
bacterium (coccus) of diameter 0.5 m, the ratio of the
surface area to the volume is 12 x 10
6
m
1
, while for an
ameba of diameter 150 m the ratio is only 4 x 10
4
m
1
(the ameba can increase this by sticking out some
pseudopods). Thimann
33
estimated that for a 90-kg
human, the ratio is only 30 m
1
.
When food is limited, some bacteria such as the
Bacillus form spores. These are compact little cells
that form inside the vegetative cell and are therefore
called endospores. They sometimes have only 1/10
the volume of the parent cell. Their water content is
very low, their metabolic rate is near zero, and they are
al.
9
extremely resistant to heat and further desiccation.
Under suitable conditions, the spores can germinate
and renew their vegetative growth. Spore formation is
one of several examples of the development of special-
ized cells or differentiation among prokaryotes.
8. Photosynthetic and Nitrogen-Fixing
Prokaryotes
It is likely that the earth was once a completely
anaerobic place containing water, ammonia, methane,
formaldehyde, and more complicated organic com-
pounds. Perhaps the first forms of life, which may
have originated about 3.5 x 10
9
years ago, resembled
present-day anaerobic bacteria. The purple and green
photosynthetic bacteria may be related to organisms
that developed at a second stage of evolution: those
able to capture energy from sunlight. Most of these
gram-negative photosynthetic bacteria are strict anaer-
obes. None can make oxygen as do higher plants.
Rather, the hydrogen needed to carry out the reduction
of carbon dioxide in the photosynthetic process is
obtained by the splitting of inorganic compounds, such
as H
2
S, thiosulfate, or H
2
, or is taken from organic
compounds. Today, photosynthetic bacteria are found
principally in sulfur springs and in deep lakes, but at
one time they were probably far more abundant and
the only photosynthetic organisms on earth.
Before organisms could produce oxygen a second
complete photosynthetic system, which could cleave
H
2
O to O
2
, had to be developed. The simplest oxygen-
No one knows how life began. Theories ranging
from the biblical accounts to recent ideas about the
role of RNA are plentiful but largely unsatisfying. In
the 1800s the great physical chemist Arrhenius was
among scientists that preferred the idea held by
some scientists today that a seed came from outer
space. Until recently the only concrete data came
from fossils. Making use of a variety of isotopic
dating methods it can be concluded that cyanobac-
teria were present 2.2 10
9
years ago and eukaryotes
1.4 10
9
years ago. About 0.5 10
9
years ago the
Cambrian explosion led to the appearance of vir-
tually all known animal phyla. Many of these then
became extinct about 0.2 10
9
years ago.
New insights published in 1859
a
were provided
by Charles Darwin. However, his ideas were only
put into a context of biochemical data after 1950 when
sequencing of proteins and later nucleic acids began.
From an astonishingly large library of sequence data
available now we can draw one firm conclusion:
Evolution can be observed;
b
it does involve mutation of
DNA. Comparisons of sequences among many
species allow evolutionary relationships to be
proposed.
c-e
In general these are very similar to
those deduced from the fossil record. They support
the idea that evolution occurs by natural selection
and that duplication of genes and movements of
large pieces of DNA within the genome have oc-
curred often. As many as 900 ancient conserved
regions of DNA in the E. coli genome correspond-
ing to those in human, nematode, and yeast DNA
are thought to date back perhaps 3.5 10
9
years.
f
However, nobody has explained how life evolved
before there was DNA.
One of the first scientists to devote his career to
biochemical evolution was I. V. Oparin,
g
who
published a book on the origin of life in 1924.
Oparin and J. B. S. Haldane, independently, proposed
that early life was anaerobic and that energy was
provided by fermentation. In 1951 Stanley Miller
built an apparatus that circulated CH
4
, NH
3
, H
2
O,
and H
2
, compounds thought to be present in a
primitive atmosphere, past an electric discharge.
He found glycine, alanine, -alanine, and other amino
acids among the products formed.
h
Schrdinger
pointed out that a flux of energy through a system
will tend to organize the system. The solar energy
passing through the biosphere induces atmospheric
circulation and patterns of weather and ocean
currents.
i,j
Perhaps in the primordeal oceans organic
compounds arose from the action of light and light-
ning discharges. These compounds became catalysts
for other reactions which eventually evolved into a
rudimentary cell-less metabolism. It is a large jump
from this to a cell! Among other problems is the lack
of any explanation for the development of individual
cells or of their genomes. However, because it helps
to correlate much information we will always take an
evolutionary approach in this book and will discuss the
beginnings a little more in later chapters.
BOX 1-C IN THE BEGINNING
a
Maynard-Smith, J. (1982) Nature (London) 296, 599601
b
Lenski, R. E., and Travisano, M. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 68086814
c
Wilson, A. C. (1985) Sci. Am. 253(Oct), 164173
d
Eigen, M., Gardiner, W., Schuster, P., and Winkler-Oswatitsch, R.
(1981) Sci. Am. 244(Apr), 88118
e
Doolittle, R. F. (1992) Protein Sci. 1, 191200
f
Green, P., Lipman, D., Hillier, L., Waterston, R., States, D., and
Claverie, J.-M. (1993) Science 259, 17111716
g
Broda, E. (1980) Trends Biochem. Sci. 5, IVV
h
Miller, S. L. (1953) Science 117, 528529
i
Mason, S. (1993) Trends Biochem. Sci. 18, 230231
j
Welch, G. R. (1996) Trends Biochem. Sci. 21, 452
A. The Simplest Living Things
10 Chapter 1. The Scene of Action
E. coli
St
Cp
Nu
N
M
Ct
G
CW
P
DI
ER
BM
Gl
M
Mb
L
D
TJ
C
SG
Mv
5 m 4 3 2 1 0
V
Gap
Abbreviations:
BM, basement membrane
ER, rough endoplasmic reticulum
(with ribosomes attached; smooth
ER is depicted nearer the nucleus
and on the right side of the cell.)
DI, deep indentation of plasma
membrane
GI, glycogen granules
Gap, space ~10-20 nm thick
between adjacent cells
M, mitochondrion
Mb, microbody
L, lysosome
D, desmosome
TJ, tight junction
Mv, microvilli
C, cillium
SG, secretion granule
V, vacuole
Nu, nucleolus
G, Golgi apparatus
CW, cell wall (of a plant)
Ct, centrioles
P, plasmodesmata
N, nucleus
Cp, chloroplast
St, starch granule
Figure 1-6 The average eukaryotic cell. This composite drawing shows the principal organelles of both animal and plant
cells approximately to the correct scale. (Adapted from a drawing by Michael Metzler.)
producing creatures existing today are the cyano-
bacteria,
34
also known as blue-green algae. Many
cyanobacteria are unicellular, but others such as Oscil-
latoria, a slimy plant that often coats the inside walls
of household aquaria, consist of long filaments about 6
m in diameter (see Fig. 1-11). All cyanobacteria contain
two groups of pigments not found in other prokaryotes:
chlorophyll a and -carotene, pigments that are also
found in the chloroplasts of true algae and in higher
plants. A recently discovered group of bacteria, the
prochlorophytes, are even closer to chloroplasts in
their pigment composition.
35
In addition to pigmented cells, some cyanobacteria
contain paler cells known as heterocysts. They have a
specialized function of fixing molecular nitrogen. The
development of the ability to convert N
2
into organic
nitrogen compounds represents another important
evolutionary step. Because they can both fix nitrogen
and carry out photosynthesis, the blue-green algae
have the simplest nutritional requirements of any
organisms. They need only N
2
, CO
2
, water, light,
and minerals for growth.
Evolution of the photosynthetic cleavage of water
to oxygen was doubtless a major event with far-
reaching consequences. Biologists generally believe
that as oxygen accumulated in the earths atmosphere,
the obligate anaerobes, which are poisoned by oxygen,
became limited to strictly anaerobic environments.
Meanwhile, a new group of bacteria, the aerobes,
appeared with mechanisms for detoxifying oxygen
11 B. Eukaryotic Cells
and for using oxygen to oxidize complex organic
compounds to obtain energy.
B. Eukaryotic Cells
Cells of the eukaryotes contain true nuclei and
are much larger and more complex internally than are
those of prokaryotes. The nucleus of a cell contains
most of its DNA and is separated from the cytoplasm
by membranes. Within the cytoplasm are various
organelles with characteristic structures. These
include mitochondria, lysosomes, peroxisomes,
and centrioles. Eukaryotic cells come in so many
sizes and shapes and with so many specialized features
that it is impossible to say what is typical. Nevertheless,
Fig. 1-6 is an attempt to portray some sort of average
cell, partly plant and partly animal.
As can be seen from Table 1-2, which lists the
diameters and volumes of several roughly spherical
cells, there is a great variation in size. However, a
diameter of 1020 m may be regarded as typical for
both plants and animals. For growth of a large cell such
as the ovum, many adjacent cells assist in synthesis of
foodstuffs which are transferred to the developing egg
cell. Plant cells are often large but usually 90% or more
of the cell is filled with a vacuole or tonoplast,
36
which is drawn unrealistically small in Fig. 1-6. The
metabolically active protoplasm of plant cells often
lies in a thin layer at their peripheries.
Many cells are far from spherical; for example,
human red blood cells are discs 8 x 8 x 1 to 2 m with
a volume of 80 m
3
. Plant fiber cells may be several
millimeters in length. Nerve cells of animals have long
extensions, the axons, which in the human sometimes
attain a length of a meter. Muscle cells fuse to give
very long multinucleate fibers.
1. The Nucleus
In a typical animal cell the nucleus has a diameter
of ~5 m and a volume of 65 m
3
. Except at the time
of cell division, it is densely and almost uniformly
packed with DNA. The amount of DNA present is
larger than that in bacteria as is indicated in Table 1-3.
Yeast contains about three times as much genetic
matter as E. coli and a human being or a mouse about
700 times as much. However, genes are sometimes
duplicated in higher organisms and large amounts of
repetitive DNA of uncertain significance are often
present. Some amphibians have 25 times more DNA
per cell than do humans. The fruit fly Drosophila
contains about 13,600 functioning genes and a human
being perhaps 50,000.
37
Because of its acidic character, DNA is stained by
basic dyes. Long before the days of modern biochem-
istry, the name chromatin was given to the material in
the nucleus that was colored by basic dyes. At the time
of cell division, the chromatin is consolidated into
distinct chromosomes which contain, in addition to
15% DNA, about 10% RNA and 75% protein.
Nearly all of the RNA of the cell is synthesized
(transcribed) in the nucleus, according to the instruc-
tions encoded in the DNA. Some of the RNA then
moves out of the nucleus into the cytoplasm where it
functions in protein synthesis and in some other ways.
Many eukaryotic genes consist of several sequences
that may be separated in the DNA of a chromosome by
intervening sequences of hundreds or thousands of
base pairs. The long RNA transcripts made from these
split genes must be cut and spliced in the nucleus to
form the correct messenger RNA molecules which are
then sent out to the ribosomes in the cytoplasm.
Each cell nucleus contains one or more dense
nucleoli, regions that are rich in RNA and may contain
1020% of the total RNA of cells. Nucleoli are sites
of synthesis and of temporary storage of ribosomal
RNA, which is needed for assembly of ribosomes. The
nuclear envelope is a pair of membranes, usually a
few tens of nanometers apart, that surround the nucleus.
The two membranes of the pair separate off a thin
perinuclear space (Fig. 1-7). The membranes contain
pores ~130 nm in diameter with a complex structure
(see Fig. 27-8).
38,39
There is a central channel ~42 nm in
diameter, which provides a route for controlled passage
of RNA and other large molecules from the nucleus
into the cytoplasm and also from the cytoplasm to the
nucleus. Smaller ~10 nm channels allow passive
diffusion of ions and small molecules.
TABLE 1-2
Approximate Sizes of Some Cells
Diameter Approximate
Cell (m) volume (m
3
)
E. coli 1 1.0
Small thymus cell 6 120
Liver cell 20 4,000
Human ovum (mature) 120 500,000
Hens egg (white excluded) 20,000 4 10
12
Yeast cell 10 500
Onion root (meristematic cell) 17 2,600
Parenchyma cell of a fruit 1,000 1 10
8
12 Chapter 1. The Scene of Action
2. The Plasma Membrane
The thin (8 nm) outer cell mem-
brane or plasma-lemma (Fig. 1-7)
controls the flow of materials into
and out of cells, conducts impulses
in nerve cells and along muscle
fibrils, and participates in chemical
communication with other cells.
Deep infoldings of the outer mem-
brane sometimes run into the cyto-
plasm. An example, is the T system
of tubules which functions in excita-
tion of muscle contraction (Figs. 19-7,
19-21). Surfaces of cells designated
to secrete materials or to absorb
substances from the surrounding
fluid, such as the cells lining kidney
tubules and pancreatic secretory
cells, are often covered with very
fine projections or microvilli which
greatly increase the surface area.
In other cases projections from one
cell interdigitate with those of an
adjacent cell to give more intimate
contact.
3. Vacuoles, Endocytosis, and
Lysosomes
Cells often contain vacuoles or
smaller vesicles that are separated
from the cytosol by a single mem-
brane. Their content is often quite
Mycoplasma genitalium
ac
0.580 1 482
Rickettsia prowazekii
d
1.11 1 834
Haemophilus influenzae
c,eg
1.83 1 1,709
Methanococcus janaschii (an archaeon)
h
1.66 1 1,738
Bacillus subtilis
i
4.16 1
Escherichia coli
j,k
4.64 1 4,288
Myxococcus xanthis
f
9.2 1
Synechocystis sp. (a cyanobacterium)
l
3.57 1 3,169
Saccharomyces cerevisiae (a yeast)
k,m-p,t
13.5 17 6,241
Giardia lamblia (a protozoan)
q
12
Plasmodium falciparum (malaria parasite)
v
25-30 14
Dictyostelium discoideum (a slime mold)
r
34
Caenorhabditis elegans (a nematode)
s,t
97 6 18,424
Sea Urchin 900
Drosophila melanogaster (fruit fly)
u,t
180 4 13,601
Danio rerio (zebrafish)
w
1,700 2.5
Fugu rubripes (pufferfish)
x
400 60,000
S. African lungfish
y
102,000 19
Mus musculus (mouse)
z
3,000 20 80,000
Bos (cow)
aa
~3,000 30 80,000
Homo sapiens (human)
bb,cc
~3,000 23 50,000
150,000
Arabidopsis thaliana (green plant)
dd,ee
115.4 5 25,498
Rice
ff-hh
450 12
Maize
ii
or Wheat
ff
~2,700
Lily
jj
>100,000
TABLE 1-3
Haploid Genome Sizes for Several Organisms
Organism; see footnotes
for sequence information
Millions of
nucleotide
base pairs
(Mb)
Estimated
number
of
genes
Number
of
chromosomes
(haploid)
a
Fraser, C. M., and 28 other authors (1995) Science 270, 397403
b
Goffeau, A. (1995) Science 270, 445446
c
Brosius, J., Robison, K., Gilbert, W., Church, G. M., and Venter, J. C. (1996)
Science 271, 13021304
d
Andersson, S. G. E., Zomorodipour, A., Andersson, J. O., Sicheritz-Pontn,
T., Alsmark, U. C. M., Podowski, R. M., Nslund, A. K., Eriksson, A.-S.,
Winkler, H. H., and Kurland, C. G. (1998) Nature (London) 396, 133140
e
Fleischmann, R. D., and 39 other authors (1995) Science 269, 496512
f
He, Q., Chen, H., Kupsa, A., Cheng, Y., Kaiser, D., and Shimkets, L. J. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 95849587
g
Mrzek, J., and Karlin, S. (1996) Trends Biochem. Sci. 21, 201202
h
Bult, C. J., and 39 other authors; corresponding author Venter, J. C. (1996)
Science 273, 10581073
i
Azevedo, V., Alvarez, E., Zumstein, E., Damiani, G., Sgaramella, V., Ehrlich,
S. D., and Serror, P. (1993) Proc. Natl. Acad. Sci. U.S.A. 90, 60476051
j
Blattner, F. R. and 16 other authors (1997) Science 277, 14531462
k
Winzeler, E. A., and 51 other authors (1999) Science 285, 901906
l
Kaneko, T., and 23 other authors (1996) DNA Res. 3, 109136. See also
http://www.kazusa.or.jp/cyano
m
Dujon, B., and 107 other authors (1994) Nature (London) 369, 371378
n
Taguchi, T., Seko, A., Kitajima, K., Muto, Y., Inoue, S., Khoo, K.-H., Morris,
H. R., Dell, A., and Inoue, Y. (1994) J. Biol. Chem. 269, 87628771
o
Johnston, M., and 34 other authors. (1994) Science 265, 20772082
p
Williams, N. (1995) Science 268, 15601561
q
Sogin, M. L., Gunderson, J. H., Elwood, H. J., Alonso, R. A., and Peattie, D.
A. (1989) Science 243, 7577
r
Loomis, W. F., and Insall, R. H. (1999) Nature (London) 401, 440 441
s
The C. elegans Sequencing Consortium (1998) Science 282, 20122018
(See this article for list of authors.)
t
Rubin, G. M., and 54 other authors. (2000) Science 287, 2204 2215
u
Adams, M. D., and 194 other authors. (2000) Science 287, 2185 2195
v
Su, X.-z, Ferdig, M. T., Huang, Y., Huynh, C. Q., Liu, A., You, J., Wootton, J. C.,
and Wellems, T. E. (1999) Science 286, 13511353
w
Postlethwait, J. H., Johnson, S. L., Midson, C. N., Talbot, W. S., Gates, M.,
Ballinger, E. W., Africa, D., Andrews, R., Carl, T., Eisen, J. S., Horne, S.,
Kimmel, C. B., Hutchinson, M., Johnson, M., and Rodriguez, A. (1994)
Science 264, 699703
x
Fishman, M. C. (1999) Proc. Natl. Acad. Sci. U.S.A. 96, 10554 10556
y
Kornberg, A., and Baker, T. A. (1992) DNA Replication, 2nd ed., pp. 1921,
Freeman, New York
z
Dietrich, W. F., Copeland, N. G., Gilbert, D. J., Miller, J. C., Jenkins, N. A.,
and Lander, E. S. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 1084910853
aa
Anonymous (1994) Nature (London) 368, 167
bb
Schuler, G. D. and 102 other authors (1996) Science 274, 540546
cc
Koonin, S. E. (1998) Science 279, 3637
dd
Olson, M. V. (1993) Proc. Natl. Acad. Sci. U.S.A. 90, 43384344
ee
The Arabidopsis Genome Initiative, (2000) Nature 408, 796815
ff
Stevens, J. E. (1994) Science 266, 11861187
gg
Shimamoto, K. (1995) Science 270, 17721773
hh
Singh, K., Ishii, T., Parco, A., Huang, N., Brar, D. S., and Khush, G. S. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 61636168
ii
Carels, N., Barakat, A., and Gernardi, G. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 1105711060
jj
Alberts, B., Bray, D., Lewis, J., Raff, M., Roberts, K., and Watson, J. D. (1994)
Molecular Biology of the Cell, 3rd ed., Garland, New York
13
Figure 1-7 Electron micrograph of a thin section of a young epidermal cell of a sunflower. The tissue was fixed and stained
with uranyl acetate and lead citrate. Clearly visible are the nucleus (N), mitochondria (M), chloroplasts (C), a Golgi body
dictyosome (G), endoplasmic reticulum, vacuole (V), cell wall, plasmodesmata, and cuticle (upper right, thin dark layer).
Micrograph courtesy of H. T. Horner.
out or excess cell parts including mitochondria.
Lysosomes are vital components of cells,
41
and several
serious human diseases result from a lack of specific
lysosomal enzymes.
4. The Endoplasmic Reticulum and Golgi
Membranes
Although cytoplasm is fluid and in some organisms
can undergo rapid streaming, the electron microscope
acidic.
40
Small vesicles sometimes bud inward from
the plasma membrane in a process called endocytosis.
In this manner the cell may engulf particles (phago-
cytosis) or droplets of the external medium (pinocytosis).
The resulting endocytotic vesicles or endosomes often
fuse with lysosomes, which are small acidified vesicles
containing a battery of enzymes powerful enough to
digest almost anything in the cell. In cells that engulf
bits of food (e.g., ameba) lysosomes provide the diges-
tive enzymes. Lysosomes also take up and digest
denatured or damaged proteins and may digest worn
N
C
G
V
M
B. Eukaryotic Cells
14 Chapter 1. The Scene of Action
has revealed that within the liquid portion, the cytosol,
there is a complex network of membranes known as
the endoplasmic reticulum (ER). The membranes
of the ER form tubes, vesicles, and flattened sacs called
cisternae. The intracisternal spaces appear to connect
with the perinuclear space and to a series of 312
flattened, slightly curved disk-shaped membranes
known as the Golgi apparatus (Figs. 1-7, 20-8).
42,43
This organelle was first reported by Camillo Golgi in
1898.
44
Its existence was long doubted, but it is known
now to play a vital role in metabolism.
The ER, the Golgi membranes, and secretion gran-
ules apparently represent an organized system for
synthesis of secreted protein and formation of new
membranes. Parts of the ER, the rough endoplasmic
reticulum are lined with many ribosomes of 2125 nm
diameter. While resembling those of bacteria, these
eukaryotic ribosomes are about 50% heavier (4 x 10
6
Da).
The smooth endoplasmic reticulum lacks ribosomes
but proteins made in the rough ER may be modified in
the smooth ER, e.g., by addition of carbohydrate chains.
Small membrane vesicles break off from the smooth
ER and pass to the Golgi membranes which lie close to
the smooth ER on the side toward the center of the cell.
Here additional modification reactions occur (Chapter
20). At the outer edges the membranes of the Golgi
apparatus pinch off to form vacuoles which are often
densely packed with enzymes or other proteins.
These secretion granules move to the surface and are
released from the cell. In this process of exocytosis
the membranes surrounding the granules fuse with
the outer cell membrane. The rough ER appears to
contribute membrane material to the smooth ER and
Golgi apparatus, while material from Golgi membranes
can become incorporated into the outer cell membrane
and into lysosomes. Outer mitochondrial membranes
and membranes around vacuoles in plant cells may
also be derived directly from the ER. Outer membrane
materials are probably recycled by endocytosis.
The term microsome, frequently met in the bio-
chemical literature, refers to small particles of 50150
nm diameter which are mostly fragments of the ER
together with some material from the plasma mem-
brane. Microsomes are formed when cells are ground
or homogenized. Upon centrifugation of the disrupted
cells, nuclei and other large fragments sediment first,
then the mitochondria. At very high speeds (e.g., at
100,000 times the force of gravity) the microsomes,
whose masses are 10
8
10
9
Da, settle. With the electron
microscope we see that in the microsomes the mem-
brane fragments have closed to give small sacs to the
outside of which the ribosomes still cling:
5. Mitochondria, Plastids, and Peroxisomes
Mitochondria, complex bodies about the size of
bacteria and bounded on the outside by a double mem-
brane (Figs. 1-6 and 1-7), are present in all eukaryotic
cells that use oxygen for respiration. The numbers
per cell appear to vary from the one for certain tiny
trypanosomes to as many as 3 x 10
5
in some oocytes.
Liver cells often contain more than 1000 mitochondria
apiece.
45
Study of ultrathin serial sections of a single
yeast cell by electron microscopy has shown that under
some growth conditions all of the yeast mitochondria
are interconnected.
46
More recent evidence from new
imaging procedures, e.g. using the green fluorescent
protein (Box 23-A)
46a, 46b
also supports the idea that
mitochondria are interconnected in a reticulum that
can become fragmented under some conditions. The
inner membrane of a mitochondrion is often highly
folded to form the cristae (crests). The outer mem-
brane is porous to small molecules but the passage of
substances into and out of the inner space of the mito-
chondrion, known as the matrix, is tightly controlled by
the inner membrane. Although some of the oxidative
chemical activites of the cells are located in the ER and in
peroxisomes, the major energy-yielding reactions for
aerobic organisms are found in the mitochondria, which
are also the principal site of utilization of oxygen.
Within each mitochondrion is a small circular molecule
of DNA whose genes encode only a few of the many
proteins needed in this organelle. Also present within
mitochondria are ribosomes of a size similar to those
of bacteria and smaller than those lining the rough ER.
Plastids are organelles of plant cells that serve
a variety of purposes.
45
Most important are the
chloroplasts, the chlorophyll-containing sites of photo-
synthesis. Like mitochondria they contain folded
internal membranes (see Fig. 23-19) and several small
molecules of DNA.
Fragile organelles, the peroxisomes or microbodies,
occur in many cells.
4750
In green leaves they may occur
in numbers up to one-third those of mitochondria.
Peroxisomes are often about the size of mitochondria
but have only a single membrane and do not contain
DNA. They often contain an apparently crystalline
core. The single membrane of peroxisomes is porous
to small molecules such as sucrose. This permits these
organelles to be separated from mitochondria by cen-
trifugation in a sucrose gradient where the microbodies
assume a density of about 1.25 g/cm
3
compared to 1.19
for the impervious mitochondria.
Peroxisomes are rich in enzymes that produce and
decompose hydrogen peroxide. They often make a
major contribution to the oxidative metabolism of
cells. In germinating oilseeds glyoxysomes, a type of
peroxisome, contain enzymes that catalyze reactions of
the biosynthetic glyoxylate pathway of metabolism.
51
Organelles that resemble peroxisomes in appearance
Endoplasmic
reticulum
Microsomes
15
but which are functionally more closely related to
mitochondria are the hydrogenosomes of anaerobic
protozoa.
52
As the name suggests, these organelles are
the site of formation of molecular hydrogen, a common
product of anaerobic metabolism.
6. Centrioles, Cilia, Flagella, and Microtubules
Many cells contain centrioles,
53
little cylinders
about 0.15 m in diameter and 0.5 m long, which are
not enclosed by membranes. Each centriole contains
a series of fine microtubules of 25 nm diameter. A
pair of centrioles are present near the nucleus in most
animal cells and play an important role in cell division.
Together with surrounding materials they form the
centrosome. However, centrioles have never been
observed in plant cells.
Related in structure to centrioles are the long
flagella and shorter cilia (the two words are virtually
synonymous) which are commonly present as organ-
elles of locomotion in eukaryotic cells. Stationary cells
of our own bodies also often have cilia. For example,
there are 10
9
cilia/cm
2
in bronchial epithelium.
54
Modified flagella form the receptors of light in our
eyes and of taste in our tongues. Flagella and cilia
have a diameter of about 0.2 m and a characteristic
internal structure. Eleven hollow microtubules of ~24
nm diameter are usually arranged in a 9 + 2 pattern
with nine pairs of fused tubules surrounding a pair of
single tubules (Figs. 1-8 and 19-23). Each microtubule
Figure 1-8 Structure of cilia and flagella of eukaryotes. After P. Satir.
57
resembles a bacterial flagellum in appearance, but
there are distinct and significant chemical differences.
The basal body of the flagellum, the kinetosome
(Fig. 1-8), resembles a centriole in structure, dimensions,
and mode of replication. Recently a small 69 megabase
pair DNA has been found in basal bodies of the protozoan
Chlamydomonas.
55,56
Microtubules similar to those found in flagella are
also present in the cytoplasm. Together with thinner
microfilaments of several kinds they form an internal
cytoskeleton that provides rigidity to cells.
58,59
Microtubules also form the spindle of dividing cells.
In nerve axons (Chapter 30) the microtubules run
parallel to the length of the axons and are part of a
mechanical transport system for cell constituents.
7. Cell Coats, Walls, and Shells
Like bacteria, most cells of higher plants and
animals are surrounded by extracellular materials.
Plants have rigid walls rich in cellulose and other
carbohydrate polymers. Outside surfaces of plant cells
are covered with a cuticle containing layers of a poly-
ester called cutin and of wax (Fig. 1-6). Surfaces of
animal cells are usually lined with carbohydrate mole-
cules which are attached to specific surface proteins to
formglycoproteins. Spaces between cells are filled
with such cementing substances as pectins in plants
and hyaluronic acid in animals. Insoluble proteins
such as collagen and elastin surround connective
tissue cells. Cells that lie on a surface
(epithelial and endothelial cells) are
often lined on one side with a thin,
collagen-containing basement
membrane (Figs. 1-6 and 8-31).
Inorganic deposits such as calcium
phosphate (in bone), calcium carbonate
(eggshells and spicules of sponges), and
silicon dioxide (shells of diatoms) are
laid down, often by cooperative action
of several or many cells.
C. Inheritance, Metabolic
Variation, and Evolution
of Eukaryotes
The striking differences between
eukaryotic and prokaryotic cells have
led to many speculations about the
evolutionary relationship of these two
great classes of living organisms. A
popular theory is that mitochondria,
which are characteristic of most eukary-
otes, arose from aerobic bacteria. After
cyanobacteria had developed and oxygen
B. Eukaryotic Cells
0.5 m
E. coli
This cilium is about 8 m long.
Lengths range from about 2 m
to 2 mm with 1020 m most common.
10
20
30
40 50
0
Basal body (kinetosome)
Cross-section, 2.5
Secondary fiber
Membrane
Microtubules (24 nm diameter)
in 9 + 2 array
Arms on microtubules
at 1722 nm intervals
Typical movement of a long
flagellum of a eukaryote
Cross-section,
2.5
16 Chapter 1. The Scene of Action
In 1908 Archibald Garrod
a,b
proposed that cysti-
nuria (Chapter 8) and several other defects in amino
acid and sugar metabolism were inborn errors of
metabolism, i.e. inherited diseases. Since that time
the number of recognized genetic defects of human
metabolism has increased at an accelerating rate to
~4000.
ce
Hundreds of other genetic problems have
also been identified. For over 800 of these the
defective gene has been mapped to a specific
chromosome.
f
An example is sickle cell anemia
(Box 7-B) in which a defective hemoglobin differs
from the normal protein at one position in one of its
constituent polypeptide chains. Many other defects
involve loss of activity of some important enzyme.
Most genetic diseases are rare, affecting about
one person in 10,000. However, cystic fibrosis
affects one in 2500. There are so many metabolic
diseases that over 0.5% of all persons born may
develop one. Many die at an early age. A much
greater number (>5%) develop such conditions as
diabetes and mental illness which are, in part, of
genetic origin. Since new mutations are always
arising, genetic diseases present a problem of con-
tinuing significance.
At what rate do new mutations appear? From
the haploid DNA content (Table 1-2) we can estimate
that the total coding capacity of the DNA in a human
cell exceeds two million genes (actually two million
pairs of genes in diploid cells). However, only a frac-
tion of the DNA codes for proteins. There are per-
haps 50,000 pairs of structural genes in human DNA.
The easily detectable rate of mutation in bacteria is
about 10
6
per gene, or 10
9
per base per replication.
g
As a result of sophisticated proofreading and
repair systems, it may be as low as 10
10
per base in
humans.
h
Thus, in the replication of the 3 x 10
9
base
pairs in diploid human chromosomes we might
anticipate about one mistake per cell division. Only
about 1/50 of these would be in structural genes and
potentially harmful. Thus, if there are 10
16
division
cycles in a normal life span
h
each parent may pass on
to future generations about 2 mutations in protein
sequences. The ~10
14
body cells (somatic cells) also
undergo mutations which may lead to cancer and to
other problems of aging. Most mutations may be
harmless or nearly so and a few may be beneficial.
However, many are damaging and some are lethal.
If a mutation is lethal, a homozygote will not survive
and will be lost in an early (and usually undetected)
spontaneous abortion. Healthy individuals carry as
many as ten lethal recessive mutations as well as at
least 35 autosomal recessive mutations of a seriously
harmful type. Harmful dominant mutations are also
frequent in the population. These include an elevated
lipoprotein content of the blood and an elevated cho-
lesterol level which are linked to early heart disease.
Biochemical disorders are also important because
of the light they shed on metabolic processes. No
other species is observed as carefully as Homo sapiens.
As a consequence frequent reference will be made to
genetic diseases throughout the book. A goal is to
find ways to prevent or ameliorate the effects of these
disorders. For example, in the treatment of phenyl-
ketonuria (Chapter 25) or of galactosemia (Chapter
20), a change in the diet can prevent irreversible
damage to the brain, the organ most frequently
affected by many of these diseases. Injection of a
missing enzyme is giving life to victims of Gauchers
disease (Chapter 20). In many other cases no satis-
factory therapy is presently available, but the possi-
bilities of finding some way to supply missing
enzymes or to carry out genetic surgery are among
the most exciting developments of contemporary
medical biochemistry (Chapter 26).
Photomicrograph of human male metaphase chromosomes. Photo
Researchers
a
Garrod, A. E. (1909) Inborn Errors of Metabolism, Oxford, London
b
Bearn, A. G. (1993) Archibald Garrod and the Individuality of Man,
Oxford, New York
c
Scriver, C. R., Beaudet, A. L., Sly, W. S., and Valle, D., eds. (1995)
The Metabolic and Molecular Bases of Inherited Disease, 7th ed.,
McGraw-Hill, New York
d
Davies, K. E. (1992) Molecular Basis of Inherited Disease, Oxford,
New York
BOX 1-D INHERITED METABOLIC DISEASES
e
McKusick, V. A. (1994) Mendelian Inheritance in Man, 11th ed.,
Johns Hopkins Univ. Press, Baltimore, Maryland
f
McKusick, V. A., Amberger, J., and Steinberg, J. (1994) J. NIH
Res. 6, 115134
g
Watson, J. D. (1976) Molecular Biology of the Gene, 3rd ed.,
Benjamin, Menlo Park, California (p. 254)
h
Koshland, D. E., Jr. (1994) Science 266, 1925
17
had become abundant, a symbiotic relationship could
have arisen in which small aerobic bacteria lived
within cells of larger bacteria that had previously been
obligate anaerobes. Sequence similarities of proteins
suggest that these symbionts may have been related to
present-day methanogens
60
and thermophilic sulfur
bacteria.
61
The aerobes presumably used up any oxy-
gen present, protecting the surrounding anaerobic
organisms from its toxicity. The relationship became
permanent and led eventually to the mitochondria-
containing eukaryotic cell.
6265
Further symbiosis with
cyanobacteria or prochlorophytes could have led to the
chloroplasts of the eukaryotic plants.
A fact that supports such ideas is the existence
among present-day organisms of many endosymbiotic
relationships. For example, the green paramecium
(Paramecium bursaria) contains, within its cytoplasm,
an alga (Chlorella), a common green plant that is quite
capable of living on its own. Perhaps by accident it
took up residence within the paramecium.
62
Some
dinoflagellates (Fig. 1-9) contain endosymbiotic cyano-
bacteria
66
and recently a ciliate that contains endo-
symbiotic purple photosynthetic bacteria has been
discovered.
67
These bacteria do not produce O
2
but
utilize products of the host ciliates metabolism such
as acetate, lactate, and H
2
as electron donors for photo-
syntheses. They also utilize O
2
for respiration and
may protect their hosts from the toxicity of O
2
, just as
may have happened in the distant past. According to
this theory the symbionts would eventually have lost
their photosynthetic ability and have become mito-
chondria. The relationship of mitochondria to bacteria
is also supported by many biochemical similarities.
Fossils of bacteria and blue-green algae have been
obtained from rocks whose age, as determined by
geochemical dating, is more than 3 10
9
years.
68,69
However the first eukaryotic cells may have appeared
about 1 10
9
years ago
70
and started to evolve into the
more than one million species that now exist.
1,71,72
1. A Changing Genome
How is it possible for the genome of an organism
to increase in size as it evolved from a lower form to a
higher one? Simple mutations that cause alterations in
protein sequences could lead to changes in form and
behavior of the organisms but could not, by themselves,
account for the increase in genetic material that accom-
panied evolution. As a result of new techniques of
genetic mapping and determining the sequence of
nucleotides in DNA we are rapidly acquiring a detailed
knowledge of the organization of the genome. It has
been found that genes are often present as duplicate
but not entirely identical copies. This suggests that
there are mechanisms by which cells can acquire extra
copies of one or more genes. Indeed it seems probable
that at some time in the past the entire genome of
bacteria was doubled and that it was later doubled
again.
73
Evidence for this is that the masses of bacterial
chromosomes group around values of 0.5, 1.4, and
2.7 x 10
9
Da. Genes can also be duplicated during the
process of genetic recombination, which is discussed
in Chapter 27. In addition, the size of the genome may
have increased by incorporation of genetic material
from extrachromosomal plasmids.
A possible advantage to a cell possessing an extra
copy of a gene is that the cell would survive even when
mutations rendered unusable the protein encoded by
one of the copies. As long as one of the genes remained
good, the organism could grow and reproduce.
The extra, mutated gene could be carried for many
generations. As long as it produced only harmless,
nonfunctioning proteins there might be little selection
pressure to eliminate it and it might undergo repeated
mutations. After many mutations and many genera-
tions later, the protein for which it coded could prove
useful to the cell in some new way.
An example of evolution via gene duplication is
provided by the oxygen-carrying proteins of blood.
It appears that about a billion years ago, the gene for
an ancestral globin, the protein of hemoglobin, was
doubled. One gene evolved into that of present-day
globins and the other into the gene of the muscle
protein myoglobin. Still later, the globin gene again
doubled leading to the present-day and chains of
hemoglobin (Chapter 7). These are two distinctly
different but related protein subunits whose genes are
not even on the same chromosome. To complicate the
picture further, most human beings have two or more
copies of their chain gene
74
as well as genes for fetal
and embryonic forms of hemoglobin. However, some
populations have lost one or more chain genes. Thus,
the genome changes in many details, even today.
2. Genetic Recombination, Sex, and
Chromosomes
Bacteria usually reproduce by simple fission. The
single DNA molecule of the chromosome is duplicated
and the bacterium divides, each daughter cell receiving
an identical chromosome. However, genetic recombi-
nation, which is accomplished in several ways by
bacteria (Chapter 27), provides a deliberate process
for mixing of genes. This process has been most fully
developed in eukaryotic organisms that undergo sexual
reproduction. The growth of a multicelled individual
begins with the fusion of two haploid gametes, an egg
and a spermatozoon. Each gamete carries a complete
set of genetic instructions, and after the nuclei fuse the
fertilized egg or zygote is diploid. Each diploid cell
contains two complete sets of genetic blueprints of
quite different origin. Even if a gene from one parent
C. Inheritance, Metabolic Variation, and Evolution of Eukaryotes
18 Chapter 1. The Scene of Action
is defective, the chances are that the gene from the other
parent will be good. Sexual reproduction and the
associated genetic recombination also provide a means
for mixing of genes.
When eukaryotic cells prepare to divide in the
process called mitosis (Fig. 26-11), the DNA molecules
of the nucleus, which become spread out through a large
volume, coil and fold. Together with proteins and other
molecules they form the compact bodies known as
chromosomes. Some organisms, such as Ascaris (a round-
worm), have only two chromosomes, a homologous
pair, one inheritied from the father and one from the
mother. Both chromosomes divide in every mitotic cell
division so that every cell of the organism has the homo-
logous pair. Higher organisms usually have a larger
number of chromosomes. Thus, humans have 23 homo-
logous pairs. The mouse has 20, the toad 11, onions 8,
mosquitos 3, and Drosophila 4. Human chromosomes
vary in size but are usually 46 m long and ~1 m in
diameter.
By the successive divisions of mitosis, a single
fertilized eukaryotic egg cell can grow to an adult.
Less than 50 successive mitotic divisions will produce
the ~10
14
cells of a human. However, formation of
gametes, which are haploid, requires the special pro-
cess of meiosis (Fig. 26-12), by which the number of
chromosomes is divided in half. During meiosis one
chromosome of each of the homologous pairs of the
diploid cell is passed to each of the gametes that are
formed. In an organism such as Ascaris, which con-
tains only a single pair of chromosomes, a gamete
receives either the chromosome of maternal origin or
that of paternal origin but not both. In organisms that
have several pairs of chromosomes, one chromosome of
each pair is passed to the gamete in a random fashion
during meiosis. Most gametes receive some chromo-
somes of maternal and some of paternal origin. An
important feature of meiosis is the genetic recombina-
tion that occurs during crossing-over. In this process,
the strands of DNA are cut and genetic material is
exchanged between the chromosomes of maternal and
paternal origin. Thus, crossing-over breaks the linkage
between genes and provides for greater variability in
the offspring than would otherwise be possible. Each
of us receives half of our genes from our mother and
half from our father, but some of these genes have been
inherited from each grandparent on both sides of the
family, some from each great-grandparent, etc.
Many genes are passed down through many
generations without substantial change, but others
are evidently designed to be scrambled readily within
somatic cells. Cell surface proteins
75
and antibody
molecules are among the proteins whose genes undergo
alteration during growth and differentiation of the
tissues of the body (Chapter 32).
3. Haploid and Diploid Phases
In human beings and other higher animals, meiosis
leads directly to formation of the gametes, the egg and
sperm cells. These fuse to form a diploid nucleus and
the adult develops by repeated mitosis of the diploid
cells. While meiosis also occurs in the life cycle of all
eukaryotic creatures, it is not always at a point corre-
sponding to that in the human life cycle. Thus, the
cells of many protozoa and of fungi are ordinarily
haploid. When two haploid nuclei fuse to form a
diploid cell, meiosis quickly occurs to produce haploid
individuals again. Among lower plants and animals
there is often an alternation of haploid and diploid
phases of the life cycle. For example, gametes of ferns
fall to the ground and germinate to form a low-growing
green mosslike haploid or gametophyte form. The
latter produces motile haploid gametes which fuse to a
diploid zygote that grows into the larger and more obvious
sporophyte form of the fern.
It is presumably the ability to survive as a hetero-
zygote, even with one or more highly deleterious
mutations, that has led to the dominance of the diploid
phase in higher plants and animals.
76
However, to
the biochemical geneticist organisms with a haploid
phase offer experimental advantages because recessive
mutants can be detected readily.
D. Survey of the Protists
Unicellular eukaryotes have traditionally been
grouped together with multicellular organisms in
which all cells have similar functions, with little or no
differentiation into tissues, as the kingdom Protista.
77,78
The fungi may also be included or may be regarded as
a separate kingdom.
79
With present-day emphasis on
DNA sequence comparisons the traditional classifica-
tion is changing, however.
26
1. Protozoa
Among the best known of the animal-like protista
is the ameba (subphylum Sarcodina or Rhizopoda).
The most striking feature of the ameba (Fig. 1-9) is
its method of locomotion, which involves the trans-
formation of cytoplasm from a liquid state to a semi-
solid gel. As the ameba moves, the cytoplasm at the
rear liquifies and flows to the front and into the
extending pseudopodia where it solidifies along the
edges. The ameba poses several important biochemical
questions: What chemistry underlies the reversible
change from liquid to solid cytoplasm? How can the
cell membranes break and reform so quickly when an
ameba engulfs food particles?
80
Relatives of the ameba include the Radiolaria,
19
marine organisms of remarkable symmetry with com-
plex internal skeletons containing the carbohydrate
polymer chitin together with silica (SiO
2
) or strontium
sulfate. The Foraminifera deposit external shells of
calcium carbonate or silicon dioxide. Over 20,000
species are known and now as in the distant past their
minute shells fall to the bottom of the ocean and form
limestone deposits.
Tiny ameboid parasites of the subphylum Sporozoa
attack members of all other animal phyla. Several
genera of Coccidia parasitize rabbits and poultry
causing enormous damage. Humans are often the
victims of species of the genus Plasmodium (Fig. 1-9)
which invade red blood cells and other tissues to cause
Figure 1-9 A few well-known protists.
malaria, one of our most serious ailments on a world-
wide basis.
8184
Throughout history malaria has prob-
ably killed more persons than any other disease.
Toxoplasma gondii is another parasite which, in its
haploid phase, is found throughout the world in wild
animals and in humans. Although its presence usually
elicits no symptoms, it sometimes causes blindness
and mental retardation in children and can be fatal to
persons with AIDS. Its sexual cycle occurs exclusively
in cats.
85,86
Another subphylum of protozoa, the Mastigophora,
are propelled by a small number of flagella and are
intermediate between animals and the algae. One of
these is Euglena viridis, a small freshwater organism
with a long flagellum in front, a
flexible tapered body, green chloro-
plasts, and a light-sensitive eye-
spot which it apparently uses to
keep itself in the sunshine (Fig. 1-9).
Euglena is also able to live as a
typical animal if there is no light.
Treatment with streptomycin (Box
20-B) causes Euglena to lose its
chloroplasts and to become an animal
permanently. The dinoflagellates
(Fig. 1-9), some colorless and some
green, occur in great numbers
among the plankton of the sea.
Giardia lamblia is a troublesome
intestinal parasite.
The hemoflagellates are respon-
sible for some of our most terrible
diseases. Trypanosomes (genus
Trypanosoma) invade the cells of the
nervous system causing African
sleeping sickness. Mutating their
surface proteins frequently by gene-
scrambling mechanisms, these and
other parasites are able to evade the
immune response of the host.
87,88
For the same reason it is difficult
to prepare vaccines against them.
Other flagellates live in a symbiotic
relationship within the alimentary
canals of termites (Fig. 1-9) and
roaches. Termites depend upon
bacteria that live within the cells of
these symbiotic protozoans to pro-
vide the essential enzymes needed
to digest the cellulose in wood.
Members of the subphylum
Ciliophora, structurally the most
complex of the protozoa, are cov-
ered with a large number of cilia
which beat together in an organized
pattern.
89
The following question
immediately comes to mind: How
D. Survey of the Protists
50 m
Trichonympha lives in
the gut of termites
Wood particles
digested with help
of symbiotic bacteria
E. colifood
for protozoa
Euglena viridis.
Is it a plant
or an animal?
Chloroplasts
A dinoflagellate
A second
flagellum
lies in the
groove
Trypanosoma
gambiense
(causes African
sleeping sickness)
A red blood cell
Malaria-causing
Plasmodium inside
Gelled cytosol
turns fluid here
Tetrahymena
Amoeba proteus
Cytosol gels
in this region
Entamoeba histolytica
(causes amebic dysentery)
Flow of cytosol
20 Chapter 1. The Scene of Action
are the cilia able to communicate with each other to
provide this organized pattern? Two ciliates that are
often studied by biochemists are Tetrahymena (Fig. 1-9),
one of the simplest, and Paramecium, one of the more
complex.
The Myxomycetes or slime molds are more
closely related to protozoa than to fungi.
90
Members
of the family Acrasieae, the best studied member of
which is Dictyostelium discoideum, start life as small
amebas. After a time, when the food supply runs
low, some of the amebas begin to secrete pulses of a
chemical attractant cyclic AMP. Neighboring amebas
respond to the pulses of cyclic AMP by emitting their
own pulses about 15 s later, then moving toward the
original source.
91,92
The ultimate effect is to cause the
amebas to stream to centers where they aggregate
and form fungus-like fruiting bodies. Asexual spores
are formed and the life cycle begins again. Other
Myxomycetes grow as a multinucleate (diploid)
plasmodium containing millions of nuclei but no
individual cell membranes. Physarum polycephalum, a
species whose plasmodium may spread to a diameter
of 30 cm, has become popular with biochemists.
The 800,000 nuclei per square millimeter all divide
synchronously.
2. Fungi
Lacking photosynthetic ability, living most often in
soil but sometimes in water, the fungi are represented by
almost half as many species (~10
5
) as are the vascular
plants.
93
The distinguishing characteristics of fungi
are the lack of chlorophyll and growth as a series of
many branched tubules (usually 68 m diameter), the
hyphae, which constitute the mycelium. The hyphae
are not made up of separate cells but contain a mass of
protoplasm with many nuclei. Only occasional septa
divide the tubules. Most fungi are saprophytic, living
on decaying plants or animal tissues. However, others
are parasites that produce serious and difficult-to-treat
infections in humans. An important medical problem
is the lack of adequate antibiotics for treating fungal
infections (mycoses).
9496
On the other hand, fungi
produce important antibiotics such as penicillin. Still
others form some of the most powerful toxins known!
The lower fungi or Phycomycetes include simple
aquatic molds and mildew organisms. Higher fungi
are classified as Ascomycetes or Basidiomycetes
according to the manner in which the sexual spores are
born. In the Ascomycetes these spores are produced in
a small sac called an ascus (Fig. 1-10). Each ascus
contains four or eight spores in a row, a set of four
representing the results of a single pair of meiotic
divisions. A subsequent mitotic division will give eight
spores. This is one of the features that has made Neu-
rospora crassa (Fig. 1-10) a favorite subject for genetic
studies.
97
The ascospores can be dissected out in order
from the ascus and cultivated separately to observe the
results of crossing-over during meiosis.
Neurospora also reproduces via haploid spores
called conidia. The haploid mycelia exist as two mating
types and conidia or mycelia from one type can fertilize
cells in a special body (the protoperithecium) of the other
type to form zygotes. The latter immediately undergo
meiosis and mitosis to form the eight ascospores.
Among other Ascomycetes are the highly prized edible
truffles and morels. However, most mushrooms and
puffballs are fruiting bodies of Basidiomycetes. Other
Basidiomycetes include the rusts, which cause enor-
mous damage to wheat and other grain crops.
Yeasts are fungi adapted to life in an environment
of high sugar content and which usually remain uni-
cellular and reproduce by budding (Fig. 1-10). Occa-
sionally the haploid cells fuse in pairs to form diploid
cells and sexual spores. Some yeasts are related to the
Ascomycetes, others to Basidiomycetes. Saccharomyces
cerevisiae, the organism of both bakers and brewers
yeast, is an Ascomycete. It can grow indefinitely in
either the haploid or diploid phase. The genetics
and biochemistry of this yeast have been studied
extensively.
98102
The genome is relatively small with
13.5 x 10
6
base pairs in 17 chromosomes. The sequence
of the 315,000 base pairs of chromosome III was deter-
mined in 1992
101,102
and the sequence of the entire
genome is now known.
103
Fungi often grow in symbiotic association with
other organisms. Of special importance are the
mycorrhizae (fungus roots) formed by colonization
of fine roots by beneficial soil fungi. Almost all plants
of economic importance form mycorrihizae.
104
3. Algae
Algae are chlorophyll-containing eukaryotic organ-
isms which may be either unicellular or colonial.
105
The colonial forms are usually organized as long fila-
ments, either straight or branched, but in some cases
as blades resembling leaves. However, there is little
differentiation among cells. The gold-brown, brown,
and red algae contain special pigments in addition to
the chlorophylls.
The euglenids (Euglenophyta) and dinoflagellates
(Pyrrophyta), discussed in the protozoa section, can
equally well be regarded as algae. The bright green
Chlorophyta, unicellular or filamentous algae, are
definitely plants, however. Of biochemical interest is
Chlamydomonas, a rather animal-like creature with two
flagella and a carotenoid-containing eyespot or stigma
(Fig. 1-11). Chlamydomonas contains a single chloroplast.
The pyrenoid, a center for the synthesis of starch,
lies, along with the eyespot, within the chloroplast. The
organism is haploid with plus and minus strains
21
Figure 1-10 Two frequently studied fungi. Top (including ascus): the yeast
Saccharomyces cerevisiae. Below: Neurospora crassa showing various stages.
After J. Webster.
93
and motile gametes. Zygotes immediately undergo
meiosis to form haploid spores. With a well-established
genetic map, Chlamydomonas is another important organ-
ism for studies of biochemical genetics.
106
The filamentous Ulothrix shows its relationship
to the animals through formation of asexual spores
with four flagella and biflagellate gametes. Only the
zygote is diploid. On the other hand, the incomparably
beautiful Spirogyra (Fig. 1-11) has no motile cells. The
ameboid male gamete flows through a tube formed
between the two mating cells, a behavior suggesting a
relationship to higher green plants.
Some unicellular algae grow to a remarkable size.
One of these is Acetabularia (Fig. 1-11), which lives in
the warm waters of the Mediterranean and other
tropical seas. The cell contains a single nucleus which
lies in the base or rhizoid portion. In the mature alga,
whose life cycle in the laboratory is 6 months, a cap of
characteristic form develops. When cap development
is complete, the nucleus divides into about 10
4
second-
ary nuclei which migrate up the stalk and out into the
rays of the cap where they form cysts. After the cap
decays and the cysts are released, meiosis occurs and
the flagellated gametes fuse in pairs to form zygotes
which again grow into diploid algae.
Because of its large size and the
location of the nucleus in the base,
the cells can be cut and grafted.
Nuclei can be removed or trans-
planted and growth and develop-
ment can be studied in the presence
or absence of a nucleus.
107110
The
green algae Volvox live in wheel-like
colonies of up to several thousand
cells and are useful for biochemical
studies of differentiation.
111
Look through the microscope
at almost any sample of algae from
a pond or aquarium and you will
see little boatlike diatoms slowly
gliding through the water. The most
prominent members of the division
Chrysophyta, diatoms are character-
ized by their external shells of
silicon dioxide. Large and ancient
deposits of diatomaceous earth
contain these durable silica skeletons
which are finely marked, often with
beautiful patterns (Fig. 1-11). The
slow motion of diatoms is accom-
plished by streaming of protoplasm
through a groove on the surface of
the cell. Diatoms are an important
part of marine plankton, and it is
estimated that three-fourths of the
organic material of the world is
produced by diatoms and dino-
flagellates. Like the brown algae,
Chrysophyta contain the pigment
fucoxanthin.
Other groups of algae are the
brown and red marine algae or
seaweed. The former (Phaeophyta)
include the giant kelps from which
the polysaccharide algin is obtained.
The Rhodophyta are delicately
branched plants containing the red
pigment phycoerythrin. The poly-
saccharides, agar and carrageenin,
D. Survey of the Protists
Neurospora crassa
Macroconidia
50 m
Microconidia
Protoperithecium
Nucleus passing
through pore
in septum
10
Ascus containing
8 ascospores
Saccharomyces cerevisiae
Ascus
22 Chapter 1. The Scene of Action
50 100 m 0
Chlamydomonas
A diatom
Chlorella, a common symbiont
in Hydra, sponges, and protozoa.
Chlorella also lives free and has
often been used in studies of
photosynthesis.
Scenedesmus
Cosmarium,
a bright green
desmid
Oscillatoria, a blue-green
alga, a prokaryotic
organism
Spirogyra
Conjugation
tube
Spore
Acetabularia
(actual size)
E. coli
a popular additive to chocolate drinks and other foods,
come from red algae.
Symbiotic associations of fungi with either true
algae or with cyanobacteria are known as lichens.
Over 15,000 varieties of lichens grow on rocks and in
other dry and often cold places. While the algae
appear to benefit little from the association, the fungi
penetrate the algae cells and derive nutrients from
them.
112
Although either of the two partners in a lichen
can be cultured separately, the combination of the two
is capable of producing special pigments and phenolic
substances known as depsides which are not formed
by either partner alone.
Figure 1-11 A few species of algae.
23
Figure 1-12 Some lower forms of Metazoa. (A) Mesozoa (25 cells). After C. P. Hickman.
116
(B) A small asconoid sponge.
After C. A. Villee, W. F. Walker, Jr., and R. D. Barnes.
117
(C) Ameboid cells of a sponge forming spicules. After Hickman.
E. The Variety of Animal Forms
In this section, we will consider only a few bio-
chemical and other aspects of multicellular animals or
Metazoa. The sudden appearance of a large number
of Metazoans about 0.5 x 10
9
years ago
113,114
may have
been an outcome of the appearance of split genes (see
Section B, 1). As a result of gene duplication the coding
pieces of split genes, the exons, could be moved to
new locations in a chromosome where they could have
become fused with other pieces of DNA to form entirely
new genes.
115
1. Major Groups of Multicellular Animals
The simplest metazoa are tiny symbiotic worms
of the phylum (or subkingdom) Mesozoa, which live
in the kidneys of deep sea-dwelling cephalopods
(octopi and squid). Each worm is made up of only 25
cells in a single layer enclosing one or a small number
of elongated axial cells (Fig. 1-12). Mesozoa have been
regarded as parasitic, but they appear to facilitate
excretion of NH
3
by the host through acidification of
the urine.
118,119
Porifera or sponges are the most primitive of
multicelled animals.
120
They lack distinct tissues but
contain several specialized types of cells. The body is
formed by stationary cells that pump water through
the pores to bring food to the sponge. Within the body
amebocytes work in groups to form the spicules of
calcium carbonate, silicon dioxide, or the protein
spongin (Fig. 1-12). Sponges appear to lack a nervous
system.
Individuals of the next most complex major phy-
lum, Cnidaria (formerly Coelenterata), are radially
symmetric with two distinct cell layers, the endoderm
and ectoderm. Many species exist both as a polyp or
hydra form (Fig. 1-13) and as a medusa or jellyfish.
The jellyfish apparently has no brain but the ways in
which its neurons interconnect in a primitive radial net
are of interest. The Cnidaria have a very simple body
form with remarkable regenerative powers. The fresh-
water hydra, a creature about 1 cm long (Fig. 1-13),
contains a total of ~10
5
cells. A complete hydra can be
regenerated from a small piece of tissue if the latter
contains some of both the inner and the outer cell
layers.
121,122
The body of flatworms (phylum Platyhelminthes)
consists of two external cell layers (endoderm and
ectoderm) with a third layer between. A distinct excre-
tory system is present. In addition to a nerve net
resembling that of the Cnidaria, there are a cerebral
ganglion and distinct eyes. One large group of flat-
worms, the planarians (typically about 15 mm in
length, Fig. 1-14), inhabit freshwater streams. They are
said to be the simplest creatures in which behavior can
be studied.
E. The Variety of Animal Forms
Osculum
Spicule
Choanocyte
Spongocoel
Porocyte
Mesenchyme
Amebocyte
Incurrent pore
Pinacocyte
Polar
cells Calotte
Axial cell
nucleus
Axoblasts
Dicyema, a
member of the
phylum Mesozoa
Thickener
Founder
A B C
24 Chapter 1. The Scene of Action
Interstitial cell
Stinging cell
Gland cell
Sperm
Muscle cell
inner layer
Egg
Muscle cell
outer layer
Muscle cell
outer layer
Muscle cell
inner layer
Interstitial cell
Gland cell
Manubrium
Gonad
Radial canal
Medusa
Many parasitic flatworms (tapeworms and flukes)
attack higher organisms.
124
Among them are the
Schistosoma, tiny worms that are transmitted to
humans through snails and which attack the blood
vessels. The resulting schistosomiasis is one of the
most widespread debilitating diseases on earth today,
affecting 200 million people or more.
125,126
The roundworms (Nematoda)
127129
have, in
addition to the enteron (alimentary tract), a separate
body cavity. Free-living nematodes abound in water
and soil but many species are parasitic. They do enor-
mous damage to plants and to some animal species.
Trichina, hookworms
129a
, and filaria worms attack
humans. However, in the laboratory the 1-mm-long,
810-cell nematode, Caenorhabditis elegans (Fig. 1-14) has
become an important animal. In 1963 Sydney Brenner
launched what has become a worldwide effort to make
this tiny worm the equivalent in the animal kingdom of
Figure 1-13 (A) Hydra. After Loomis.
123
(B) The medusa stage of Obelia, a
hydroid coelenterate.
123a
A
B
E. coli in the bacterial world.
129
The
10
8
nucleotides in the worms six
chromosomes contain ~13,600
genes. C. elegans has become an
important animal in which to study
differentiation. Already the exact
lineage of every cell has been traced,
as has every connection among the
302 neurons in the animals nervous
system. The related rotifers,
130
with
whirling wheels of cilia on their
heads (Fig. 1-14) and transparent
bodies, are a delight to the micro-
scopist. Like nematodes, they are
cell constant organisms. The total
number of cells in the body is con-
stant as is that in almost every part
of every organ. Part of the develop-
mental plan of such organisms is a
programmed cell death (Chapter 32).
The Annelida (segmented
worms)
131
are believed to be evolu-
tionary antecedents of the arthropods.
Present-day members include earth-
worms, leeches, and ~10
5
species of
marine polychaetes. Annelids have
a true body cavity separate from the
alimentary canal and lined by a
peritoneum. They have a well-
developed circulatory system and
their blood usually contains a type
of hemoglobin.
About 10
6
species of arthropods
(80% of all known animals) have
been described. Most are very
small.
72
These creatures, which
have a segmented exoskeleton of
chitin and other materials, include
the horseshoe crabs, the Arachnida
(scorpions, spiders, and mites), the Crustacea, Myri-
opoda (centipedes and millipedes), and the Insecta.
Important biochemical problems are associated with
the development and use of insecticides and with our
understanding of the metamorphosis that occurs
during the growth of arthropods.
132
The fruit fly
Drosophila melanogaster has provided much of our basic
knowledge of genetics and continues to be the major
species in which development is studied.
133134a
Among the molluscs (phylum Mollusca) the
squids and octopuses have generated the most interest
among biochemists. The neurons of squid contain giant
axons, the study of which has led to much of our
knowledge of nerve conduction. Octopuses show
signs of intelligence not observed in other invertebrates
whose nervous reactions seem to be entirely prepro-
grammed. The brains of some snails contain only 10
4
neurons, some of which are unusually large. The
25
Warning: Not everything in this book is true.
Despite all efforts to get it right, there are unin-
tentional errors and misinterpretations of experi-
mental results. Indeed, the history of biochemistry
is replete with accounts of experimental findings
that were interpreted incorrectly. Yet, the ideas
expressed often stimulated others to develop a
more correct picture later. The same is true today.
Students should be critical, should look at experi-
mental details, and consult original literature as
much as possible.
Progress in science depends both upon careful
observations and measurements and upon imagi-
native interpretations of unexpected findings.
Speculative ideas, a number of which are men-
tioned in this book, provide an important stimulus
in science. They should neither be ignored nor
accepted as facts. I have tried to write in such
a way that established facts will not often be
confused with speculation.
BOX 1-E ERRORS, MISCONCEPTIONS,
AND SPECULATION
Echinodermata or spiny-skinned animals (starfish,
sea urchins, and sea cucumbers) are regarded as a
highly advanced phylum. Their embryological devel-
opment has been studied intensively.
The phylum Chordata, to which we ourselves
belong, includes not only the vertebrates but also more
primitive marine animals that have a spinal cord.
Among these primitive species, which may be related
to early ancestral forms, are the tunicates or sea squirts.
They have a very high concentration of vanadium in
their blood.
2. Cell Types and Tissues
Isolated animal cells in tissue culture, no matter
how highly differentiated, tend to revert quickly to
one of three basic types known as epitheliocytes,
mechanocytes, and amebocytes. Epitheliocytes are
closely adherent cells derived from epithelial tissues
and thought to be related in their origins to the two
surface layers of the embryonic blastula. Mechano-
cytes, often called fibroblasts or fibrocytes, are
derived from muscle, supporting, or connective tissue.
Like the amebocytes, they arise from embryonic mes-
enchymal tissue cells that have migrated inward from
the lower side of the blastula (Chapter 32). Neurons,
neuroglia, and lymphocytes are additional distinct
cell types.
Figure 1-14 (A) A planarian, length 15 mm. After
Hickman.
116
Diagram of digestive and nervous sytems;
cutaway section shows ventral mouth. Small drawing
shows pharynx extended through ventral mouth. (B) The
nematode Caenorhabditis elegans. Ascaris is very similar in
appearance. From Buchsbaum.
77
(C) A rotifer, Philodina
(~10
3
cells). After C. A. Villee et al.
117
A
B
C
E. The Variety of Animal Forms
Flame cell
Digestive gland
Mouth
Pharynx (mastax)
Jaws
Stomach
Ovary
Protonephridial
tubule
Intestine
Bladder
Pedal glands
Foot
Anus
Eye
Ciliated
crown
Lateral nerve cord
Intestine
(anterior trunk)
Diverticulum
Mouth
Pharyngeal
opening
Intestine
(right posterior trunk)
Cerebral ganglia
Pharynx
Transverse nerve
Pharyngeal
chamber
Mouth
Pharynx
26 Chapter 1. The Scene of Action
Tissues. Cells aggregate to form four major kinds
of tissue. Epithelial tissues line the primary surfaces of
the body: the skin, the digestive tract, urogenital tract,
and glands. External skin is composed of flat platelike
squamous epithelial cells whereas internal surfaces are
often formed by colummar epithelial cells. Glands
(sweat, oil, mammary, and internal secretory) as well
as the sensory organs of the tongue, nose, and ear are
all composed of epithelial cells. Epithelial cells are
among the most highly polarized of cells. One side of
each cell faces the outside, either air or water, while
the other side is often directly against a basement
membrane.
Supporting and connective tissues include the fatty
adipose tissue as well as cartilage and bone. Both
of the latter contain large amounts of intercellular
material or ground substance consisting largely of
complex polymers. Embryonic fibroblasts differentiate
into white fibers, which produce collagen, and yellow
fibers, which form elastin. The fibrils of both of these
proteins are assembled in the intercellular space
where they are embedded in the ground substance.
Osteoblasts form bone by deposition of calcium
phosphate in 37 m thick layers within a ground
substance that contains special proteins.
A third tissue is muscle, which is classified into
three types: striated (voluntary skeletal muscle),
cardiac (involuntary striated muscle), and smooth
(involuntary) muscle. There are two major groups of
cells in nervous tissue, the fourth tissue type. Neurons
are the actual conducting cells whose cell membranes
carry nerve impulses. Several kinds of glial cells lie
between and around the neurons.
Blood cells. Blood and the linings of blood vessels
may be regarded as a fifth tissue type.
135,135a
The
human body contains 5 x 10
9
erythrocytes or red
blood cells per ml, a total of 2.5 x 10
13
cells in the five
liters of blood present in the body. Erythrocytes are
rapidly synthesized in the bone marrow. The nucleus
is destroyed, leaving a cell almost completely filled
with hemoglobin. With an average lifetime of 125
days, human red blood cells are destroyed by leuko-
cytes in the spleen and liver.
The white blood cells or leukocytes are nearly a
thousandfold less numerous than red cells. About 7 x 10
6
cells are present per ml of blood. There are three types
of leukocytes: lymphocytes (~26% of the total),
monocytes (~7% of the total), and polymorphonuclear
leukocytes or granulocytes (~70% of the total).
Lymphocytes are about the same size as erythrocytes
and are made in lymphatic tissue. Individual lympho-
cytes may survive for as long as ten years. They func-
tion in antibody formation and are responsible for
maintenance of long-term immunity.
Monocytes, two times larger, are active in ingesting
bacteria. These cells stay in the blood only a short
time before they migrate into the tissues where they
become macrophages,
136
relatively fixed phagocytic
cells. Macrophages not only phagocytize and kill
invading bacteria, protozoa, and fungi but also destroy
cancer cells. They also destroy damaged cells and
cellular debris as part of the normal turnover of tissues.
They play an essential role in the immune system by
processing antigens and in releasing stimulatory
proteins.
Granulocytes of diameter 912 m are formed in
the red bone marrow. Three types are distinguished
by staining: neutrophils, eosinophils, and basophils.
Neutrophils are the most numerous phagocytic cells of
our blood and provide the first line of defense against
bacterial infections. The functions of eosinophils and
basophils are less well understood. The number of
eosinophils rises during attacks of hay fever and asthma
and under the influence of some parasites, while the
basophil count is increased greatly in leukemia and
also by inflammatory diseases. Granules containing
histamine, heparin, and leukotrienes are present in the
basophils. Blood platelets or thrombocytes are tiny
(23 m diameter) cell-like bodies essential for rapid
coagulation of blood. They are formed by fragmenta-
tion of the cytoplasm of bone marrow megakaryocytes.
One mature megakaryocyte may contribute 3000 plate-
lets to the 13 x 10
8
per ml present in whole blood.
Cell culture. Laboratory growth of isolated animal
cells has become very important in biochemistry.
137
Sometimes it is necessary to have many cells with as
nearly as possible identical genetic makeup. Such
bacterial cells are obtained by plating out the bacteria
and selecting a small colony that has grown from a
single cell to propagate a pure strain. Similarly,
single eukaryotic cells may be selected for tissue cul-
ture and give rise to a clone of cells which remains
genetically identical until altered by mutations.
The culture of embryonic fibroblasts is used to
obtain enough cells to perform prenatal diagnosis of
inherited metabolic diseases (Box 1-D). Tissue culture
is easiest with embryonic or cancer cells, but many
other tissues can be propagated. However, the cells
that grow best and which can be propogated indefi-
nitely are not entirely normal; the well-known HeLa
strain of human cancer cells which was widely grown
for many years throughout the world contains 7080
chromosomes per cell compared with the normal 46.
3. Communication
Plants are able to maintain their form because the
cells are surrounded by thick walls that cement the
cells together. However, animal cells lack rigid walls
and must be held together by specialized contacts.
138,139
Contacts between cells of both plants and animals are
27
Figure 1-15 Electron micrographs of cell junctions of three types. (A) Freeze-fractured zona occludens (occlusion zone)
between epithelial cells of the rat small intestine. The tight junctions are repesented as a meshwork of ridges (in the P or
protoplasmic fracture face) or grooves (in the E fracture face which looks toward the extracellular space). These represent the
actual sites of membrane fusion. Microvilli are seen in the lower part of the photograph. From D. S. Friend and N. B. Gilula.
141
(B) Thin cross section of tight junction between mouse hepatocytes. The arrows indicate points of membrane fusion. From
Gilula.
142
Copyright 1975 by The Williams & Wilkins Co., Baltimore. (C) A freeze-fractured septate junction from ciliated
epithelium of a mollusc. This type of junction forms a belt around the cells. Fracture face P (central depressed area) contains
parallel rows of membrane particles that correspond to the arrangmenet of the intracellular septa seen in thin sections. The
surrounding fracture face E contains a complementary set of grooves. Particles in nonjunctional membrane regions (upper
right corner) are randomly arranged. (D) Thin section of a septate junction of the type shown in (C). The plasma membranes
of the two cells are joined by a periodic arrangement of electron-dense bars or septa, which are present within the intercellular
space. Note the Golgi membranes in the lower right part of the photograph. (C) and (D) are from N. B. Gilula
143
A B
E. The Variety of Animal Forms
200 nm 100 nm
100 nm 200 nm
C D
28 Chapter 1. The Scene of Action
E F
100 nm
100 nm
Figure 1-15 (continued) (E) Desmosomes (macula adherens) in rat
intestinal epithelium. Features include a wide (2535 nm) intercellular
space containing dense material, two parallel cell membranes, a dense
plaque associated with the cytoplasmic surface and cytoplasmic tono-
filaments that converge on the dense plaque. From Gilula.
143
(F) Freeze-
fractured surface through gap junctions between communicating cells in
culture. Both a large junction and a smaller one below (arrow) can be
seen. (G) Gap junctions in thin section. (F) and (G) are from N. B. Gilula.
144
important for a second reason: Cells must communicate, one with
another.
Cell contacts and junctions. Many epithelial cells, e.g., those
lining the border of kidney tubules and secretory glands, form
tight junctions with adjacent cells. Electron microscopy shows
that in these junctions the outer portions of the membranes actual-
ly fuse in some places (Fig. 1-15). One way in which this has been
G
100 nm
29 F. Higher Plants and Plant Tissues
demonstrated is to freeze a tissue rapidly and to frac-
ture it in the frozen state within a vacuum chamber.
The fractured tissue is kept at about 100C in a vacuum
for a short time while water molecules evaporate from
the fractured surfaces. A thin plastic replica is then
made of the etched surfaces, which sometimes pass
through tight junctions revealing details of their struc-
ture (Fig. 1-15, A, E). Study of electron micrographs of
such surfaces shows that some cells are completely
surrounded by belts of tight junctions, sometimes
referred to as occlusion zones or terminal bars.
Tight junctions between endothelial cells of blood
capillaries in the brain prevent free diffusion of com-
pounds from the blood stream into brain cells and
form the bloodbrain barrier.
140
Tight junctions
between neurons and adjacent cells surround the
nodes of Ranvier (Chapter 30).
Contacts of another type, known as septate
desmosomes or adhesion discs, form a belt around
the cells of invertebrate epithelia. In these contacts a
space of ~18 nm between adjacent cell membranes is
bridged in a number of places by thin walls. Behind
the desmosomes the membrane is often backed up at
these points by an electron-dense region to which are
attached many fine microfilaments of ~610 nm
diameter (Fig. 1-15, D).
One method of communication between cells is
by passage of chemical substances through special
junctions which, because of their appearance in electron
micrographs of thin sections (Fig. 1-15, G) are known
as gap junctions.
139,145,146
Gap junctions may cover
substantial areas of the cell interface. In cross section,
a thin 34 nm gap between the adjacent cell membranes
is bridged by a lattice-like structure, which may appear
in freeze-fractured surfaces as a hexagonal array of
particles (Fig. 1-15, F, lower junction). These particles
or connexons are each thought to be composed of six
protein subunits. A central channel in the connexon is
able to pass molecules of molecular mass up to about
500 Da.
147,148
Small molecules may be able to pass
freely from one cell to another through the gap junction.
Because of their low electrical resistance, gap junctions
allow electrical coupling of cells. Such junctions
form the electrotonic synapses that link some neurons
to other excitable cells. Heart cells are all electrically
coupled through gap junctions.
149
Another type of communicating junction is also
found in synapses of the nervous system. At these
specialized contacts a nerve impulse transmitted along
the membrane of one neuron triggers the release of a
neurotransmitter, a chemical substance that passes
across the gap between cells of the synapse and initiates
a nerve impulse in the second neuron (Chapter 30).
Cell recognition. Cells of higher organisms are
able to recognize other cells as identical, as belonging
to another tissue, or as being foreign. This ability is
developed most highly by cells of the immune system
but is possessed to some extent by others. For example,
cells of sponges can be separated by partial digestion
of the protein cement that holds them together.
When dissociated cells from orange sponges were
mixed with those from yellow sponges, the cells
clumped together to reform small sponges.
150,151
Furthermore, orange cells stuck to orange cells and
yellow to yellow cells. Similar results have been
obtained using a mixture of cultivated liver, kidney,
and embryonic brain cells. When a wound heals,
epithelial cells grow and move across the wound
surface but they stop when they meet. Cells in tissue
culture and growing on a glass surface experience
this same contact inhibition
152
and spread to form
a unicellular layer. Cancer cells in culture do not stop
but climb one on top of the other, apparently lacking
proper recognition and communication. Many chem-
ical signals appear to pass between cells. An important
goal of contemporary biochemistry is to understand
how cells recognize each other and respond to signals
that they receive.
F. Higher Plants and Plant Tissues
Botanists recognize two divisions of higher plants.
The Bryophyta or moss plants consist of the Musci
(mosses) and Hepaticae (liverworts). These plants
grow predominantly on land and are characterized
by swimming sperm cells and a dominant gameto-
phyte (haploid) phase. Tracheophyta, or vascular
plants, contain conducting tissues. About 2 x 10
5
species are known. The ferns (class Filicineae, formerly
Pteridophyta) are characterized by a dominant diploid
plant and alternation with a haploid phase. Seed plants
are represented by two classes: Gymnosperms
(cone-bearing trees) and Angiosperms, the true
flowering plants.
Genetically the simplest of the angiosperms is
the little weed Arabidopsis thaliana, whose generation
time is as short as five weeks. Its five chromosomes
contain only 10
8
base pairs in all, the smallest known
genome among angiosperms
153
and one whose com-
plete nucleotide sequence is being determined. Its
biochemistry, physiology, and developmental biology
are under intensive study. It may become the fruit
fly of the plant kingdom.
There are several kinds of plant tissues. Undiffer-
entiated, embryonic cells found in rapidly growing
regions of shoots and roots form the meristematic
tissue. By differentiation, the latter yields the simple
tissues, the parenchyma, collenchyma, and scleren-
chyma. Parenchyma cells are among the most abun-
dant and least specialized in plants. They give rise
through further differentiation to the cambium layer,
the growing layer of roots and stems. They also
30 Chapter 1. The Scene of Action
make up the pith or pulp in the center of stems and
roots, where they serve as food storage cells.
The collenchyma, present in herbs, is composed
of elongated supporting cells and the sclerenchyma
of woody plants is made up of supporting cells with
hard lignified cell walls and a low water content.
This tissue includes fiber cells, which may be ex-
tremely long; e.g., pine stems contain fiber cells of 40
m diameter and 4 mm long.
Two complex tissues, the xylem and phloem,
provide the conducting network or circulatory system
of plants. In the xylem or woody tissue, most of the
cells are dead and the thick-walled tubes (tracheids)
serve to transport water and dissolved minerals from
the roots to the stems and leaves. The phloem cells
provide the principal means of downward conduction
of foods from the leaves. Phloem cells are joined end
to end by sieve plates, so-called because they are
perforated by numerous minute pores through which
cytoplasm of adjoining sieve cells appears to be con-
nected by strands 59 m in diameter.
154
Mature sieve
cells have no nuclei, but each sieve cell is paired with a
nucleated companion cell.
Epidermal tissue of plants consists of flat cells,
usually containing no chloroplasts, with a thick outer
wall covered by a heavy waxy cuticle about 2 m thick.
Only a few specialized cells are found in the epidermis.
Among them are the paired guard cells that surround
the small openings known as stomata on the under-
surfaces of leaves and control transpiration of water.
Specialized cells in the root epidermis form root hairs,
long extensions (~1 mm) of diameter 517 m. Each
hair is a single cell with the nucleus located near the tip.
Figure 1-16 shows a section from a stem of a typical
angiosperm. Note the thin cambium layer between
the phloem and the xylem. Its cells continuously
undergo differentiation to form new layers of xylem
increasing the woody part of the stem. New phloem
cells are also formed, and as the stem expands all of
the tissues external to the cambium are renewed and
the older cells are converted into bark.
Plant seeds consist of three distinct portions.
The embryo develops from a zygote formed by fusion
of a sperm nucleus originating from the pollen and
an egg cell. The fertilized egg is surrounded in the
gymnosperms by a nutritive layer or endosperm
which is haploid and is derived from the same game-
tophyte tissue that produced the egg. In angiosperms
two sperm nuclei form; one of these fertilizes the egg,
while the other fuses with two haploid polar nuclei
derived from the female gametophyte. (The polar nuclei
are formed by the same mitotic divisions that formed
the egg.) From this develops a 3n triploid endosperm.
G. The Chemical Composition of Cells
Water is the major component of living cells, but
the amount varies greatly. Thus, the pig embryo is
97% water; at birth a new-born pig is only 89% water.
A lean 45-kg pig may contain 67% water but a very fat
135-kg animal only 40% water. Similar variations are
encountered with other constitutents.
The water content of a tissue is often determined
by thoroughly drying a weighed sample of tissue at
low temperature in vacuum and then weighing it a
second time. The solid material can then be extracted
with a solvent that will dissolve out the fatty com-
pounds. These are referred to collectively as lipids.
After evaporation of the solvent the lipid residue may
be weighed. By this procedure a young leafy vegetable
might be found to contain 25% lipid on a dry weight
Figure 1-16 Section of the stem of an angiosperm. Enlarged sections showing tubes of the phloem (left) and xylem (right).
From S. Biddulph and O. Biddulph.
155
Drawn by Bunji Tagawa.
Ray
Sieve
tube
Sieve
plate
Fiber
Ray
Fiber
Vessel
Ray
Xylem
Pith
Phloem
Cambium
31
basis. Even very lean meats contain 1030% lipid.
The residue remaining after removal of the lipid
consists predominately of three groups of compounds:
proteins, nucleic acids, and carbohydrates. Most of
the nitrogen present in tissues is found in the proteins
and the protein content is sometimes estimated by
determining the percentage of nitrogen and multiplying
by 6.25. In a young green plant, 2030% of the dry
matter may be protein, while in very lean meat it may
reach 5070%.
A dried tissue sample may be burned at a high
temperature to an ash, which commonly amounts to
310% and is higher in specialized tissues such as
bone. It is a measure of the inorganic constituents of
tissues.
The carbohydrate content can be estimated by the
difference of the sum of lipid, protein, and ash from
100%. It amounts to 5060% in young green plants
and only 210% in typical animal tissues. In excep-
tional cases the carbohydrate content of animal tissues
may be higher; the glycogen content of oysters is 28%.
The amount of nucleic acid in tissues varies from
0.1% in yeast and 0.51% in muscle and in bacteria to
1540% in thymus gland and sperm cells. In these latter
materials of high nucleic acid content it is clear that
multiplication of % N by 6.25 is not a valid measure of
protein content. For diploid cells of the body the DNA
content per cell is nearly constant.
Table 1-4 compares the composition of a bacterium,
of a green plant, and of an active animal tissue (rat
liver). Although the solid matter of cells consists
principally of C, H, O, N, S, and P, many other chemical
elements are also present. Among the cations, Na
+
, K
+
,
Ca
2+
, and Mg
2+
are found in relatively large amounts.
Thus, the body of a 70 kg person contains 1050 g Ca
(mostly in the bones), 245 g K, 105 g Na, and 35 g Mg.
Iron (3 g), zinc (2.3 g), and rubidium (1.2 g) are the next
most abundant. Of these iron and zinc are essential to
life but rubidium is probably not. It is evidently taken up
by the body together with potassium.
The other metallic elements in the human body
amount to less than 1 g each, but at least seven of them
play essential roles. They include copper (100 mg),
manganese (20 mg), and cobalt (~5 mg). Others, such
as chromium (<6 mg), tin, and vanadium, have only
recently been shown essential for higher animals.
156,157
Nickel, lead, and others may perhaps be needed.
Nonmetallic elements predominating in the ash
are phosphorus (700 g in the human body), sulfur
(175 g), and chlorine (105 g). Not only are these three
elements essential to all living cells but also selenium,
fluorine, silicon (Box 4-B), iodine, and boron are needed
by higher animals and boron by plants (Fig. 1-17).
Iodine deficiency may affect one billion human beings
and may cause 20 million cases per year of cretinism,
or less severe brain damage.
158
What is the likelihood that other elements will be
found essential? Consider a human red blood cell, an
object of volume ~80 m
3
and containing about 3 x 10
8
protein molecules (mostly hemoglobin). About 7 x 10
5
atoms of the trace metal copper and 10
5
atoms of the
nutritionally essential tin are present in a single red
cell. Also present are 2 x 10
4
atoms of silver, a toxic
metal. Its concentration, over 10
-7
M, is sufficient that
it could have an essential catalytic function. However,
we know of none and it may simply have gotten into
our bodies from handling money, jewelry, and other
G. The Chemical Composition of Cells
TABLE 1-4
Approximate Composition of Metabolically
Active Cells and Tissues
a
Component
Rat
liver
d
(%)
Green plant
(spinach,
Spinacia
oleracea)
c
E. coli
b
(%)
H
2
O 70 93 69
Protein 15 2.3 21
Amino acids 0.4
DNA 1 0.2
RNA 6 1.0
Nucleotides 0.4
Carbohydrates 3 3.2
Cellulose 0.6
Glycogen 3.8
Lipids 2 0.3 6
Phospholipids 3.1
Neutral lipids 1.6
Sterols 0.3
Other small molecules 0.2
Inorganic ions 1 1.5
K
+
0.4
Equivalents per liter in rat liver
Amino acid residues 2.1
Nucleotide units 0.03
Glycogen (glucose units) 0.22
K
+
0.1
a
Data were not readily available for spaces left blank
b
From J. D. Watson (1976) Molecular Biology of the Gene, 3rd ed.,
p. 69, Benjamin, New York The amounts of amino acids,
nucleotides, carbohydrates, and lipids include precursors present
in the cell.
c
From B. T. Burton (1976) Human Nutrition, 3rd ed., McGraw-Hill,
New York (p. 505)
d
From C. Long, ed., (1961) Biochemists Handbook, pp. 677679, Van
Nostrand-Reinhold, Princeton, New Jersey
32 Chapter 1. The Scene of Action
BOX 1-F ABOUT THE REFERENCES
a
These are not journals but series of review and reference books. There are many other series of Advances in... and Annual
Reviews of ... that are not listed here.
b
EMBOEuropean Molecular Biology Organization
c
FASEBFederation of American Societies for Experimental Biology
Full Title Abbreviation
Advances in Carbohydrate Chemistry and Biochemistry
a
Adv. Carbohydr. Chem. Biochem.
Advances in Protein Chemistry
a
Adv. Protein Chem.
Analytical Biochemistry Anal. Biochem.
Annual Review of Biophysics and Biomolecular Structure
a
Ann. Rev. Biophys. Biomolec. Struct.
Annual Review of Biochemistry
a
Ann. Rev. Biochem.
Archives of Biochemistry and Biophysics Arch. Biochem. Biophys.
Biochemical and Biophysical Research Communications Biochem. Biophys. Res. Commun.
Biochemical Journal Biochem. J.
Biochemistry
Biochimica et Biophysica Acta Biochim. Biophys. Acta
Bioorganic Chemistry Bioorg. Chem.
Carbohydrate Research Carbohydr. Res.
EMBO Journal
b
EMBO J.
European Journal of Biochemistry Eur. J. Biochem.
FASEB Journal
c
FASEB J.
Journal of Bacteriology J. Bacteriol.
Journal of Biochemistry J. Biochem.
Journal of Biological Chemistry J. Biol. Chem.
Journal of Lipid Research J. Lipid Res.
Journal of Molecular Biology J. Mol. Biol.
Journal of the American Chemical Society J. Am. Chem. Soc.
Journal of Theoretical Biology J. Theor. Biol.
Methods in Enzymology
a
Methods Enzymol.
Nature
Nucleic Acids Research Nucleic Acids Res.
Proceedings of the National Academy of Sciences, USA Proc. Natl. Acad. Sci. U.S.A.
Science
Structure
Trends in Biochemical Sciences Trends Biochem. Sci. or TIBS
The lists of references at the ends of chapters
are provided to encourage readers to look at origi-
nal research articles. The lists are neither complete
nor critically selected, but they do increase the
information given in this book many-fold. I apolo-
gize for the important papers omitted. However,
the references that are here will help a student to
get started in reading the literature. Each reference
contains other references and names of persons
active in the field. By searching recent journal
indices or a computer database it is easy to find
additional articles by the same authors or on the
same subject.
Look at the various types of scientific articles
including reviews, preliminary reports and full
research papers. Be sure to examine those in the
primary source journals which publish detailed
research results. These articles have always been
sent to referees, active scientists, who check to see
that the experiments are described accurately, that
the authors have cited relevant literature, and that
the conclusions are logical. Some journals, e.g.,
Biochem. Biophys. Res. Comm. and FEBS Letts. are
dedicated to rapid publication of short reports but
are also refereed. Other journals provide mostly
reviews or a mixture of reviews. Periodical review
series, such as Advances in Nucleic Acid Chemistry
and Related Topics, often appear annually. Every
student who intends to become a professional
biochemist should consider purchasing the Annual
Review of Biochemistry each year. This indispensible
source of current information on most aspects of
biochemistry is available to students at a very low
price.
Many journal papers are difficult to read. To
start, pick papers that have an understandable
introduction. Choose reviews that are short, such
as those in Trends in Biochemical Sciences. Then go
on to the more comprehensive ones. Never sit back
and hope that your computer will automatically
fetch just what you need! Many journals carry papers
of biochemical importance. Those specializing in
biochemistry include the following:
33 G. The Chemical Composition of Cells
Essential to all animals
and plants
Essential to several classes
of animals and plants
Believed essential to a
variety of species
Possible essential trace
elements for some species
H
K
C N
P S
O
Mg
Ca Mn Fe Cu Zn
B F
Br Se
Cd
V
Mo
Co
Li Be
Sc Ti
Hf
Y
La
Zr
Na
R
Cs
Sr
Ba
Nb
Ta W
Cr
Re
Tc Ru
Os Ir
Rh
Pt
Pd
Ni
Au
Ag
Hg Tl
In
Ga
Al
Ge
Sn
Pb Bi
S
As
Po
Te
At Rn
Xe
Kr
Ar
Ne
He
Cl
I
Si
Figure 1-17 Elements known to be essential to living things (after da Silva and Williams
157
). Essential elements are enclosed
within shaded boxes. The 11 elementsC, H, O, N, S, P, Na, K, Mg, Ca, and Cl make up 99.9% of the mass of a human being.
An additional 13 are known to be essential for higher animals in trace amounts. Boron is essential to higher plants but
apparently not to animals, microorganisms, or algae.
silver objects. The red blood cell also contains boron
and aluminum (3 x 10
5
atoms each), arsenic (7 x 10
5
atoms), lead (7 x 10
4
atoms), and nickel (2 x 10
4
atoms).
Of the elements (uranium and below) in the periodic
table, only four (Ac, Po, Pa, and Ra) are present, on the
average, in quantities less than one atom per cell.
156
Of the apparently nonessential elements, several,
e.g., Cs, Rb, Sr, and Ni (possibly essential) are not toxic
at low concentrations. Others, such as Sb, As, Ba, Be,
Cd, Pb, Hg, Ag, Tl, and Th, are highly toxic.
The ionic compositions of tissues and of body fluids
vary substantially. Blood of marine organisms is similar
to that of seawater in its content of Na
+
, Cl
-
, Ca
2+
, and
Mg
2+
. Blood of freshwater and terrestrial organisms
contains about ten times less Na
+
and Cl
-
and several
times less Ca
2+
and Mg
2+
than is present in seawater,
but it is nevertheless relatively rich in these ions.
In general, cells are rich in K
+
and Mg
2+
, the K
+
predominating by far, and are poor in Na
+
and Ca
2+
.
Chloride is the principal inorganic anion, but organic
carboxylate and phosphate groups contribute most of
the negative charges (Table 1-4), many of which are
fixed to proteins or other macromolecules. Ling esti-
mated that cells typically contain about 1.66 M of amino
acid residues in their proteins. Of these residues, 10%
have negatively charged side chains and 8% positively
charged. The difference is a net negative charge
amounting to 33 mM within cells.
159.
34 Chapter 1. The Scene of Action
1. May, R. M. (1992) Sci. Am. 267(Oct), 4248
2. Stanier, R. Y., Ingraham, J. L., Wheelis, M. L.,
and Painter, P. R. (1986) The Microbial World,
5th ed., Prentice-Hall, Englewood Cliffs, New
Jersey
3. Brock, T. D., Smith, D. W., and Madigan, M. T.
(1988) Biology of Microorganisms, 5th ed.,
Prentice-Hall, Englewood Cliffs, New Jersey
4. Neidhardt, F. C., ed. (1987) Escherichia coli and
Salmonella typhimurium, Am. Soc. for
Microbiology, Washington, D.C.
5. Neidhardt, F. C., Ingraham, J. L., and
Schaechter, M. (1990) Physiology of the Bacterial
Cell, Sinauer, Sunderland, Mass.
6. Maniloff, J., and Morowitz, H. J. (1972)
Bacteriol. Rev. 36, 263290
7. Razin, S. (1978) Microbiol. Rev. 42, 414470
8. Barile, M. F., Razin, S., Tully, J. G., and
Whitcomb, R. F., eds. (1979) The Mycoplasmas,
Vol. I, Academic Press, New York
9. Watson, J. D. (1976) Molecular Biology of the
Gene, 3rd ed., Benjamin, Menlo Park,
California (p. 61)
10. Horowitz, D. M., and Sanders, J. K. M. (1994)
J. Am. Chem. Soc. 116, 26952702
10a. Gansheroff, L. J., and OBrien, A. D. (2000)
Proc. Natl. Acad. Sci. U.S.A. 97, 2959 2961
11. Fraser, C. M., Gocayne, J. D., White, O.,
Adams, M. D., Clayton, R. A., Fleischmann, R.
D., Bult, C. J., Kerlavage, A. R., Sutton, G.,
Kelley, J. M., Fritchman, J. L., Weidman, J. F.,
Small, K. V., Sandusky, M., Fuhrmann, J.,
Nguyen, D., Utterback, T. R., Saudek, D. M.,
Phillips, C. A., Merrick, J. M., Tomb, J.-F.,
Dougherty, B. A., Bott, K. F., Hu, P.-C., Lucier,
T. S., Peterson, S. N., Smith, H. O., Hutchison,
C. A., III, and Venter, J. C. (1995) Science 270,
397403
12. Sonea, S. (1988) Nature (London) 331, 216
13. Costerton, J. W., Geesey, G. G., and Cheng, K.-
J. (1978) Sci. Am. 238(Jan), 8695
14. Macnab, R. M. (1987) in Escherichia coli and
Salmonella typhimurium (Niedhardt, F. C., ed),
pp. 7083, Am. Soc. for Microbiology,
Washington, D.C.
15. Margulis, L., Ashen, J. B., Sonea, S., Sol, M.,
and Guerrero, R. (1993) Proc. Natl. Acad. Sci.
U.S.A. 90, 69666970
16. Goldstein, S. F., Charon, N. W., and Kreiling,
J. A. (1994) Proc. Natl. Acad. Sci. U.S.A. 91,
34333437
17. Levine, M. M. (1985) N. Engl. J. Med. 313,
445447
18. Kuehn, M. J., Heuser, J., Normark, S., and
Hultgren, S. J. (1992) Nature (London) 356,
252255
19. Alper, J. (1993) Science 262, 1817
20. Holt, J. G., Krieg, N. R., Sneath, P. H. A.,
Staley, J. T., and Williams, S. T., eds. (1994)
Bergeys Manual of Determinative Bacteriology,
9th ed., Williams & Wilkins, Baltimore,
Maryland
21. Doolittle, R. F. (1992) Protein Sci. 1, 191 200
22. Schwartz, R. M., and Dayhoff, M. O. (1978)
Science 199, 395403
23. Cavalier-Smith, T. (1986) Nature (London) 324,
416417
24. Jones, W. J., Nagle, D. P., Jr., and Whitman, W.
B. (1987) Microbiol. Rev. 51, 135177
25. Woese, C. R. (1987) Microbiol. Rev. 51, 221271
26. Sogin, M. L., Gunderson, J. H., Elwood, H. J.,
Alonso, R. A., and Peattie, D. A. (1989) Science
243, 7577
27. Garrett, R. A., Dalgaard, J., Larson, N., Kjems,
J., and Mankin, A. S. (1991) Trends Biochem.
Sci. 16, 2226
28. Searcy, D. G. (1982) Trends Biochem. Sci. 7,
183185
29. Wheelis, M. L., Kandler, O., and Woese, C. R.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89,
29302934
30. Woese, C. R., Kandler, O., and Wheelis, M. L.
(1990) Proc. Natl. Acad. Sci. U.S.A. 87, 4576
4579
30a. Doolittle, W. F. (1999) Science 284, 2124 2128
30b. Pennisi, E. (1999) Science 284, 1305 1307
31. Cavalier-Smith, T. (1992) Nature (London) 356,
570
32. Angert, E. R., Clements, K. D., and Pace, N. R.
(1993) Nature (London) 362, 239241
33. Thimann, K. V. (1963) The Life of Bacteria, 2nd
ed., Macmillan, New York
34. Stanier, R. Y., and Cohen-Bazire, G. (1977)
Ann. Rev. Microbiol. 31, 225274
35. Morden, C. W., and Golden, S. S. (1989)
Nature (London) 337, 382385
36. Marme, D., Marre, E., and Hertel, R., eds.
(1982) Plasmalemma and Tonoplast: Their
Functions in the Plant Cell, Elsevier, Amsterdam
37. Collins, F. S. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 1082110823
38. Hinshaw, J. E., Carraghen, B. O., and
Milligan, R. A. (1992) Cell 69, 11331141
39. Dingwall, C., and Lasky, R. (1992) Science 258,
942947
40. Nolta, K. V., Padh, H., and Steck, T. L. (1991)
J. Biol. Chem. 266, 1831818323
41. de Duve, C. (1975) Science 189, 186194
42. Griffiths, G., and Simons, K. (1986) Science
234, 438443
43. Hartley, S. M. (1992) Trends Biochem. Sci. 17,
325327
44. Golgi, C. (1898) Arch. Ital. Biol. 30, 60 and 278
45. Kirk, J. T. O., and Tilney-Bassett, R. A. E.
(1978) The Plastids: Their Chemistry, Structure,
Growth, and Inheritance, 2nd ed., Elsevier-
North Holland Biomedical Press, Amsterdam
46. Hoffmann, H. P., and Avers, C. J. (1973)
Science 181, 749751
46a. Capaldi, R. A. (2000) Trends Biochem. Sci. 25,
212 214
46b. Rutter, G. A., and Rizzuto, R. (2000) Trends
Biochem. Sci. 25, 215 221
47. Fukui, S., and Tanaka, A. (1979) Trends
Biochem. Sci. 4, 246249
48. Kindl, H., and Lazarow, P. B., eds. (1982)
Peroxisomes and Glyoxysomes, Vol. 386, Annals
of the New York Academy of Sciences, New
York
49. de Duve, C. (1983) Sci. Am. 248(May), 7484
50. Masters, C., and Crane, D. (1995) The
Peroxisome: A Vital Organelle, Cambridge Univ.
Press, London
51. Lord, J. M., and Roberts, L. M. (1980) Trends
Biochem. Sci. 5, 271274
52. Cerkasov, J., Cerkasovova, A., Kulda, J., and
Vilhelmova, D. (1978) J. Biol. Chem. 253, 1207
1214
53. Glover, D. M., Gonzalez, C., and Raff, J. W.
(1993) Sci. Am. 268(Jun), 6268
54. Olmsted, J. B., and Borisy, G. G. (1973) Ann.
Rev. Biochem. 42, 507540
55. Hall, J. L., Ramanis, Z., and Luck, D. J. L.
(1989) Cell 59, 121132
56. Hyams, J. S. (1989) Nature (London) 341, 485
486
57. Satir, P. (1961) Sci. Am. 204(Feb), 108116
58. Lazarides, E. (1980) Nature (London) 283, 244
256
59. Amos, L. A., and Amos, W. B. (1991) Molecules
of the Cytoskeleton, Guilford, New York
60. Iwabe, N., Kuma, K., Hasegawa, M., Osawa,
S., and Miyata, T. (1989) Proc. Natl. Acad. Sci.
U.S.A. 86, 93559359
61. Lake, J. A. (1991) Trends Biochem. Sci. 16, 4650
References
62. Margulis, L. (1981) Symbiosis in Cell Evolution,
Freeman, San Francisco, California
63. Hurst, L. D. (1994) Nature (London) 369, 451
64. Gupta, R. S., and Golding, G. B. (1996) Trends
Biochem. Sci. 21, 166171
65. de Duve, C. (1996) Sci. Am. 274 (Apr), 5057
66. Markert, C. L., Shaklee, J. B., and Whitt, G. S.
(1975) Science 189, 102114
67. Fenchel, T., and Bernard, C. (1993) Nature
(London) 362, 300
68. Schopf, J. W. (1993) Science 260, 640646
69. Horgan, J. (1993) Sci. Am. 269(Aug), 24
70. Doolittle, W. F. (1980) Trends Biochem. Sci. 5,
146149
71. May, R. M. (1986) Nature (London) 324, 514515
72. Seger, J. (1989) Nature (London) 337, 305306
73. Riley, M., and Anilionis, A. (1978) Ann. Rev.
Microbiol. 32, 519560
74. Goossens, M., Dozy, A. M., Embury, S. H.,
Zachariades, Z., Hadjiminas, M. G.,
Stamatoyannopoulos, G., and Kan, Y. W.
(1980) Proc. Natl. Acad. Sci. U.S.A. 77, 518521
75. Williams, R. O., Young, J. R., and Majiwa, P.
A. O. (1979) Nature (London) 282, 847849
76. Kondrashov, A. S. (1988) Nature (London) 336,
435440
77. Buchsbaum, R., Buchsbaum, M., Pearse, J.,
and Pearse, V. (1987) Animals Without
Backbones, 3rd ed., Univ. Chicago Press,
Chicago, Illinois
78. Margulis, L., and Schwartz, K. V. (1999) Five
Kingdoms: An Illustrated Guide to the Phyla of
Life on Earth, 3rd ed., Freeman, New York
79. Laver, W. G., Air, G. M., Dopheide, T. A., and
Ward, C. W. (1980) Nature (London) 283, 454
457
80. McKanna, J. A. (1973) Science 179, 8890
81. Miller, L. H. (1992) Science 257, 3637
82. Wyler, D. J. (1993) N. Engl. J. Med. 329, 3137
83. Miller, L. H., Good, M. F., and Milon, G.
(1994) Science 264, 18781883
84. Nussenzweig, R. S., and Long, C. A. (1994)
Science 265, 13811382
85. Sibley, L. D., and Boothroyd, J. C. (1992) Nature
(London) 359, 8285
86. Soldati, D., and Boothroyd, J. C. (1993) Science
260, 349352
87. Donelson, J. E., and Turner, M. J. (1985) Sci.
Am. 252(Feb), 4451
88. Blum, M. L., Down, J. A., Gurnett, A. M.,
Carrington, M., Turner, M. J., and Wiley, D. C.
(1993) Nature (London) 362, 603609
89. Gall, J. G., ed. (1986) The Molecular Biology of
Ciliated Protozoa, Academic Press, Orlando,
Florida
90. Olive, L. S. (1975) The Mycetozoans, Academic
Press, New York
91. Kimmel, A. R., ed. (1989) Molecular Biology of
Dictyostelium Development, Liss, New York
92. Traynor, D., Kessin, R. H., and Williams, J. G.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89, 8303
8307
93. Webster, J. (1980) Introduction to Fungi, 2nd
ed., Cambridge Univ. Press, New York
94. Davies, D. A. L., and Pope, A. M. S. (1978)
Nature (London) 273, 235236
95. Georgopapadakou, N. H., and Walsh, T. J.
(1994) Science 264, 371373
96. Sternberg, S. (1994) Science 266, 16321634
97. Davis, R. H., and DeSerres, F. J. (1970) Meth.
Enzymol. 17A, 79148
98. Botstein, D., and Fink, G. R. (1988) Science 240,
14391443
99. Pringle, J. R., Broach, J. R., and Jones, E. W.,
eds. (19911994) The Molecular and Cellular
Biology of the Yeast Saccharomyces, Cold
Springs Harbor Laboratory Press, Cold
Spring Harbor, New York (3 Vols.)
35
100. Wheals, A. E., Rose, A. H., and Harrison, S. J.,
eds. (1995) The Yeasts, 2nd ed., Vol. 6,
Academic Press, San Diego, California
101. Gimeno, C. J., Ljungdahl, P. O., Styles, C. A.,
and Fink, G. R. (1992) Cell 68, 10771090
102. Oliver, S. G., and 146 other authors (1992)
Nature (London) 357, 3846
103. Williams, N. (1995) Science 268, 15601561
104. Ruehle, J. L., and Marx, D. H. (1979) Science
206, 419422
105. Canter-Lund, H., and Lund, J. W. G. (1995)
Freshwater Algae: Their Microscopic World
Explored, Biopress, Bristol, England
106. Harris, E. H. (1989) The Chlamydomonas
Sourcebook, Academic Press, San Diego,
California
107. Brachet, J. L. A. (1965) Endeavour 24, 155161
108. Bonotto, S., Kefeli, V., and Puiseux-Dao, S.
(1979) Developmental Biology of Acetabularia,
Elsevier-North Holland, Amsterdam
109. Berger, S., and Kaever, M. J. (1992)
Dasycladales: An Illustrated Monograph of a
Fascinating Algal Order, Oxford Univ. Press,
New York
110. Menzel, D., and Elsner-Menzel, C. (1990)
Protoplasm 157, 5263
111. Jaenicke, L. (1982) Trends Biochem. Sci. 7, 6164
112. Ahmadjian, V. (1962) in Physiology and
Biochemistry of Algae, Vol. 2 (Lewin, R. A., ed),
pp. 817822, Academic Press, New York
113. Morris, S. C. (1993) Nature (London) 361, 219
221
114. Levinton, J. S. (1992) Sci. Am. 267(Nov), 8491
115. Keese, P. K., and Gibbs, A. (1992) Proc. Natl.
Acad. Sci. U.S.A. 89, 94899493
116. Hickman, C. P. (1973) Biology of the Invertebrates,
Mosby, St. Louis, Missouri
117. Villee, C. A., Walker, W. F., Jr., and Barnes, R.
D. (1973) General Zoology, Saunders,
Philadelphia, Pennsylvania
118. Lapan, E. A., and Morowitz, H. (1972) Sci. Am.
227(Dec), 94101
119. Lapan, E. A. (1975) Comp. Biochem. Physiol.
52A, 651657
120. Bergquist, P. R. (1978) Sponges, Hutchinson,
London
121. Grimmelikhuijzen, C. J. P., and Schaller, H. C.
(1979) Trends Biochem. Sci. 4, 265267
122. Lenhoff, H. M., and Lenhoff, S. G. (1988) Sci. Am.
258(Apr), 108113
123. Loomis, W. F. (1959) Sci. Am. 200(Apr), 150
123a. Hickman, C. P. Integrated Principles of Zoology
(1966) Mosby, St. Louis
124. Trager, W. (1986) Living Together: The Biology of
Animal Parasitism, Plenum, New York
125. Cherfas, J. (1991) Science 251, 630631
126. Kolberg, R. (1994) Science 264, 18591861
127. Wood, W. B., ed. (1988) The Nematode
Caenorhabditis elegans, Cold Spring Harbor
Lab. Press, Cold Spring Harbor, New York
128. Kenyon, C. (1988) Science 240, 14481453
129. Roberts, L. (1990) Science 248, 13101313
129a. Hotez, P. J., and Pritchard, D. I. (1995) Sci. Am.
272(Jun), 68 74
130. Dumont, H. J., and Green, J., eds. (1980)
Rotatoria, p. 42, Junk, The Hague
131. Mill, P. J., ed. (1978) Physiology of Annelids,
Academic Press, New York
132. Florkin, M., and Scheer, B. T., eds. (1970, 1971)
Chemical Zoology, Vol. 5 and 6, Academic
Press, New York
133. Goldstein, L. S. B., and Fyrberg, E. A., eds.
(1994) Drosophila melanogaster: Practical Uses in
Cell and Molecular Biology, Academic Press,
San Diego, California
134. Lawrence, P. (1992) The Making of a Fly: The
Genetics of Animal Design, Blackwell, Oxford
134a. Rubin, G. M., and Lewis, E. B. (2000) Science
287, 22162218
135. Maddy, A. H. (1992) Trends Biochem. Sci. 17,
125126
135a. Harris, J. R., ed. (1991) Blood Cell Biochemistry,
Plenum, New York (Three volumes)
136. Johnston, R. B., Jr. (1988) N. Engl. J. Med. 318,
747754
137. Freshney, R. I. (1994) Culture of Animal Cells,
3rd ed., Wiley-Liss, New York
138. Feldman, J., Gilula, N. B., and Pitts, J. D., eds.
(1978) Intercellular Junctions and Synapses,
Halsted Press, New York
139. Staehelin, L. A., and Hull, B. E. (1978) Sci. Am.
238(May), 141153
140. Bradbury, M. (1979) The Concept of a Blood-
Brain Barrier, Wiley, Chichester, New York
141. Friend, D. S., and Gilula, N. B. (1972) J. Cell
Biol. 53, 771
142. Gilula, N. B. (1975) Cellular Membranes and
Tumor Cells, Williams & Wilkins Co.,
Baltimore, Maryland (p. 221)
143. Gilula, N. B. (1974) in Cell Communication
(Cox, R. P., ed), pp. 129, Wiley, New York
144. Gilula, N. B. (1972) Nature (London) 235, 262
265
145. Zimmer, D. B., Green, C. R., Evans, W. H., and
Gilula, N. B. (1987) J. Biol. Chem. 262, 7751
7763
146. Hertzberg, E. L., and Johnson, R. G., eds.
(1988) Gap Junctions, Liss, New York
147. Robinson, S. R., Hampson, E. C. G. M.,
Munro, M. N., and Vaney, D. I. (1993) Science
262, 10721074
148. Stauffer, P. L., Zhao, H., Luby-Phelps, K.,
Moss, R. L., Star, R. A., and Muallem, S. (1993)
J. Biol. Chem. 268, 1976919775
149. Veenstra, R. D., and DeHaan, R. L. (1986)
Science 233, 272274
150. Moscona, A. A. (1961) Sci. Am. 205(Sep), 143
162
151. Alberts, B., Bray, D., Lewis, J., Raff, M.,
Roberts, K., and Watson, J. D. (1983) Molecular
Biology of the Cell, pp. 679680, Garland Publ.,
New York
152. Heaysman, J. E. M. (1978) Intl. Rev. Cytol. 55,
4966
153. Palca, J. (1989) Science 245, 131
154. Lloyd, A. M., Barnason, A. R., Rogers, S. G.,
Byrne, M. C., Fraley, R. T., and Horsch, R. B.
(1986) Science 234, 464466
155. Biddulph, S., and Biddulph, O. (1959) Sci. Am.
200(Feb), 4449
156. Bowen, H. J. M. (1966) Trace Elements in
Biochemistry, Academic Press, New York
157. Frasto da Silva, J. J. R., and Williams, R. J. P.
(1991) The Biological Chemistry of the Elements:
The Inorganic Chemistry of Life, Clarendon
Press, Oxford
158. Hetzel, B. S. (1994) N. Engl. J. Med. 331, 1770
1771
159. Ling, G. N. (1962) A Physical Theory of the
Living State, Ginn (Blaisdell), Boston,
Massachusetts
References
36 Chapter 1. The Scene of Action
1. Describe the principal structural or organizational
differences between prokaryotic and eukaryotic
cells.
2. Describe two or more principal functions of proteins
within cells, one function of DNA, two or more
functions of RNA, and one function of lipids.
3. Compare the chemical makeup of ribosomes, of
cell membranes, and of bacterial flagella.
4. Assume the following dimensions: Mycoplasma,
sphere, 0.33 m diameter; E. coli, cylinder, 0.8 m
diameter x 2 m; liver cell, sphere 20 m; root hair,
cylinder, 10 m diameter x 1 mm.
a. Calculate for each cell the total volume, the
mass in grams and in daltons (assume a specific
gravity of 1.0).
b. Assume that bacterial ribosomes are approxi-
mately spherical with a diameter of 23 nm.
What is their volume? If the mass of a bacterial
ribosome is 2.7 x 10
6
daltons, what is its appar-
ent density (divide mass by volume)? Experi-
mentally the buoyant density of bacterial
ribosomes in a cesium chloride gradient
(Chapter 5) is about 1.6 g/cm
3
. How can this
difference be explained? If eukaryotic ribo-
somes are 1.17 times larger than bacterial
ribosomes in linear dimensions, what is the
volume of a eukaryotic ribosome?
c. What fraction of volume of E. coli consists of cell
wall, of plasma membrane, of ribosomes (assume
15,000 are present)? If a cell of E. coli is 80%
water, what fraction by weight of the total solids
consists of ribosomes? Of DNA (assuming 2
chromosomes per cell)?
d. What fraction by volume of a liver cell is
composed of ribosomes, of plasma membrane,
of mitochondria (assume 1000 mitochondria)?
What fraction is accounted for by the nucleus?
5. a. What is the molar concentration of an enzyme
of which only one molecule is present in an
E. coli cell?
b. Assume that the concentration of K
+
within an
E. coli cell is 150 mM. Calculate the number of
K
+
ions in a single cell.
c. If the pH inside the cell is 7.0, how many H
+
ions are present?
6. If chromosomes (and chromatin) are 15% DNA,
what will be the mass of 23 pairs of chromosomes
in a human diploid cell? If the nucleus has a
diameter of 5 mm and a density of 1.1 g/cm
3
, what
fraction by weight of the nucleus is chromatin?
7. Compare the surface to volume ratios for an E. coli
cell, a liver cell, the nucleus of a eukaryotic cell, a
root hair. If a cell of 20 m diameter is 20%
covered with microvilli of 0.1 m diameter and 1
m length centered on a 0.2 m spacing, how
much will the surface/volume ratio be increased?
8. It has been shown that the code for specifying a
particular amino acid in a protein is determined
by a sequence of three nucleotides (a codon) in a
DNA chain. There are four different kinds of
nucleotide units in DNA. How many different
codons exist? Note that this is larger than the
number of different amino acids (20) that are
incorporated into proteins plus the three stop
(termination) codons (see Tables 5-5 and 5-6 for a
list of codons).
Study Questions
37
9. State two similarities and two differences between
cyanobacteria (blue-green algae) and green algae.
10. Compare the sizes and structures of bacterial and
eukaryotic flagella.
11. How much larger in volume is a typical eukaryotic
cell compared to a bacterium?
12. Compare the structure and properties of mito-
chondria, chloroplasts, and peroxisomes.
13. What are the possible origins of mitochondria and
chloroplasts? What evidence can you cite to
support your answer?
14. How many different kinds of polymers, e.g.
proteins, RNA, that are present in or around living
cells, can you name? Can you name some sub-
groups in any of your categories?
15. Compare the composition of these three, especially
with respect to C, H, O, N, S, P, Fe, Cu, Al, and Si:
a. The earths crust
b. Ocean water
c. Cytoplasm
Study Questions
38 Chapter 2. Amino Acids, Peptides, and Proteins
Contents
A. Structural Principles for Small Molecules
1. Bond Angles
2. Bond Lengths
3. Contact Distances
4. Asymmetry: Right-Handed and Left-Handed
Molecules
The D- and L- families of amino acids
The RS notation for configuration
Diastereoisomers
Geometrical isomers
5. Conformations: The Shapes that Molecules can
Assume
6. Tautomerism and Resonance
B. Forces between Molecules and between
Chemical Groups
1. Van der Waals Forces
2. Attraction between Charged Groups
(Salt Linkages)
3. Hydrogen Bonds
4. The Structure and Properties of Water
5. Hydration of Polar Molecules and Ions
6. Hydrophobic Interactions
C. Amino Acids and Peptides
1. Properties of -Amino Acids
2. Acidic and Basic Side Chains
3. The Peptide Unit
4. Polypeptides
D. The Architecture of Folded Proteins
1. Conformations of Polypeptide Chains
2. The Extended Chain Structures
Pleated sheets
Twisted sheets
Properties of sheets
Cylinders and barrels
Beta propellers
3. Helices
Properties of helices
Stacking of helices in proteins
Coiled coils
Helix bundles
4. Polyglycine II and Collagen
5. Turns and Bends
6. Domains, Subunits, and Interfaces
7. Packing of Side Chains
8. The Network of Internal Hydrogen Bonds
E. Folding Patterns and Protein Families
1. Complex Folding Patterns
2. Symmetry
3. Effects of Sequence on Folding
F. Chemical Modification and Crosslinking
1. Disulfide Crosslinkages
2. Other Crosslinkages
39
39
40
40
41
42
42
43
43
43
45
46
46
47
47
49
50
50
51
51
55
55
56
59
59
61
62
63
64
65
67
68
69
70
70
71
72
72
74
75
75
76
76
77
78
79
80
80
G. Dynamic Properties of Proteins
1. Motion of Backbone and Side Chains
2. Conformational Changes
3. Denaturation and Refolding
4. Effects of pH and Solvent
5. Irreversible Damage to Proteins
H. Design and Engineering of Proteins
References
Study Questions
81
81
81
82
83
84
85
86
91
Boxes
Box 2-A Proteins of Blood Plasma
Box 2-B Silks
Box 2-C The Nobel Prizes
Tables
Table 2-1 The Sizes of Some Atoms
Table 2-2 Structure and Chemical Properties
of Side Chain Groups of Amino Acids
Table 2-3 Approximate Torsion Angles for Some
Regular Peptide Structures
Table 2-4 Classification of Protein Residues
According to Their Tendencies to
Form Helix, Structure, and
Turns
58
67
83
41
52
61
78
A spiders orb-web is formed by extrusion of a concen-
trated protein solution and stretching of the resulting
fiber. The cross-strands, which are stronger than steel,
resemble silkworm silk. The molecules contain micro-
crystalline sheet domains that are rich in Gly-Ala repeats
as well as polyalanine segments. The capture spiral is
formed from much more elastic molecules that contain
many -turn-forming sequences. These assume a spring-
like spiral. See Box 2-B.
John Gerlach/DRK PHOTO
39
Thousands of different proteins make up a very
large fraction of the machinery of a cell. Protein
molecules catalyze chemical reactions, carry smaller
molecules through membranes, sense the presence of
hormones, and cause muscle fibers to move. Proteins
serve as structural materials within cells and between
cells. Proteins of blood transport oxygen to the tissues,
carry hormones between cells, attack invading bacteria,
and serve in many other ways. No matter what bio-
logical process we consider, we find that a group of
special proteins is required.
The amino acid units that make up a protein mole-
cule are joined together in a precise sequence when the
protein is made on a ribosome. The chain is then folded,
often into a very compact form. Sometimes the chain
is then cut in specific places. Pieces may be discarded
and parts may be added. A metal ion, a coenzyme
derived from a vitamin, or even a single methyl group
may be attached to form the biologically active protein.
The final product is a complex and sophisticated
machine, often with moving parts, that is exquisitely
designed for its particular role.
The biological functioning of a protein is determined
both by the properties of the chemical groups in the
amino acids that are joined to form the protein chain and
by the way the chain is folded. The ways in which the
different parts of the protein interact with each other
and with other molecules are equally important. These
interactions play a major role in determining the folding
pattern and also provide much of the basis for the bio-
logical functioning of proteins. Similar considerations
apply also to carbohydrates, nucleic acids, and other bio-
polymers. For these reasons it is appropriate to review
some fundamentals of molecular structure and geometry.
A. Structural Principles for Small Molecules
Stable organic molecules are held together by
covalent bonds which are usually very strong. The
standard Gibbs energies of formation (G
f
)

of many
covalent single bonds are of the order of 400 kJ/mol
(96 kcal/mol). The bonds have definite directions,
which are measured by bond angles and definite
bond lengths.
1. Bond Angles
Because of the tetrahedral arrangement of the four
bonds around single-bonded carbon atoms and most
phosphorus atoms, all six of the bond angles about the
central atom have nearly the same tetrahedral angle of
109.5.
Bond angles within chains of carbon atoms in organic
compounds vary only slightly from this, and even
atoms that are attached to fewer than four groups
usually have similar angles; for example, the HOH
angle in a water molecule is 105, and the HNH
angles of ammonia are 107. In ethers the COC
angle is 111. However, bond angles of only 101 are
present in H
2
O
2
and of 92 in H
2
S and PH
3
.

See Chapter 6 for a review of thermodynamics


C
109.5
O P
O
O
O
Phosphate ion
3
Amino Acids, Peptides, and Proteins
2
40 Chapter 2. Amino Acids, Peptides, and Proteins
Figure 2-1 Packing of molecules of suberic acid HOOC
(CH
2
)
6
COOH in a crystal lattice as determined by neutron
diffraction.
2
Notice the pairs of hydrogen bonds that join the
carboxyl groups at the ends of the molecules and also the
close contact of hydrogen atoms between the chains. Only
the positions of the hydrogen nuclei were determined; the
van der Waals radii have been drawn around them. How-
ever, the radii were originally determined from X-ray and
neutron diffraction data obtained from many different
crystalline compounds.
The presence of double bonds leads to planarity
and to compounds with bond angles of 120, the inter-
nal angle in a hexagon. The planar geometry imposed
upon an atom by a double bond is often transmitted
to an adjacent nitrogen or oxygen atom as a result of
resonance (Section 6). For example, the amide groups
that form the peptide linkage in proteins (see Fig. 2-5)
are nearly planar and the angles all fall within four
degrees of 120.
2. Bond Lengths
Chemists describe bond lengths as the distances
between the nuclei of bonded atoms. The CC single
bond has a length of 0.154 nm (1.54 ). The CO
bond is 0.01 nm shorter (0.143 nm), and the typical
CH bond has a length of 0.109 nm. The CN bond
distance is halfway between that for CC and CO
(0.149 nm). Other lengths, such as that of OH, can be
estimated from the covalent radii given in Table 2-1.
The length of a double bond between any two
atoms (e.g., C=C) is almost exactly 0.020 nm less than
that for a single bond between the same atoms. If
there is resonance, hence only partial double bond
character, the shortening is less. For example, the
length of the CC bond in benzene is 0.140 nm; the
CO distances in the carboxylate anion are 0.126 nm.
Using simple geometry, it is easy to calculate
overall lengths of molecules; here are two distances
worth remembering:
In the preceding simplified structural formula for
benzene the six hydrogen atoms have been omitted.
Resonance between the two possible arrangements
of the three double bonds
1
is indicated by the circle.
Chemical shorthand of the following type is used
throughout the book. Carbon atoms may be represented
by an angle or the end of a line, but other atoms will
always be shown.
3. Contact Distances
Covalent bond distances and angles tell us how
the atomic nuclei are arranged in space but they do not
tell us anything about the outside surfaces of mole-
cules. The distance from the center of an atom to the
point at which it contacts an adjacent atom in a packed
structure such as a crystal (Fig. 2-1) is known as the
van der Waals radius. The ways in which biological
molecules fit together are determined largely by the
van der Waals contact radii. These, too, are listed in
Table 2-1. In every case they are approximately equal
to the covalent radius plus 0.08 nm. Van der Waals radii

Carboxylate ion
C
O
O

C
O
O

C
C
C
0.254
nm
Distance between
alternate C atoms in fully
extended hydrocarbon chain
Distance across
a benzene ring
0.31 nm
COOH C
O
OH
CONHR C
O
C
C
CH
2
CH
3
H
H
C
CH
3
CH
3
P OH
O
O

N
H
R
H

=
phospho group

H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
O
O
O
O
O
O
Hydrogen Oxygen Carbon
41
explain in words how a right and a left hand differ.
However, since most biochemical compounds are
asymmetric,
3
it is important to be able to visualize
these molecules in three dimensions and to draw their
structures on paper. One of the best ways of learning
to do this is to study molecular models. You may
learn the most by making your own models (see
Appendix).
Whenever four different groups are bonded to a
central carbon atom, the molecule is asymmetric and
the four groups can be arranged in two different
configurations. Consider alanine, one of the alpha
()-amino acids from which proteins are built.
It is called an -amino acid because the amino group
is attached to the (or number 2) carbon atom. To
indicate its three-dimensional structure on a flat piece
of paper, the bonds that project out of the plane of the
paper and up toward the reader are often drawn as
elongated triangles, while bonds that lie behind the
plane of the paper are shown as dashed lines. The
isomer of alanine having the configuration about
the -carbon atom shown in the following structural
formulas is called S-alanine or L-alanine. The isomer
which is a mirror image of S-alanine is R-alanine or
D-alanine. Pairs of R and S compounds (see Section B
for definitions) are known as enantiomorphic forms
or enantiomers.
Notice that in the foregoing drawings, the carboxyl
group ( ), abbreviated COOH, is shown as
havinglost a proton to form the carboxylate ion COO

.
Likewise, the amino group (NH
2
) has gained a proton
to form the NH
3
+
ion. The resulting dipolar ionic or
zwitterionic structure is the one that actually exists
for amino acids both in solution and in crystals.
A. Structural Principles for Small Molecules
are not as constant as covalent radii because atoms can
be squeezed a little, but only enough to decrease
the contact radii by 0.0050.01 nm. The radii of space-
filling molecular models are usually made a little
smaller than the actual scaled van der Waals radii to
permit easier assembly.
4. Asymmetry: Right-Handed and Left-
Handed Molecules
The left hand looks much like the right hand, but
they are different. One is the mirror image of the other.
A practical difference is that your right hand will not
fit into a left-handed glove. Despite our daily acquain-
tance with handedness it may seem difficult to
COO

C H
3
+
N H
CH
3
2()
3()
Alanine
According to the
Fisher projection
formula it is
L-alanine.
H
O
C N
C
H
3
N
+
C
CH
3
H
O
O

C
H
3
N
+
H
COO

H
3
C
C H
3
+
N H
COO

CH
3
C H
3
+
N COO

CH
3
H

S-Alanine (L-Alanine)
C
O
OH
H 0.030 0.12
F 0.064 0.138 0.130
C 0.077 0.16
N 0.070 0.160 0.160
O 0.066 0.154 0.154
Cl 0.099 0.178 0.158
Si 0.117
P 0.110 0.19
S 0.104 0.203 0.160
Br 0.114 0.184 0.154
I 0.133 0.213 0.176
Se 0.215 0.170
0.20
0.170
TABLE 2-1
The Sizes of Some Atoms
ac
a
From Pauling, L. (1960) The Nature of the Chemical Bond, 3rd ed.,
Cornell Univ. Press, Ithaca, New York (pp. 224227 and 260).
b
Covalent radii for two atoms can be summed to give the inter-
atomic distance. The van der Waals radii determine how closely
molecules can pack. The closest observed contacts between atoms
in macromolecules are approximately 0.02 nm less than the sum
of the van der Waals radii. From Sasisekharian, V., Lakshminara-
yanan, A. V., and Ramachandran, G. N. (1967) in Conformation of
Biopolymers, Vol. 2 (Ramachandran, G. N., ed), Academic Press,
New York, (p. 641)
c
Nyburg, S. C and Faerman, C. H. (1985) Acta Crystal. B41, 274 279
Shapes of many atomic surfaces are elliptical. The major radius
a applies to sideways contacts and the minor radius b to polar
contacts along a covalent bond axis. Distances are for atoms singly
bonded to C and may differ slightly if bonds are to other atoms.
d
For distances in multiply by 10.
van der Waals radii (nm)
d
Half-thickness of
aromatic molecules
a
Sideways
contact
c
Element
Covalent
radius
(nm)
a,b,d
b
Polar
contact
c
Radius of
methyl group
42 Chapter 2. Amino Acids, Peptides, and Proteins
The D- and L- families of amino acids. The
amino acids of which proteins are composed are related
to L-alanine but have various side chains (R groups) in
place of the methyl group of alanine. In the preceding
section the structure of L-alanine was given in four
different ways. To recognize them all as the same
structure, we can turn them in space to an orientation
in which the carboxyl group is at the top, the side
chain (CH
3
) is down, and both project behind the
paper. The amino group and hydrogen atom will then
project upward from the paper at the sides as shown
below. According to a convention introduced at the
beginning of this century by Emil Fischer, an amino acid
is L if, when oriented in this manner, the amino group lies to
the left and D if it lies to the right.
Fischer further proposed that the amino acid in this
orientation could be projected onto the paper and
drawn with ordinary lines for all the bonds. This gives
the previously shown Fischer projection formula of
L-alanine.
Although the D and L system of designating configu-
ration is old it is still widely used. Remember that D and
L refer to the absolute configurations about a selected
reference atom in the molecule; for an amino acid this
is the number 2 or -carbon. A quantity that is related
to the asymmetry of molecules is the experimentally
measurable optical rotation (Chapter 23). The sign of
the optical rotation (+ or ) is sometimes given together
with the name of a compound, e.g., D(+)-glucose. The
older designations d (dextro) and l (levo) indicated +
and , respectively. However, compounds with the D
configuration may have either + or optical rotation.
In older literature optical isomerism of the type
represented by D and L pairs was usually discussed in
terms of asymmetric carbon atoms or asymmetric
centers. Now the terms chiral (pronounced ki-ral)
molecules, chiral centers, and chirality (Greek:
handedness) are preferred.
The RS notation for configuration. This nota-
tion, devised by Cahn, Ingold, and Prelog, provides an
unambiguous way of specifying configuration at any
chiral center.
4,5
It is especially useful for classes of
compounds for which no well-established DL system
is available. The groups or atoms surrounding the
central carbon atom, or other central atom, are ranked
according to a priority sequence. The priority of a
group is determined by a number of sequence rules,
the first of which is (1) Higher atomic number precedes
lower. In the following illustration, the priorities of the
groups in D-alanine are indicated by the letters a > b >
c > d. The highest priority (a) is assigned to the NH
2
groups which contain nitrogen bonded to the central
atom. To establish the configuration, the observer
views the molecule down the axis connecting the
central atom to the group having the lowest priority,
i.e., to group d. Viewed in this way, the sequence of
groups a, b, and c can either be that of a right-handed
turn (clockwise) as shown in the drawing or that of a
left-handed turn (counterclockwise).
To establish the priority sequence of groups first
look at the atoms that are bonded directly to the central
atom, arranging them in order of decreasing atomic
number. Then if necessary, move outward to the next
set of atoms, again comparing atomic numbers. In the
case of alanine, groups b and c must be ordered in this
way because they both contain carbon directly bonded
to the central atom. When double bonds are present at
one of the atoms being examined, e.g., the carboxyl
group in alanine, imagine that phantom atoms that
replicate the real ones are present at the ends of the
bonds:
These phantom atoms fill out the valences of the atoms
involved in the multiple bonds and are considered to
have zero atomic number and zero mass. They are not
considered in establishing priorities.
If the first rule and the expansion of multiple
bonds are not sufficient to establish the priority, use
these additional rules: (2) Higher atomic mass precedes
lower. (3) When a double bond is present Z precedes
E (see Geometrical isomers). For ring systems a cis
arrangement of the highest priority substituents pre-
cedes trans. (4) When a pair of chiral centers is present
R,R or S,S precedes R,S or S,R. (5) An R chiral center
precedes S. For further details see Eliel et al.
5
and Bentley.
6
The following groups are ordered in terms of decreas-
ing priority
6
: SH > OR > OH > NHCOCH
3
> NH
2
>
COOR > COOH > CHO > CH
2
OH > C
6
H
5
> CH
3
>
3
H >
2
H > H.
Although the RS system is unambiguous, closely
The view down the axis and toward the group of lowest priority (d),
which lies behind the page. The right-handed turn indicates the
configuration R (rectus = right); the opposite configuration is S
(sinister = left).
C
O
OH
C
O
OH
(O)
(C)
phantom atoms
C H
3
+
N H
COO

R
C H NH
3
+
COO

R
L-Amino acid D-Amino acid
d H
COOH
NH
2
CH
3
b
a
c
observer
R-Alanine (d-Alanine)
c
b
d
a
43 A. Structural Principles for Small Molecules
Configurations of amide or ester linkages may also
be specified in this manner. This is possible because
the CN bond of an amide has partial double-bond
character, as to a lesser extent does the CO bond to
the bridge oxygen in an ester. In this case, assign the
lowest priority to the unshared electron pair on the
ester bridge oxygen.
An amide of the Z configuration is ordinarily referred to
as trans in protein chemistry because the main chain
atoms are trans.
5. Conformations: The Shapes That Molecules
Can Assume
As important to biochemists as configurations, the
stable arrangements of bonded atoms, are conforma-
tions, the various orientations of groups that are caused
by rotation about single bonds.
5,8
In many molecules
such rotation occurs rapidly and freely at ordinary
temperatures. We can think of a CH
3
group as a kind
of erratic windmill, turning in one direction, then
another. However, even the simplest molecules have
preferred conformations, and in more complex structures
rotation is usually very restricted.
Consider a molecule in which groups A and B are
joined by two CH
2
(methylene) groups. If A and B are
pulled as far apart as possible, the molecule is in its
fully extended anti or staggered conformation:
Groups A and B are said to be antiperiplanar (ap) in
this conformation. Not only are A and B as far apart as
possible but also all of the hydrogen atoms are at their
maximum distances one from the other. This can be
seen by viewing the molecule down the axis joining
related compounds that belong to the same configura-
tional family in the DL system may have opposite con-
figurations in the RS system. Thus, L-cysteine (side
chain CH
2
SH) has the R configuration. This is one of
the reasons that the DL system is still used for amino
acids and sugars.
Diastereoisomers. Whereas compounds with
one chiral center exist as an enantiomorphic pair,
molecules with two or more chiral centers also exist as
diastereoisomers (diastereomers). These are pairs of
isomers with an opposite configuration at one or more
of the chiral centers, but which are not complete mirror
images of each other. An example is L-threonine which
has the 2S, 3R configuration. The diastereoisomer with
the 2S, 3S configuration is known as L-allo-threonine.
L-isoleucine, whose side chain is CH(CH
3
) CH
2
CH
3
,
has the 2S, 3R configuration. It can be called 2(S)-
amino-3(R)-methyl-valeric acid but the simpler name
L-isoleucine implies the correct configuration at both
chiral centers.
Sometimes the subscript s or g is added to a D or L
prefix to indicate whether the chirality of a compound
is being related to that of serine, the traditional config-
urational standard for amino acids, or to that of gly-
ceraldehyde. In the latter case the sugar convention
(Chapter 4) is followed. In this convention the config-
urations of the chiral centers furthest from C1 are
compared. Ordinary threonine is L
s
- or D
g
-threonine.
The configuration of dextrorotatory (+)-tartaric acid
can be described as 2R, 3R, or as D
s
, or as L
g
.
Biochemical reactions are usually stereospecific
and a given enzyme will catalyze reactions of mole-
cules of only a single configuration. A related fact is
that proteins ordinarily consist entirely of amino acids
of the L series.
Geometrical isomers. The RS system also gives
an unambiguous designation of geometrical isomers
containing a double bond.
5,7
At each end of the bond,
select the group of highest priority. If these two groups
lie on the same side of the double bond the configuration
is Z (from the German zusammen, together); if on
opposite sides E (entgegen, opposite).
C
a b
C
a' b'
C
a b
C
b' a'
Z E
a and a' higher
priority than
b or b'
C
O C
N
C H
C
O C
N
H C
C
O C
O
C
b'
Z or trans E or cis
b' a' b' a' a'
a b a b a b
COOH
HCOH
HOCH
COOH
2R, 3R-tartaric acid
H H
H H
A
B
C
C
A
H B H
H
H
H
A
B
H
H
H
The antiperiplanar
conformation
View down the axis joining the
C atoms (Newton projection)
44 Chapter 2. Amino Acids, Peptides, and Proteins
the carbon atoms (Newman projection). Rotation of
the second carbon atom 180 around the single bond
yields the eclipsed conformation in which groups A
and B are synperiplanar.
If A and B are large bulky groups they will bump
together, attainment of the eclipsed conformation will
be almost impossible, and rotation will be severely
restricted. Even if A and B are hydrogen atoms (ethane),
there will be a rotational barrier in the eclipsed con-
formation which amounts to 12 kJ (3 kcal) per mole
because of the crowding of the hydrogen atoms as
they pass each other.
5,9
This can be appreciated readily
by examination of space-filling molecular models.
If groups A and B are methyl groups (butane), the
steric hindrance between A and B leads to a rotational
barrier of 25 kJ ( 6 kcal) per mole. The consequence
of this simple fact is that in fatty acids and related
substances and in polyethylene the chains of CH
2
groups tend to assume fully extended zigzag confor-
mations.
In addition to this extended conformation there
are two gauche (skewed or synclinal) conformations
which are only slightly less stable than the staggered
conformation and in which A and B interfere only if
they are very bulky. In one of the two gauche confor-
mations B lies to the right of A and in the other to the
left of A when viewed down the axis.
These two conformations are related to right-handed
and left-handed screws, respectively. The threads on
an ordinary right-handed household screw, when
viewed down the axis from either end, move back-
ward from left to right in the same fashion as do the
groups A and B in the illustration. The angle is the
torsion angle and is positive for right-handed con-
formations. Gauche conformations are important in
many biological molecules; for example, the sugar
alcohol ribitol stacks in crystals in a sickle confor-
mation,
6
in which the chain starts out (at the left) in
the zigzag arrangement but shifts to a gauche con-
formation around the fourth carbon atom, thereby
minimizing steric interference between the OH groups
on the second and fourth carbons.
The complete series of possible conformations is shown
in Fig. 2-2.
A B
H H H
H
One of the two gauche or synclinal conformations
H
A
B
H
H
H

H
OH
1 2
5
3
4
OH
H
H
H
H
HO
OH
H
OH
H
Ribitol in sickle conformation
A B
H H H H
A
H H
(B)
Eclipsed or synperiplanar conformation
Figure 2-2 Description of conformations about a single
bond in the terminology of Klyne and Prelog
10,11
using the
Newman projection. Group A is on the front atom at the top:
the conformation is given for each possible position of group
B on the other atom.
A
B
30
90 90
anticlinal
+ac
anticlinal
ac
antiperiplanar (trans)
ap (t)
a +ap
synclinal
(gauche)
+sc (g
+
)
synclinal
sc (g

)
synperiplanar (sp)
sp +sp
a +ac

sc (g

) +sc (g
+
)
In the chain of methylene units, the hydrogen
atoms on alternate carbon atoms of the fully extended
chain barely touch (Fig. 2-1) but larger atoms cannot
be accommodated. Thus, when fluorine atoms of van
der Waals radius 0.135 nm replace the hydrogen atoms
of radius 0.12 nm, a fully extended chain is no longer
possible. For this reason the torsion angle in polyfluo-
roethylene is changed from the 180 of polyethylene to
166, enough to relieve the congestion but not enough
to cause severe eclipsing of the fluorines on adjacent
carbons. The resulting helical structure is reminis-
cent of those occurring in proteins and other biopoly-
mers. We see that helix formation can be a natural
result of steric hindrance between groups of atoms.
45
Conformations of ring-containing molecules are dealt
with in Chapter 4.
6. Tautomerism and Resonance
Many simple organic compounds exist as mix-
tures of two or more rapidly interconvertible isomers
or tautomeric forms. Tautomers can sometimes be
separated one from the other at low temperatures
where the rate of interconversion is low. The classic
example is the oxo-enol (or keto-enol) equilibrium
(Eq. 2-1).
Although usually less stable than the oxo (keto) form,
the enol is present in a small amount. It is formed
readily from the oxo tautomer by virtue of the fact that
hydrogen atoms attached to carbon atoms that are
immediately adjacent to carbonyl (C=O) groups are
remarkably acidic. Easy dissociation of a proton is a
prerequisite for tautomerism. Since most hydrogen
atoms bound to carbon atoms do not dissociate readily,
tautomerism is unusual unless a carbonyl or other
activating group is present.
Since protons bound to oxygen and nitrogen
atoms usually do dissociate readily, tautomerism also
exists in amides and in ring systems containing O and
N (Eqs. 2-2 to 2-5).
(2-1)
(2-5)
A. Structural Principles for Small Molecules
The tautomerism in Eq. 2-2 is the counterpart of
that in the oxo-enol transformation. However, the
equilibrium constant for aqueous conditions favors
form A very strongly. 2-Pyridone is tautomerized to
2-hydroxypyridine (Eq. 2-3) to a greater extent. Pyri-
midines (Eq. 2-4) and purines can form a variety of
tautomers. The existence of form D of Eq. 2-4 is the
basis for referring to uracil as dihydroxypyrimidine.
However, the di-oxo tautomer A predominates. Pyri-
doxine (vitamin B
6
) exists in water largely as the di-
polar ionic tautomer B (Eq. 2-5) but in methanol as
the uncharged tautomer A. In a pair of tautomers,
a hydrogen atom always moves from one position to
another and the lengths and bond character of these
bonds also change.
The equilibrium constant for a tautomeric inter-
conversion is simply the ratio of the mole fractions
of the two forms; for example, the ratio of enol to oxo
forms of acetone
12
in water at 25C is 6.0 x 10
9
, while
that for isobutyraldehyde is 1.3 x 10
4
. The ratio of
2-hydroxypyridine to 2-pyridone is about 10
3
in water
but increases to 0.6 in a hydrocarbon solvent and to
2.5 in the vapor phase.
13,14
The ratio of dipolar ion to
uncharged pyridoxine (Eq. 2-5) is 4 at 25C in water.
15
The ratios of tautomers B, C, and D to the tautomer A of
uracil (Eq. 2-4) are small, but it is difficult to measure
them quantitatively.
16
These tautomeric ratios are
defined for given overall states of protonation (see
Eq. 6-82). The constants are independent of pH but
will change if the overall state of protonation of the
molecule is changed. They may also be altered by
R C C R'
O
R C C R'
OH
H
H
Oxo form
H
Enol
A B
N
HO
CH
2
OH
CH
2
OH
H
3
C N

O
CH
2
OH
CH
2
OH
H
3
C
Pyridoxine (vitamin B
6
)
H
+
chain
torsion
angle,
180
Polyethylene
torsion
angle,
166
Polyfluoroethylene (Teflon)
H H
H H
F F
F F
(2-2)
(2-3)
(2-4)
R N C R'
O
R N C R'
O H
H
A B
N O N OH
H
2-Pyridone 2-Hydroxypyridine
N
N
H
O
H
N
N
H
OH
O O
N
N
O
HO
N
N
OH
HO
A B
C D
H
46 Chapter 2. Amino Acids, Peptides, and Proteins
changes in temperature or solvent or by binding to a
protein or other molecule.
It is important to distinguish tautomerism from
resonance, a term used to indicate that the properties
of a given molecule cannot be represented by a single
valence structure but can be represented as a hybrid of
two or more structures in which all the nuclei remain
in the same places. Only bonding electrons move to
convert one resonance form into another. Examples
are the enolate anion, which can be thought of as a
hybrid of structures A and B, and the amide linkage,
which can be represented by a similar pair of resonance
forms.
A double-headed arrow is often used to indicate that
two structures drawn are resonance structures rather
than tautomers or other separable isomers.
Although they are distinctly different, tautomer-
ism and resonance are related. Thus, the acidity of
carbon-bound hydrogens in ketones, which allows
formation of enol tautomers, results from the fact that
the enolate anion produced by dissociation of one of
these hydrogens is stabilized by resonance. Similarly,
tautomerism in the imidazole group of the amino acid
histidine is related to resonance in the imidazolium
cation. Because of this resonance, if a proton approaches
structure A of Eq. 2-6 and becomes attached to the left-
hand nitrogen atom (N

), the positive charge in the


resulting intermediate is distributed over both nitrogen
atoms. This makes the proton on N

acidic, permitting
it to dissociate to tautomer B.
Note: The nitrogen atom designated N

(or ND1)
11
in
Eq. 2-6 may also be called N1 or N

(pros-N). Likewise,
N

(NE2) may be designated N3 or N

(tele- N). Since


N

has sometimes also been called N3, it is best not to


use the numerical designations for the nitrogen atoms.
The tautomeric ratio of B to A for histidine in
water (Eq. 2-6) has been estimated, using
15
N- and
13
C-NMR, as 5.0 when the -amino group is proto-
nated and as 2.5 when at high pH it is unprotonated.
17
This tautomerism of the imidazole group is probably
important to the function of many enzymes and other
proteins; for example, if N

of structure A (Eq. 2-6) is


embedded in a protein, a proton approaching from
the outside can induce the tautomerism shown with
the release of a proton in the interior of the protein,
perhaps at the active site of an enzyme. The form
protonated on N

(B of Eq. 2-6), which is the minor


form in solution, predominates in some positions
within proteins.
18
B. Forces between Molecules and between
Chemical Groups
The structure of living cells depends very much
on the covalent bonds within individual molecules
and on covalent crosslinks that sometimes form
between molecules. However, weaker forces acting
between molecules and between different parts of the
same molecule are responsible for many of the most
important properties of biochemical substances. These
are described as van der Waals forces, electrostatic
forces, hydrogen bonds, and hydrophobic interac-
tions. In the discussion that follows the thermody-
namic quantities H, S, and G will be used. If
necessary, please see Chapter 6 for definitions and
a brief review.
1. Van der Waals Forces
All atoms have a weak tendency to stick together,
and because of this even helium liquifies at a low
enough temperature. This is a result of the van der
Waals or London dispersion forces which act strongly
only at a very short distance. These forces arise from
electrostatic attraction between the positively charged
nucleus of one atom and the negatively charged electrons
of the other.
1921
Because nuclei are screened by the
electron clouds surrounding them, the force is weak.
The energy (enthalpy) of binding of one methylene
(CH
2
) unit into a monomolecular layer of a fatty
acid is about H = 1.7 kJ/mol.
22
Although this is
a small quantity, when summed over the 16 or more
carbon atoms of a typical fatty acid the binding energy
is substantial. When a methylene group is completely
surrounded in a crystalline hydrocarbon, its van der
Waals energy, as estimated from the heat of sublimation,
is 8.4 kJ/mol; that of H
2
O in liquid water at the melt-
ing point of ice is 15 kJ/mol.
22
NH N H
+
NH
+
HN N
+
H HN
4

N + H
+
HN
Imidazolium cation

B
A
Imizadole
group
A B
R
H
C C
O
R' R
H
C C
O

R'

Enolate anion
(2-6)
47 B. Forces Between Molecules and Between Chemical Groups
While van der Waals forces between individual
atoms act over very short distances, they can be felt
at surprisingly great distances when exerted by large
molecules or molecular aggregates.
23
Forces between
very smooth surfaces have been measured experimen-
tally at distances as great as 10 nm and even to 300
nm.
23a
However, these long-range van der Waals
forces probably depend upon layers of oriented
water molecules on the plates
23
(see also Section 5).
2. Attraction between Charged Groups
(Salt Linkages)
Fixed positive and negative charges attract each
other strongly. Consider a carboxylate ion in contact
with NH
3
+
or with an ion of calcium:
From the van der Waals radii of Table 2-1 and the ionic
crystal radius of Ca
2+
of 0.10 nm, we can estimate an
approximate distance between the centers of positive
and negative charge of 0.25 nm in both cases. It is of
interest to apply Coulombs law to compute the force
F between two charged particles which are almost in
contact. Let us choose a distance of 0.40 nm (4.0 )
and apply Eq. 2-7.
In this equation r is the distance in meters, q and q
are the charges in coulombs (one electronic charge =
1.6021 x 10
19
coulombs), is the dielectric constant,
and F is the force in newtons (N). The force per mole
is NF where N is Avogadros number.
An uncertainty in this kind of calculation is in the
dielectric constant , which is 1.0 for a vacuum, about
2 for hydrocarbons, and 78.5 for water at 25C. If is
taken as 2, the force for r = 0.40 nm is 4.3 x 10
14
N/mol.
The force would be twice as great for the Ca
2+
COO

case. To move two single charges further apart by just


0.01 nm would require 4.3 kJ/mol, a substantial amount
of energy. However, if the dielectric constant were that
of water, this would be reduced almost 40-fold and the
electrostatic force would not be highly significant in
binding. It is extremely difficult to assign a dielectric
constant for use in the interior of proteins.
23b
For
charges spaced far apart within proteins the effective
dielectric constant is usually as high as 3060.
24
For
closely spaced charges in hydrophobic niches it may be
as low as 24.
2527
A calculation that is often made is the work required
to remove completely two charges from a given distance
apart (e.g., 0.40 nm) to an infinite distance (Eq. 2-8).
If = 2, this amounts to 174 kJ/mol for single charges
at a distance of 0.40 nm; 69 kJ/mol at 1 nm; and only
6.9 kJ/mol at 10 nm, the distance across a cell mem-
brane. We see that very large forces exist between
closely spaced charges.
Electrostatic forces are of great significance in
interactions between molecules and in the induction of
changes in conformations of molecules. For example,
attraction between COO

and NH
3
+
groups occurs
in interactions between proteins. Calcium ions often
interact with carboxylate groups, the doubly charged
Ca
2+
bridging between two carboxylate or other polar
groups. This occurs in carbohydrates such as agarose,
converting solutions of these molecules into rigid gels
(Chapter 4).
Individual macromolecules as well as cell surfaces
usually carry a net negative charge at neutral pH. This
causes the surfaces to repel each other. However, at a
certain distance of separation the van der Waals attrac-
tive forces will balance the electrostatic repulsion.
21
Protruding hydrophobic groups may then interact and
may tether bacteria or other particles at a fixed distance,
often ~5 nm, from a cell surface.
28
3. Hydrogen Bonds
One of the most important weak interactions
between biologically important molecules is the hydro-
gen bond (H-bond).
29,30,30a
These bonds are the result
of electrostatic attraction caused by the uneven distri-
bution of electrons within covalent bonds. For example,
the bonding electron pairs of the HO bonds of water
molecules are attracted more tightly to the oxygen
atoms than to the hydrogen atoms. A small net positive
charge is left on the hydrogen and a small net negative
charge on the oxygen. Such polarization of the water
molecules can be indicated in the following way:
(2-7)
C
O
O
C
O
O

N
H
H
H
+

~0.25 nm ~0.24 nm
Ca
2+
H
H

+
O
F = 8.9875 10
9
newtons
qq'
r
2

W(kJ mol
-1
) = 8.9875 10
6
N
qq'
r
138.9
r (in nm)
=
(2-8)
48 Chapter 2. Amino Acids, Peptides, and Proteins
Here the
+
and

indicate a fraction of a full charge


present on the hydrogen atoms and on the nonbonded
electron pairs of the oxygen atom, respectively. Mole-
cules such as H
2
O, with strongly polarized bonds, are
referred to as polar molecules and functional groups
with such bonds as polar groups. They are to be
contrasted with such nonpolar groups as the CH
3
group in which the electrons in the bonds are nearly
equally shared by carbon and hydrogen.
A hydrogen bond is formed when the positively
charged end of one of the dipoles (polarized bonds)
is attracted to the negative end of another dipole.
Water molecules tend to hydrogen bond strongly one
to another; each oxygen atom can be hydrogen-bonded
to two other molecules and each hydrogen to another
water molecule. Thus, every water molecule can have
up to four hydrogen-bonded neighbors.
Many groups in proteins, carbohydrates, and nucleic
acids form hydrogen bonds to one another and to
surrounding water molecules. For example, an imida-
zole group of a protein can bond to an OH group of
an amino acid side chain or of water in the following
ways:
Remember that hydrogen bonds are always formed
between pairs of groups, with one of them, often C=O
or C=N-, containing the negative end of a dipole and
the other providing the proton. The proton acceptor
group, often OH or NH and occasionally SH, and even
CH in certain structures,
31,31a,31b
donates an unshared
pair of electrons. Dashed arrows are sometimes drawn
from the hydrogen atom to the electron donor atom
to indicate the direction of a hydrogen bond. Do not
confuse these arrows with the curved arrows that
indicate flow of electrons in organic reactions.
The strength of hydrogen bonds, as measured by
the bond energy, varies over the range 1040 kJ/mol.
The stronger the hydrogen bond the shorter its length.
Because hydrogen atoms can usually not be seen in
X-ray structures of macromolecules, the lengths of
hydrogen bonds are often measured between the
surrounding heavy atoms:
A typical OH- - - O hydrogen bond will have a length
of about 0.31 nm; a very strong hydrogen bond may
be less than 0.28 nm in length, while weak hydrogen
bonds will approach 0.36 nm, which is the sum of the
van der Waals contact distances plus the OH bond
length. Beyond this distance a hydrogen bond cannot
be distinguished easily from a van der Waals contact.
Hydrogen bonds are strongest when the hydrogen
atom and the two heavy atoms to which it is bonded
are in a straight line. For this reason hydrogen bonds
tend to be linear. However, the dipoles forming the hydro-
gen bond do not have to be colinear for strong hydrogen
bonding:
32,32a
There is some preference for hydrogen
bonding to occur in the direction of an unshared electron
pair on the oxygen or nitrogen atom.
3335
Both ammonia, NH
3
, and the NH
2
groups of proteins
are good electron donors for hydrogen bond forma-
tion. However, the hydrogen atoms of uncharged NH
2
groups tend to be poor proton donors for H-bonds.
36
Do hydrogen bonds have some covalent character?
The answer is controversial.
36a,36b,36c
Hydrogen bonding is important both to the inter-
nal structure of biological macromolecules and in
interactions between molecules. Hydrogen bonding
often provides the specificity necessary to bring sur-
faces together in a complementary way. Thus, the
location of hydrogen-bond forming groups in surfaces
between molecules is important in ensuring an exact
alignment of the surfaces.
37
The hydrogen bonds do
not always have to be strong. For example, Fersht and
coworkers, who compared a variety of mutants of an
enzyme of known three-dimensional structure, found
that deletion of a side chain that formed a good hydro-
gen bond to the substrate weakened the binding energy
by only 26 kJ/mol. However, loss of a hydrogen
bond to a charged group in the substrate caused a loss
of 1520 kJ/mol of binding energy.
37
Study of mutant
A linear OH- - -O hydrogen bond with
dipoles at an angle one to another.
N N H O
H
N N H
O

+
H

+
or
O H O
length of a
hydrogen bond
O H O
C

+
O
H
H
H O
H
H
H
O H
O
H
O H
H
A water molecule
hydrogen bonded
to four other
water molecules;
note the
tetrahedral
arrangement of
bonds around the
central oxygen.
49
proteins created by genetic engineering is now an
important tool for experimentally investigating the
biological roles of hydrogen bonding.
3739
4. The Structure and Properties of Water
Water is the major constituent of cells and a re-
markable solvent whose chemical and physical prop-
erties affect almost every aspect of life. Many of these
properties are a direct reflection of the fact that most
water molecules are in contact with their neighbors
entirely through hydrogen bonds.
4048
Water is the
only known substance for which this is true.
In ordinary ice all of the water molecules are
connected by hydrogen bonds, six molecules forming
a hexagonal ring resembling that of cyclohexane. The
structure is extended in all directions by the formation
of additional hydrogen bonds to adjacent molecules
(Fig. 2-3). As can be seen in this drawing, the molecules
in ice assume various orientations in the hexagonal
array, and frequently rotate to form their hydrogen
bonds in different ways. This randomness remains as
the temperature is lowered, and ice is one of few sub-
stances with a residual entropy at absolute zero.
49,50
Ice is unusual also in that the molecules do not assume
closest packing in the crystal but form an open struc-
ture. The hole through the middle of the hexagon and
on through the hexagons lying below it is 0.06 nm in
diameter.
The short hydrogen-bond length (averaging 0.276
nm) in ice indicates of strong bonding. The heat of
sublimation (H) of ice is 48.6 kJ/mol. If the van der
Waals dispersion energy of 15 kJ/mol is subtracted
from this, the difference of 34 kJ/mol can be attributed
entirely to the hydrogen bondstwo for each molecule.
Their average energy is 17 kJ/mol apiece. However,
some of the hydrogen bonds are stronger and others
weaker than the average.
51
In a gaseous water dimer the hydrogen bond is linear,
a fact that suggests some covalent character.
52
Its length is distinctly greater than that in ice. This
is one of a number of pieces of evidence suggesting
cooperativity in formation of chains of hydrogen
bonds.
40,53,54
Consider the following three trimers
for which theoretical calculations have predicted the
indicated hydrogen bond energies.
40
In the first case
the central water molecule donates two protons for
hydrogen-bond formation; in the second it accepts the
protons. In the third case it is both an electron acceptor
and a donor. The OH dipoles are oriented head to tail
and the hydrogen bonds are stronger than in the other
cases. Long chains of similarly oriented hydrogen
bonds exist in ice and this may account for the short
hydrogen bond lengths. Closed rings of hydrogen
bonds oriented to give a maximum cooperative effect
also exist in liquid water clusters
55
and within proteins,
carbohydrates, and nucleic acids.
53,54
The nature of liquid water is still incompletely
understood,
46-48,56
but we know that water contains ice-
like clusters of molecules that are continually breaking
up and reforming in what has been called a flickering
cluster structure. Judging by the infrared spectrum
of water, about 10% of the hydrogen bonds are broken
when ice melts.
41
A similar conclusion can be drawn
from the fact that the heat of melting of ice is 5.9 kJ/mol.
It has been estimated that at 0C the average cluster
contains about 500 water molecules.
41
At 50C there
are over 100 and at the boiling point about 40. Although
most molecules in liquid water are present in these
clusters, the hydrogen bonds are rapidly broken and
reformed in new ways, with the average lifetime of a
given hydrogen bond being 10
12
s.
B. Forces Between Molecules and Between Chemical Groups
O H
H
H
O
H
0.298 nm
27 kJ/mol
43 kJ/mol
27 kJ/mol
O
H H
O O
H H
O
H H
O O
H
H
O
H H
H H
O O
H H
H H H H
Figure 2-3 Six water molecules in the lattice of an ice
crystal. The hydrogen bonds, which connect protons with
electron pairs of adjacent molecules, are shown as dashed
lines.
0.10 nm
0.276 nm
50 Chapter 2. Amino Acids, Peptides, and Proteins
(2-9)
5. Hydration of Polar Molecules and Ions
Water molecules are able to hydrogen bond not
only to each other but also to polar groups of dissolved
compounds. Thus, every group that is capable of
forming a hydrogen bond to another organic group
is also able to form hydrogen bonds of a somewhat
similar strength with water. For this reason, hydrogen
bonding is usually not a significant force in holding
small molecules together in aqueous solutions. Polar
molecules that stick together through hydrogen bond-
ing when dissolved in a nonpolar solvent often do not
associate in water. How then can biochemists assert
that hydrogen bonding is so important in biochemistry?
Part of the answer is that proteins and nucleic acids
can be either properly folded with hydrogen bonds
formed internally or denatured with hydrogen bonds
from those same groups to water. The Gibbs energy
change between these two states is small.
Every ion in an aqueous solution is surrounded
by a shell of oriented water molecules held by the
attraction of the water dipoles to the charged ion.
The hydration of ions has a strong influence on all
aspects of electrostatic interactions and plays a domi-
nant role in determining such matters as the strength
of acids and bases, the Gibbs energy of hydrolysis of
ATP, and the strength of bonding of metal ions to
negatively charged groups. For example, the previously
considered interaction between carboxylate and calcium
ions would be much weaker if both ions retained their
hydration shells.
Consider the following example. G for dissocia-
tion of acetic acid in water is +27.2 kJ/mol (Table 6-5).
The enthalpy change H for this process is almost
zero (0.1 kJ/mol) and S is consequently 91.6 J K
1
.
This large entropy decrease reflects the increased
amount of water that is immobilized in the hydration
spheres of the H
+
and acetate

ions formed in the


dissociation reaction. In contrast, dissociation of NH
4
+
to NH
3
and H
+
converts one positive ion to another.
H is large (+52.5 kJ/mol) but the entropy change S
is small (2.0 J K
1
, Table 6-5).
Although effects of hydration are important in
almost all biochemical equilibria, they are difficult to
assess quantitatively. It is hard to know how many
molecules of water are freed or immobilized in a given
reaction. Charged groups in proteins are often hydrated.
However, if they are buried in the interior of the pro-
tein, they may be solvated by polarizable protein side
chain groups such as OH or by backbone or side
chain amide groups.
57,58
6. Hydrophobic Interactions
Fats, hydrocarbons, and other materials whose
molecules consist largely of nonpolar groups have a
low solubility in water and a high solubility in non-
polar solvents. Similarly, the long alkyl groups of fatty
acid esters aggregate within membranes and nonpolar
side chains of proteins are often packed together in the
centers of protein molecules. Because it is as if the
nonpolar groups fear water, this is known as the
hydrophobic effect.
5970
The terms hydrophobic forces,
hydrophobic interactions, and hydrophobic bonding
have also been used. However, the latter term can be
misleading because the hydrophobic effect arises not
out of any special attraction between nonpolar groups
but primarily from the strong internal cohesion of the
hydrogen-bonded water structure.
How strongly do nonpolar groups attract each
other in water? A partial answer can be obtained by
measuring the standard Gibbs energy G of transfer of a
hydrocarbon molecule from a dilute aqueous solution into
a dilute solution in another hydrocarbon. By studying a
series of alkanes, Abraham
62
calculated the Gibbs energy
change per CH
2
unit (Eq. 2-9) as: G
o
= 3.8 kJ/mol.
CH
2
(water) CH
2
(hexane)
G = 3.8 kJ/mol
This equation is a quantitative statement of the
fact that the CH
2
group prefers to be in a nonpolar
environment than to be surrounded by water. A similar
Gibbs energy change would be expected to accompany
the bringing together of a methylene unit from a small
molecule and a hydrophobic surface on a protein mole-
cule. However, in the latter case the accompanying
decrease in entropy would make G less negative.
What causes the decrease in Gibbs energy when
nonpolar groups associate in water? Jencks
60
suggested
that we think of the transfer of a nonpolar molecule
from a nonpolar solvent into water in two steps: (1)
Create a cavity in the water of about the right size to
accommodate the molecule. Since many hydrogen
bonds will be broken, the Gibbs energy of cavity for-
mation will be high. It will be principally an enthalpy
(H) effect. (2) Allow the water molecules in the solvent
to make changes in their orientations to accommodate
the nonpolar molecule that has been placed in the
cavity. The water molecules can move to give good
van der Waals contacts and also reorient themselves to
give the maximum number of hydrogen bonds. Since
hydrogen bonds can be formed in many different
ways in water, there may be as many or even more
hydrogen bonds after the reorientation than before.
This will be true especially at low temperature where
most water exists as large icelike clusters. For dissolved
hydrocarbons, the enthalpy of formation of the new
hydrogen bonds often almost exactly balances the
enthalpy of creation of the cavity initially so that H
for the overall process (transfer from inert solvent into
water) is small. For the opposite transfer (from water
to hydrocarbon hexane; Eq. 2-9) H is usually a small
51 C. Amino Acids and Peptides
positive number for aliphatic hydrocarbons and is
nearly zero for aromatic hydrocarbons.
Since G = H TS (see Chapter 6), it follows
that the negative value of G for hydrophobic inter-
actions must result from a positive entropy change,
which may arise from the restricted mobility of water
molecules that surround dissolved hydrophobic groups.
When two hydrophobic groups come together to form
a hydrophobic bond, water molecules are freed from
the structured region around the hydrophobic surfaces
and the entropy increases. The S for Eq. 2-9 is about
12 J deg
1
mol
1
. Attempts have been made to relate
this value directly to the increased number of orienta-
tions possible for a water molecule when it is freed
from the structured region.
64
However, interpretation
of the hydrophobic effect is complex and controver-
sial.
6571a
The formation constant K
f
for hydrophobic asso-
ciations often increases with increasing temperature.
This is in contrast to the behavior of K
f
for many asso-
ciation reactions that involve polar molecules and for
which H is often strongly negative (heat is released).
An example of the latter is the protonation of ammonia
in an aqueous solution (Eq. 2-10).
NH
3
+ H
+
NH
4
+
H = 52.5 kJ/mol
Since R1nK
f
= G/T = H/T + S, K
f
decreases
with increasing temperature if H is negative. Because
for a hydrophobic interaction with a positive value
of H K
f
increases with increasing temperature, an
increase in stability at higher temperatures is sometimes
used as a criterion for hydrophobic bonding. However,
this criterion does not always hold. For example, base
stacking interactions in polynucleotides (Chapter 5),
whose strength does not increase with increasing
temperature, are still thought to be hydrophobic.
The water molecules that are in immediate contact
with dissolved nonpolar groups are partially oriented.
They form a cagelike structure around each hydropho-
bic group. When particles surrounded by such hydra-
tion layers are 12 nm apart, they sometimes experience
either a fairly strong repulsion or an enhanced attrac-
tion caused by these hydration layers.
21,6466,72
Direct
experimental measurements have shown that these
effects extend to distances of 10 nm
21,63
and can ac-
count for the previously mentioned long-range van
der Waals forces.
Various efforts have been made to develop scales
of hydrophobicity that can be used to predict the
probability of finding a given amino acid side chain
buried within a protein or in a surface facing water.
59,73
A new approach has been provided by the study of
mutant proteins. For example, deletion of a single
CH
2
group from an interior hydrophobic region of
a protein was observed to decrease the stability of the
protein by 4.6 kJ/mol.
74
(2-10)
C. Amino Acids and Peptides
Twenty -amino acids are the monomers from
which proteins are made. These amino acids share
with other biochemical monomers a property essential
to their role in polymer formation: They contain at least
two different chemical groups able to react with each other
to form a covalent linkage. In the amino acids these are
the protonated amino (NH
3
+
) and carboxylate (COO

)
groups. The characteristic linkage in the protein poly-
mer is the peptide (amide) linkage whose formation
can be imagined to occur by the splitting out of water
between the carboxyl of one amino acid and the amino
group of another (Eq. 2-11).
This equation is not intended to imply a mecha-
nism for peptide synthesis. The equilibrium position
for this reaction in an aqueous solution favors the free
amino acids rather than the peptide. Therefore, both
biological and laboratory syntheses of peptides usually
do not involve a simple splitting out of water. Since
the dipeptide of Eq. 2-11 still contains reactive carboxyl
and amino groups, other amino acid units can be joined
by additional peptide linkages to form polypeptides.
These range from short-chain oligomers to polymers
of from 50 to several thousand amino acid units, the
proteins.
7577
1. Properties of -Amino Acids
The amino acids have in common a dipolar ionic
structure and a chiral center. They are differentiated,
one from another, by the structures of their side chain
groups, designated R in the foregoing formulas. These
groups are of varying size and chemical structure.
The side chain groups fill much of the space in the
interior of a protein molecule and also protrude from
the external surfaces of the protein where they deter-
mine many of the chemical and physical properties of
the molecule.
Table 2-2 shows the structures of the side chains
(2-11)
H
3
N CH
R
C
O
O H
+
N
H
CH
R
COO

H
H
3
N CH
R
C
O
N
H
CH
R
COO

+
+
-Amino acids
+
A peptide linkage
in a dipeptide
H
2
O
52 Chapter 2. Amino Acids, Peptides, and Proteins
TABLE 2-2
Structure and Chemical Properies of Side Chain Groups (R) of Amino Acids
Tyrosine, phenylalanine,
and tryptophan can form
hydrophobic bonds and
may be especially effective
in bonding to other flat
molecules.
Strictly a link in the peptide chain, glycine
provides a minimum of steric hindrance to
rotation and to placement of adjacent groups.
a. Glycine "side chain" = H (Gly, G)
a
d. Aromatic amino acids
These bulky groups of distinctive
shapes participate in hydrophobic
interactions in protein interiors
and in forming binding sites of
specific shapes.
b. Amino acids with alkyl groups as side chains
c. The imino acid proline. Because the side chain is fused to the -amino group, the entire structure, not just the side
chain, is shown.
Note the secondary amino group and the relatively rigid conformation.
The presence of proline strongly influences the folding of protein chains.
of the amino acids commonly found in proteins. The
complete structure is given for proline. Both the three-
letter abbreviations and one-letter abbreviations used
in describing sequences of amino acids in proteins are
also given in this table. Amino acids of groups ac of
Table 2-2 plus phenylalanine and methionine are some-
times grouped together as nonpolar. They tend to be
found in a hydrophobic environment on the inside
of a protein molecule. Groups f and i contain polar,
charged side chains which usually protrude into the
water surrounding the protein. The rest are classified
as polar but noncharged.
To get acquainted with amino acid structures,
learn first those of glycine, alanine, serine, aspartic
acid, and glutamic acid. The structures of many
other amino acids can be related to that of alanine
(R=CH
3
) by replacement of a hydrogen by another
group. Metabolic interrelationships will make it easier
to learn structures of the rest of the amino acids later.
Since the COOH groups of glutamic and aspartic
acids are completely dissociated to COO

at neutral
pH, it is customary in the biochemical literature to refer
to these amino acids as glutamate and aspartate
without reference to the nature of the cation or cations
present as counter ions. Such -ate endings are also
used for most other acids (e.g., malate, oxaloacetate,
H
C
H
H
H
H
H H
H
C
H
H
O
H
H
H
H
H
H
H
Tyrosine (Tyr, Y)
N
H
C
H
H
Phenylalanine (Phe, F) Tryptophan (Trp, W)
H Acidic protein,
pK
a
~9.510.9.
H donor in
hydrogen bonds
C C
N
C
C
C HO
O
H
H
H
H
H
H
H
H
Proline (Pro, P)
replaced by OH
in hydroxyproline
(Hyp) which occurs
in collagen
C
H
H
H C
H
C
H
H
C H
H
H
H
C
H
H
C C
H
C
H
H
H
H
H
H
C
H
C
H
H
H
C C
H
H
H
H
H
Alanine (Ala, A)
*
Valine (Val, V)
Leucine (Leu, L)
Isoleucine (Ile, I)
Phenylalanine (Phe, F) Tryptophan (Trp, W) Tyrosine (Tyr, Y)
H
C
H
H
OOC

C
H
3
N
+
H
l-Alanine
Replacement of
one H by phenyl phenylalanine
serine
cysteine
aspartic acid
by OH
by SH
by COOH
R
OOC

C
H
3
N
+
H
53 C. Amino Acids and Peptides
TABLE 2-2
(continued)
The OH group is very weakly acidic (pK
a
~13.6). It can form
esters with phosphoric acid or organic acids and is a site of
attachment of sugar rings in glycoproteins. Hydroxyl groups
of serine are found at the active centers of some enzymes.
The imidazole groups in histidine side
chains are parts of the active sites of
many enzymes. Like other basic
groups in proteins they also may bind
metal ions.
A flexible side arm with a
potentially reactive amino
group at the end. The
high pK
a
of ~10.5 means
that lysine side chains are
ordinarily protonated in
neutral solutions.
The guanidinium group has a high pK
a
of
over 12 and remains protonated under
most circumstances. It is stabilized by
resonance as indicated by the dashed line.
Guanidinium groups are often sites for
binding of phosphate or carboxylate
groups by pairs of hydrogen bonds.
e. Amino acid alcohols
f. Acidic amino acids
g. Amides of aspartic acid and glutamic acid
h. Sulfur-containing amino acids
i. Basic amino acids
C
H
OH
C
CH
3
H
H OH
Serine
(Ser, S)
Threonine
(Thr, T)
Note the second chiral center.
The L-amino acid with the
opposite configuration in the
side chain is L-allo-threonine.
*
C
H
H
C
H
H
C C
HO
C
OH
O
H
H
O
Aspartic acid (Asp, D) Glutamic acid (Glu, E)
C
H
H
C N
H
H
O
C
H
H
C C
N
O
H
H
H
H
Asparagine (Asn, N)
Glutamine (Gln, Q)
The amide group is not acidic but is polar
and participates in hydrogen bonding.
If it is uncertain whether a position in a
protein is occupied by aspartic acid or
asparagine, it may be designated Asx or
B. If glutamic or glutamine, it may be
designated as Glx or Z.
Carboxyl groups of these side chains are dissociated
at neutral pH (pK
a
values are 4.34.7) and provide
anionic () groups on the surfaces of proteins.
C
S
H
H
H
C
C
H
H
S
H
H
C H
H
H
A weak acid,
pK
a
~8.5
Cysteine (Cys, C)
Methionine (Met, M)
H
2
C
S S
CH
2
Two cysteine SH groups can be oxidatively joined
to form a disulfide bridge in the "double-headed"
amino acid cystine.
CH
2
CH
2
CH
2
CH
2
N H
H
H
Lysine (Lys, K)
+

Threonine
(Thr, T)
Methionine (Met, M)
Histidine (His, H)
Lysine (Lys, K)
N
N
C
H
H
H
(also 3 or )
(also 1 or )
H
2
CH
2
CH
2
CH
2
N C
N
N
H
H
H
H
H
+
Guanidinium ion
Hydrogen
bonds
Arginine (Arg, R)
a
The three-letter and one-letter abbreviations used for the
amino acid residues in peptides and proteins are given in
parentheses. B, J, U, X, and Z can be used to indicate
modified or unusual amino acids.
Essential in the human diet
Key
Essential if phenylalanine or cysteine is inadequate
Essential for rapid growth
This basic site accepts a proton to
form a conjugate acid of pK
a
~6.4
7.0 and carrying a positive charge.
54 Chapter 2. Amino Acids, Peptides, and Proteins
Figure 2-4 Structures of some naturally occurring peptides. Oxytocin and
vasopressin are hormones of the neurohypophysis (posterior lobe of the
pituitary gland). Adrenocorticotropin is a hormone of the adenohypophysis
(anterior pituitary). Hormones of the adenohypophysis are released under the
influence of releasing factors (regulatory factors) produced in the neighboring
hypothalamus (a portion of the brain) in response to neural stimulation.
Structures of two releasing factors are shown. Note that the -carboxyl groups
of the N-terminal glutamine residues have reacted, with loss of NH
3
, with the
neighboring terminal NH
2
groups to form cyclic amide (pyroglutamyl) groups.
78
Gramicidin S is an antibiotic made by Bacillus brevis, and surfactin is a depsi-
peptide (containing an ester linkage), a surface active antibiotic of Bacillus
subtilis. Methionine enkephalin is a brain peptide with opiate-like activity.
79
S S
H
N
O N
H
O
O
N
H
2
N O
N
HN
N-terminal Glycine amide, C-terminal
Pyroglutamyl-His-Trp-Ser-Tyr
Human vasopressin
Cys-Tyr-Ile-Gln-Asn-Cys-Pro-Arg-GlyNH
2
Ser-Tyr-Ser-Met-Glu-His-Phe-Arg-Trp-Gly-Lys-Pro-Val--Gly-Lys
Glu-Asp-Glu-Ala-Gly-Asn-Pro-Tyr-Val-Lys-Val-Pro-Arg-Arg-Lys
Ser-Ala-Glu-Ala-Phe-Pro-Leu-Glu-Phe
Pro-Arg-Leu-Gly
GlyNH2
N-terminal Glycine amide, C-terminal
Human oxytocin
Human adrenocorticotropin (ACTH)
1
O
C L-Glu
O C
O
30
15
Hypothalamic thyrotropic hormone releasing
factor (TRF); pyroglutamyl-histidyl-prolinamide
Luteinizing hormone
releasing factor (LRF)
D-Phe L-Leu L-Orn L-Val L-Pro
D-Phe L-Leu L-Orn L-Val L-Pro
Gramicidin S
L-Leu D-Leu L-Val
L-Leu D-Leu
Surfactin
Methionine enkephalin
Tyr-Gly-Gly-Phe-Met
L-Asp
Cys-Tyr-Ile-Gln-Asn-Cys-Pro-Leu-GlyNH
2
S S
phosphate, and adenylate) and in
names of enzymes (e.g., lactate
dehydrogenase).
During the formation of poly-
peptides, the -amino and carboxyl
groups of the amino acids are con-
verted into the relatively unreactive
and uncharged amide (peptide)
groups except at the two chain
termini. In many cases the terminal
amino and carboxyl groups are also
converted within cells into uncharged
groups (Chapter 10). Immediately
after the protein is synthesized its
terminal carboxyl group is often
converted into an amide. The N
terminus may be acetylated or
cyclized to a pyroglutamyl group.
Sometimes a cyclic peptide is formed
(Fig. 2-4).
The properties of polypeptides
and proteins are determined to a
large extent by the chemistry of the
side chain groups, which may be
summarized briefly as follows.
Glycine in a peptide permits a max-
imum of conformational mobility.
The nine relatively nonpolar amino
acidsalanine, valine, leucine, iso-
leucine, proline, methionine, phe-
nylalanine, tyrosine, and trypto-
phanserve as building blocks of
characteristic shape. Tyrosine and
tryptophan also participate in hydro-
gen bonding and in aromatic: aro-
matic interactions within proteins.
Much of the chemistry of pro-
teins involves the side chain func-
tional groups OH, SH, COO

,
NH
3
+
, and imidazole (Eq. 2-6) and
the guanidinium group of arginine.
The side chains of asparagine and
glutamine both contain the amide
group CONH
2
, which is relatively
inert chemically but which can
undergo hydrogen-bonding inter-
actions. The amide linkages of the
polypeptide backbone must also be
regarded as important functional
groups. Most polar groups are
found on the outside surfaces of
proteins where they can react chem-
ically in various ways. When inside
proteins they form H-bonds to the
peptide backbone and to other
polar groups.
55
2. Acidic and Basic Side Chains
The side chains of aspartic and glutamic acids
carry negatively charged carboxylate groups at pH 7
while those of lysine and arginine carry the positively
charged NH
3
+
and guanidinium ions, respectively.
At pH 7 the weakly basic imidazole group of histidine
may be partially protonated. Both the SH group of
cysteine and the phenolic OH of tyrosine are weakly
acidic and will dissociate and thereby acquire negative
charges at a sufficiently high pH.
The number of positive and negative charges on
a protein at any pH can be estimated approximately
from the acid dissociation constants (usually given as
pK
a
values) for the amino acid side chains. These are
given in Table 2-2. However, pK
a
values of buried
groups are often greatly shifted from these, especially
if they associate as ion pairs. In addition, many pro-
teins have free amino and carboxyl-terminal groups
at the opposite ends of the peptide chain. These also
participate in acidbase reactions with approximately
the following pK
a
values.
terminal COOH, pK
a
= 3.63.7
terminal NH
3
+
, pK
a
= 7.57.9
The acidbase properties of an amino acid or of
a protein are described by titration curves of the type
shown in Figs. 3-1 and 3-2. In these curves the number
of equivalents of acid or base that have reacted with an
amino acid or protein that was initially at neutral pH
are plotted against pH. The net negative or positive
electrical charge on the molecule can be read directly
from the curves. Both the net electrical charges and
the distribution of positively and negatively charged
groups are often of crucial importance to the functioning
of a protein.
Additional aspects of the acidbase chemistry of
amino acids and proteins are considered in Chapter 3,
Section A and Chapter 6, Section E. The student may
find it appropriate to study these sections at this time
and to work the associated study problems.
3. The Peptide Unit
The very ability of a protein to exist as a complex
three-dimensional structure depends upon the properties
of the amide linkages between the amino acid units.
Many of these properties follow from the fact that an
amide can be viewed as a resonance hybrid of the
following structures. Because of the partial double-
bond character, the CN bond is shorter than that of a
normal single bond and the C=O bond is lengthened.
The observed lengths in nanometers determined by
X-ray diffraction measurements are given in Fig. 2-5
(top). The partial double-bond character of the CN
bond has important consequences. The peptide unit is
nearly planar as is indicated by the dashed parallelo-
gram in Fig. 2-5.
However, the bonds around the nitrogen retain
some pyramidal character (Fig. 2-5, bottom). Even
more important is the fact that there is flexibility. As
a result, the torsion angle may vary over a range of
15 or even more from that in the planar state.
80,81,81a
The resonance stabilization of the amide linkage is
thought to be about 85 kJ/mol. Rotation around the
CN bond through 90 would be expected to require
C. Amino Acids and Peptides
C
O
N
H
C
O
N
H

+
C
O
N
H

+
C N H
2
H
2
N
HN
Guanidinium group
+
R
1
H
R
2
H
C
O
0
.
1
2
6
N
C
2
0
.1
3
2
0
.
1
0
2
H
O
-Carbon
of second
amino acid
-Carbon
of first
amino acid
H-bond length
0.280.30
116
121
122
0
.1
5
1
Distance (nm) between successive -carbon atoms in a protein.
0.38
C
1
0
.1
4
6
C
1
C
2

This angle, , may


be as great as 15

View down CN axis


(as indicated by heavy arrow above)
C
Figure 2-5 Dimensions of the peptide linkage. Interatomic
distances in nm, including the hydrogen bond length to an
adjacent peptide linkage, are indicated. The atoms enclosed
by the dotted lines all lie approximately in a plane. However,
as indicated in the lower drawing, the nitrogen atom tends
to retain some pyramidal character.
56 Chapter 2. Amino Acids, Peptides, and Proteins
the upper structure would no longer exist. Thus, the
hydrogen bonds would be weakened. We can conclude
that if an amide linkage in such a chain becomes twisted,
the hydrogen bonds that it forms will be weakened. If
there is cooperativity, the hydrogen bonds will all be
strongest when there is good planarity in all of the
amides in the chain.
Amides have very weak basic properties and pro-
tonation is possible either on the oxygen (A) or on the
nitrogen (B).
82,83
The pK
a
values for such protonation are usually
less than zero, but it is possible that a correctly placed
acidic group in a protein could protonate either oxygen
or nitrogen transiently during the action of a protein.
Protonation on oxygen would strengthen hydrogen
bonds from the nitrogen whereas protonation on
nitrogen would weaken hydrogen bonds to oxygen
and might permit rotation. The amide group has a
permanent dipole moment of 3.63 debyes oriented as
follows:
Here the arrow points toward the positive end of the
dipole.
While the trans peptide linkage shown in Fig. 2-4
is usual, the following cis peptide linkage, which is
~8 kJ/mol less stable than the trans linkage, also occurs
in proteins quite often. The nitrogen atom is usually
but not always from proline.
81,84
4. Polypeptides
The chain formed by polymerization of amino
acid molecules provides the primary structure of
a protein. Together with any covalent crosslinkages
and other modifications, this may also be called the
about this much energy. This fact immediately sug-
gests a way in which proteins may sometimes be able
to store energyby having one or more peptide units
twisted out of complete planarity.
69
An important effect of the resonance of the amide
linkage is that the oxygen atom acquires some nega-
tive charge and the NH group some positive charge.
Some of the positive charge is usually depicted as
residing on the nitrogen, but some is found on the
hydrogen atom. The latter can be pictured as arising
from a contribution of a fourth resonance form that
contains no bond to hydrogen.
Nevertheless, this picture is inadequate. Various evi-
dence indicates that the nitrogen actually carries a net
negative charge.
81a
The positive and negative ends of the dipoles in
the amide group tend to associate to form strong
hydrogen bonds. These hydrogen bonds together
with the connecting amide linkages can form chains
that may run for considerable distances through pro-
teins. The tendency for cooperativity in hydrogen
bond formation may impart unusual stability to these
chains. As with individual amide linkages, these chains
of hydrogen-bonded amides can also be thought of as
resonance hybrids:
The two structures pictured are extreme forms, the
true structure being something in between. In the
lower form, rotation about the CN bond would be
permitted but then the charge separation present in
C
O
C
1
N
H
C
2
C
O
N
cis Peptide linkages
C O
N H

+
Fourth
resonance
form
C O
N H O C
N H O C
N H
C O
N H O C
N H O C
N H

+
+
N
3.63D
46.7
C
O
H
O
N

H
HO
NH
O
N
H
H +
+
A B
57
1 10 20 30 40
A P P S V F A E V P Q A Q P V L V F K L I A D F R E D P D P R K V N L G V G A Y
50 60 70 80
R T D D C Q P W V L P V V R K V E Q R I A N N S S L N H E Y L P I L G L A E F R
90 100 110 120
T C A S R L A L G D D S P A L Q E K R V G G V Q S L G G T G A L R I G A E F L A
130 140 150 160
R W Y N G T N N K D T P V Y V S S P T W E N H D G V F T T A G F K D I R S Y R Y
170 180 190 200
W D T E K R G L D L Q G F L S D L E N A P E F S I F V L H A C A H N P T G T D P
210 220 230 240
T P E Q W K Q I A S V M K R R F L F P F F D S A Y Q G F A S G N L E K D A W A I
250 260 270 280
R Y F V S E G F E L F C A Q S F S K N F G L Y N E R V G N L T V V A K E P D S I
290 300 310 320
L R V L S Q M Q K I V R V T W S N P P A Q G A R I V A R T L S P D E L F H E W T
330 340 350 360
G N V K T M A D R I L S M R S E L R A R L E A L K T P G T W N H I T D Q I G M F
370 380 390 400
S F T G L N P K Q V E Y L I N E K H I Y L L P S G R I N M C G L T T K N L D Y V
410
A T S I H E A V T K I Q
C. Amino Acids and Peptides
covalent structure of the protein. Each monomer
unit in the chain is known as an amino acid residue.
This term acknowledges the fact that each amino acid
has lost one molecule of H
2
O during polymerization.
To be more precise, the number of water molecules lost
is one less than the number of residues. Peptides are
named according to the amino acid residues present
and beginning with the one bearing the terminal amino
group. Thus, L-alanyl-L-valyl-L-methionine has the
following structure:
Like amino acids, this tripeptide is a dipolar ion.
The same structure can be abbreviated Ala-Val-Met or,
using one-letter abbreviations, AVM. It is customary
in describing amino acid sequences to place the amino-
terminal (N-terminal) residue at the left end and the
carboxyl-terminal (C-terminal) residue at the right end.
Residues are numbered sequentially with the N-termi-
nal residue as 1. An example is shown in Fig. 2-6.
The sequence of amino acid units in a protein is
always specified by a gene. The sequence determines
how the polypeptide chain folds and how the folded
protein functions. For this reason much effort has
gone into sequencing, the determination of the
precise order of amino acid residues in a protein.
Sequences of several hundreds of thousands of pro-
teins and smaller peptides have been established and
the number doubles each year.
75,87,88,88a
Most of these
Figure 2-6 (A) The complete amino acid sequence of the cytoplasmic enzyme aspartate aminotransferase from pig heart. The
peptide has the composition Lys
19
, His
8
, Arg
26
, (Asp + Asn)
42
, Ser
26
, Thr
26
, (Glu + Gln)
41
, Pro
24
, Gly
28
, Ala
32
, Cys
5
, Val
29
, Met
6
, Ile
19
,
Leu
38
, Tyr
12
, Phe
23
, Trp
9
. The molecular mass is 46.344 kDa and the complete enzyme is a 93.147-kDa dimer containing two
molecules of the bound coenzyme pyridoxal phosphate attached to lysine-258 (enclosed in box).
85,86
(B) A stereoscopic view of a
complete enzyme molecule which contains two identical subunits with the foregoing sequence. Coordinates from Arthur Arnone.
In this wire model all the positions of all of the nearly 7000 atoms that are heavier than hydrogen are shown. The > 8000
hydrogen atoms have been omitted. The view is into the active site of the subunit on the right. The pyridoxal phosphate and the
lysine residue to which it is attached are shown with heavy lines. The active site of the subunit to the left opens to the back side as
viewed here. The drawing may be observed best with a magnifying viewer available from Abrams Instrument Corp., Lansing,
Michigan or Luminos Photo Corp., Yonkers, New York. However, with a little practice, it is possible to obtain a stereoscopic view
unaided. Hold the book with good illumination about 2030 cm from your eyes. Allow your eyes to relax as if viewing a distant
object. Of the four images that are visible, the two in the center can be fused to form the stereoscopic picture. Drawings by
program MolScript (Kraulis, 1991).
A
B
H
3
N
O
H
N
N
O
COO
H H CH
3
H
CH
H
3
C CH
3
SH
3
C
+

H
58 Chapter 2. Amino Acids, Peptides, and Proteins
BOX 2-A PROTEINS OF BLOOD PLASMA
Among the most studied of all proteins are those
present in blood plasma.
af
Their ready availability
and the clinical significance of their study led to the
early development of electrophoretic separations.
Electrophoresis at a pH of 8.6 (in barbital buffer) indi-
cated six main components. The major and one of the
fastest moving proteins is serum albumin. Trailing
behind it are the
1
-,
2
-, and -globulins, fibrinogen,
and -globulins. Each of these bands consists of
several proteins and two-dimensional separation by
electrophoresis and isoelectric focusing (Chapter 3)
reveals over 30 different proteins.
e
Many of these
contain varying numbers of attached carbohydrate
units and appear as families of spots.
Fractionation of large quantities of plasma together
with immunochemical assays has led to identification
of over 200 different proteins. Sixty or more are enzymes,
some in very small quantitites which may have leaked
from body cells. Normally plasma contains 5.7 8.0 g
of total protein per 100 ml (~1 mM). Albumin accounts
for 3.5 4.5 g/100 ml. An individuals liver synthesizes
about 12 g each day. Next most abundant are the
immunoglobulins. One of these (IgG or -globulin)
is present to the extent of 1.21.8 g/100 ml. Also present
in amounts greater than 200 mg per 100 ml are - and
-lipoproteins, the
1
antitrypsin,
2
-macroglobulin,
haptoglobin, transferrin, and fibrinogen.
Plasma proteins have many functions. One of
them, fullfilled principally by serum albumin, is to
impart enough osmotic pressure to plasma to match
that of the cytoplasm of cells. The heart-shaped human
serum albumin consists of a single 65 kDa chain of 585
amino acid residues coiled into 28 helices.
g
Three
homologous repeat units or domains each contain six
disulfide bridges, suggesting that gene duplication
occurred twice during the evolution of serum albumins.
The relatively low molecular mass and high density of
negative charges on the surface make serum albumin
well adapted for the role of maintaining osmotic pres-
sure. However, serum albumin is not essential to life.
Over 50 mutant forms have been found and at least 30
persons have been found with no serum albumin in
their blood.
h,i
These analbuminemic individuals are
healthy and have increased concentrations of other
plasma proteins.
A second major function of plasma proteins is
transport. Serum albumin binds to and carries many
sparingly soluble metabolic products, including fatty
acids, tryptophan, cysteine, steroids, thyroid hormones,
Ca
2+
, Cu
2+
, Zn
2+
, other metal ions, bilirubin, and various
drugs. There are also many more specialized transporter
proteins. Transferrin carries iron and ceruloplasmin
(an
2
globulin) transports copper. Transcortin carries
corticosteroids and progesterone, while another pro-
tein carries sex hormones. Retinol-binding protein
carries vitamin A and cobalamin-binding proteins
vitamin B
12
. Hemopexin carries heme to the liver,
where the iron can be recovered.
j
Haptoglobin binds
hemoglobin released from broken red cells and also
assists in the recycling of the iron in the heme.
k
Lipoproteins (see Table 21-1) carry phospholipids,
neutral lipids, and cholesterol esters. Most of the
mass of these substances is lipid.
Immunoglobulins,
1
-trypsin inhibitor and

2
-macroglobulin,
k
ten or more blood clotting factors;
and proteins of the complement system all have
protective functions that are discussed elsewhere in
this book. Hormones, many of them proteins, are
present in the blood as they are carried to their target
tissues. Many serum proteins have unknown or poorly
understood functions. Among these are the acute phase
proteins, whose concentrations rise in response to
inflammation or other injury.
Human serum albumin. From He and Carter.
g
a
Allison, A. C., ed. (1974) Structure and Function of Plasma
Proteins, Vol. 1, Plenum, New York
b
Allison, A. C., ed. (1976) Structure and Function of Plasma
Proteins, Vol. 2, Plenum, New York
c
Putnam, F. W., ed. (1975) The Plasma Proteins, 2nd ed., Vol. 1
and 2, Academic Press, New York
d
Blomback, B., and Hanson, L. A., eds. (1979) Plasma Proteins,
John Wiley, Chichester, Oklahoma
e
Geisow, M. J., and Gordon, A. H. (1978) Trends Biochem. Sci. 3,
169171
f
Smith, E. L., Hill, R. L., Lehman, I. R., Lefkowitz, R. J.,
Handler, P., and White, A. (1983) in Principles of Biochemistry,
Mammalian Biochemistry, 7th ed., McGraw-Hill, New York,
pp. 337
g
He, X. M., and Carter, D. C. (1992) Nature 358, 209215
h
Peters, T., Jr. (1996) All About Albumin: Biochemistry, Genetics and
Medical Applications, Academic Press, San Diego, California
i
Madison, J., Galliano, M., Watkins, S., Minchiotti, L., Porta, F.,
Rossi, A., and Putnam, F. W. (1994) Proc. Natl. Acad. Sci: USA
91, 64766480
j
Satoh, T., Satoh, H., Iwahara, S.-I., Hrkal, Z., Peyton, D. H.,
and Muller-Eberhard, U. (1994) Proc. Natl. Acad. Sci: U.S.A. 91,
84238427
k
Feldman, S. R., Gorias, S. L., and Pizzo, S. V. (1985) Proc. Natl.
Acad. Sci: U.S.A. 82, 57005704
59
have been deduced from the sequences of nucleotides
in DNA. Sequences of some small peptide hormones
and antibiotics are shown in Fig. 2-4 and that of a 412-
residue protein in Fig. 2-6. The molecular mass of a
protein can be estimated from the chain length by
assuming that each residue adds 100115 Da.
The amino acid composition varies greatly among
proteins. A typical globular protein contains all or
most of the 20 amino acids. The majority are often
present in roughly similar amounts but His, Cys, Met,
Tyr, and Trp tend to be less abundant than the others.
Specialized proteins sometimes have unusual amino
acid compositions. For example, collagen of connec-
tive tissue contains 33 mole% glycine and 21% of
proline + hydroxyproline residues; the major proteins
of saliva contain 22% of glutamate + glutamine and
2045% proline.
89
Cell walls of plants contain both
high proline and high glycine polypeptides. One from
petunias is 67% glycine.
90
Silk fibroin contains 45%
glycine and 29% alanine. A DNA repair protein of
yeast has 13 consecutive aspartate residues.
91
The
tough eggshell (chorion) of the domesticated silkmoth
Bombyx mori contains proteins with 30% cysteine.
92
Many proteins consist, in part, of repeated short se-
quences. For example, the malaria-causing Plasmodium
falciparum in its sporozoite stage is coated with a protein
that contains 37 repeats of the sequence NANP inter-
spersed with 4 repeats of NDVP.
93
These two sequences
have been indicated with single-letter abbreviations
for the amino acids.
With a large number of protein and DNA sequences
available, it has become worthwhile to compare sequences
of the same protein in different species or of different
proteins within the same or different species. Com-
puter programs make it possible to recognize similar-
ities and homologies between sequences even when
deletions and insertions have occurred.
88,88a,9497
The
term homology has the precise biological definition
having a common evolutionary origin, but it is often
used to describe any close similarity in sequence.
98
Among a pair of homologous proteins, a change at a
given point in a sequence may be either conservative,
meaning that a residue of similar character (large,
small, positively charged, nonpolar, etc.) has been
substituted, or it may be nonconservative.
D. The Architecture of Folded Proteins
All proteins are made in the same way but as the
growing peptide chains peel off from the ribosome,
each of the thousands of different proteins in a living
cell folds into its own special tertiary structure.
88a
The number of possible conformations of a protein
chain is enormous. Consider a 300-residue polypeptide
which could stretch in fully extended form for 100
nm. If the chain were folded back on itself about 13
times it could form a 7-nm square sheet about 0.5 nm
thick. The same polypeptide could form a thin helical
rod 45 nm long and 1.1 nm thick. If it had the right
amino acid sequence it could be joined by two other
similar chains to form a collagen-type triple helix of
87 nm length and about 1.5 nm diameter (Fig. 2-7).
The highly folded globular proteins vary consid-
erably in the tightness of packing and the amount of
internal water of hydration.
99,100
However, a density
of 1.4 g cm
3
is typical. With an average mass per
residue of 115 Da our 300-residue polypeptide would
have a mass of 34.5 kDa or 5.74 x 10
20
g and a volume
of 41 nm
3
. This might be approximated by a cube 3.45
nm in width, a brick of dimensions 1.8 x 3.6 x 6.3
nm, or a sphere of diameter 4.3 nm. Although protein
molecules are usually very irregular in shape,
101
for
purposes of calculation idealized ellipsoid and rod
shapes are often assumed (Fig. 2-7).
It is informative to compare these dimensions
with those of the smallest structures visible in cells; for
example, a bacterial flagellum is 13 nm in diameter
and a cell membrane 810 nm in thickness. Bricks
of the size of the 300-residue polypeptide could be
used to assemble a bacterial flagellum or a eukaryotic
microtubule. Helical polypeptides may extend
through cell membranes and project on both sides,
while a globular protein of the same chain length may
be almost completely embedded in the membrane.
1. Conformations of Polypeptide Chains
To understand how a polypeptide chain folds we
need to look carefully at the possible conformations
of the peptide units. Since each peptide unit is nearly
planar, we can think of a polypeptide as a chain of flat
units fastened together as in Fig. 2-8. Every peptide
unit is connected to the next by the -carbon of an
amino acid. This carbon provides two single bonds
to the chain and rotation can occur about both of them
(except in the cyclic amino acid proline). To specify
the conformation of an amino acid unit in a polypeptide
chain, we must describe the torsion angles about both
of these single bonds.
11,76,102
These angles are indicated
by the symbols (phi) and (psi) and are assigned
the value 180 for the fully extended chain as shown in
Fig. 2-8. Each angle is taken as zero for the impossible
conformation in which the two chain ends are in the
eclipsed conformation. By the same token, the torsion
angle (omega) around the CN bond of the amide
is 0 for a planar cis peptide linkage and 180 for the
usual trans linkage.
Since both and can vary for each residue in a
protein, there are a large number of possible confor-
mations. However, many are excluded because they
bring certain atoms into collision. This fact can be
established readily by study of molecular models.
D. The Architecture of Folded Proteins
60 Chapter 2. Amino Acids, Peptides, and Proteins
Using a computer, it is possible to study the whole
range of possible combinations of and . This has
been done for the peptide linkage by Ramachandran.
The results are often presented as plots of vs
(Ramachandran plots or conformational maps)
102a,103,103a
in which possible combinations of the two angles are
indicated by blocked out areas. The original Ramachan-
dran plots were made by representing the atoms as
hard spheres of appropriate van der Waals radii, but
the version shown in Fig. 2-9 was calculated using a
complex potential energy function to represent the
van der Waals attraction and the repulsion from close
contact.
103
This map was calculated for poly-L-alanine
but it would be very similar for most amino acids.
Notice the two areas connected by a higher energy
bridge on the left side of Fig. 2-9. The upper area
contains the pairs of torsion angles for the extended
structures as well as for collagen. The lower area
contains allowed conformations for the right-handed
helices. As can be seen from Fig. 2-10, most of the
observed conformations of peptide units in a real
protein fall into these regions. Glycyl residues are
an exception. Since glycine has no -carbon atom, the
conformations are less restricted. Out of nearly 1900
non-glycine residues in well-determined protein struc-
tures, 66 were found in disallowed areas of the Rama-
chandran diagram.
104a
These were often accommodated
by local distortions in bond angles. The positions at
which such steric strain occurs are often in regions
concerned with function.
104b
One residue, which lies
in a disallowed region in Fig. 2-10 is asparagine 297
of aspartate aminotransferase (Fig. 2-6). It is located
Figure 2-8 Two peptide units in the completely
extended conformation. The torsion angles
i
,

i
, and
i
are defined as 0 when the main chain
atoms assume the cis or eclipsed conformation.
The angles in the completely extended chain are
all 180. The distance from one carbon atom
(C

) to the next in a peptide chain is always 0.38


nm, no matter how the chain is folded.
Figure 2-7 Some idealized shapes that a 34.5 kDa protein molecule
of 300 amino acids might assume.
0 5 10 15 20 nm
Cube, 3.45 nm
Brick,
1.8 3.6 6.3 nm
Prolate spheroid,
6.8 3.4 nm
Oblate spheroid,
5.4 2.7 nm
Sphere,
diameter = 4.3 nm
pleated sheet,
7 7 nm
~0.5 nm thick
Double
helical
rope,
~21 nm
long
Collagen
triple helix,
~29 nm
long
helix, 45 nm long,
1.1 nm diam, ~80 turns
Extended,
~100 nm,
~0.5 nm
width
C
H
N
H
O C
C
O C
H N
H
H
C
R
i
R
i+1
R
i1

i
ith
amino
acid
residue
Repeat
distance
0.72 nm
61 D. The Architecture of Folded Proteins
Figure 2-10 Ramachandran plot for cytosolic aspartate
aminotransferase. The angles and were determined
experimentally from X-ray diffraction data at 0.16 nm resolu-
tion and model building. The majority of conformations are
those of helices or of structure. Glycine residues are
indicated by filled circles, while all other residues are denoted
by an . One of these (green) lies quite far from an allowed
area and must give rise to localized strain.
104b
Extreme lower
limit allowed regions by the hard-sphere criteria are shown
in outline. From coordinates of Arthur Arnone et al. (unpub-
lished).
106
adjacent to the coenzyme site. The possible confor-
mations of proline residues are limited. The angle is
always 60 20, while for the residue adjacent to the
proline N can be either ~150 or ~ 30.
105
Typical ,
angles for some regular peptide structures are given in
Table 2-3.
Conformations of side chain groups are designated
by a series of torsion angles designated
i
.
11
Within
proteins there are strong preferences for certain
i
,
2
pairs.
107
Torsion angles within proline rings are con-
sidered in Chapter 5,A,6.
2. The Extended Chain Structures
As was first pointed out by Pauling and Corey,
108,109
an important structural principle is that within proteins
the maximum possible number of hydrogen bonds
involving the C=O and NH groups of the peptide
chain should be formed. One simple way to do this is
to line up fully extended chains ( = = 180) and to
form hydrogen bonds between them. Such a structure
Figure 2-9 Potential energy distribution in the plane
for a pair of peptide units with alanyl residues calculated
using potential parameters of Scheraga and Flory. Contours
are drawn at intervals of 1 kcal (4.184 kJ) per mol going
down from 0 kcal per mol. The zero contour is dashed.
From Ramachandran et al.
104
The points marked x are for
the four ideal structures: twisted structure (), collagen (C),
right-handed helix (
R
), and the less favored left-handed
helix (
L
).
a
From Libecq, C., ed. (1992) Biochemical Nomenclature, Portland
Press, London and Chapel Hill (for the International Union of
Biochemistry and Molecular Biology).
b
Torsion angles for the fully extended chain can be designated either
+180 or 180, the two being equivalent. They are given as =
180 and = +180 to facilitate comparison with the other struc-
tures.
c
Ramachandran, G. N. (1967) Treatise on Collagen, Vol. 1, p. 124,
Academic Press, New York
d
Both and are quite variable but + = 104.
e
For residues other than these indicated the torsion angles are
about those of typical structures.
TABLE 2-3
Approximate Torsion Angles for Some Regular
Peptide Structures
a
Structure (deg.) (deg.)
Hypothetical fully extended
polyglycine chain
b
180 +180
-Poly(l-alanine) in
antiparallel-chain pleated sheet 139 +135
Parallel-chain pleated sheet 119 +113
Twisted strand 120 +140
Polyglycine II 80 +150
Poly(l-proline) II 78 +149
Collagen
c
60 15 140 15
Right-handed helix
d
57 47
Left-handed helix +57 +47
Bends: Type I, residue 2 60 30
residue 3 90 0
Type II, residue 2 60 120
residue 3 80 0
Type III, residue 2 & 3 60 30
Bulges
e
: Classical bulge residue 1 100 45
G1 bulge residue 1 85 0
180
120
60
0
60
120
180
180 120 60 0 60 120 180

180
120
60
0
60
120
180
180 120 60 0 60 120 180
62 Chapter 2. Amino Acids, Peptides, and Proteins
exists for polyglycine and resembles that in Fig. 2-11.
Notice that on the left side of this figure, the adjacent
chains run in opposite directions; hence, the term
antiparallel structure. The antiparallel arrange-
ment not only gives the best hydrogen bond formation
between chains but also permits a single chain to fold
back on itself giving a compact hairpin loop.
Pleated sheets. While a fully extended polygly-
cine chain is possible, the side chains of other amino
acids cannot be accommodated without some distor-
tion of the structure. Thus, the peptide chains in silk
fibroin have a repeat distance of 0.70 nm compared
with the 0.72 nm for the fully extended chain (Fig. 2-8).
Pauling and Corey
108
showed that this shortening of
the chain could result from rotation of angle by 40
(to 140) and rotation of in the opposite direction
by 45 (to +135) to give a slightly puckered chain.
The resulting multichain structure (shown in Fig. 2-11)
is known as a pleated sheet. As in this figure, both
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
O
R
R
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
R
R
R
R
R
R
O
R
R
R
R
R
R
R
O
O
R
R
H
R
R
R
R
O
R
R
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
O
H
H
H
H
H
H
H
H
H
R
H
R H
R
H
R
H
R
H
R
H
H
R
H
R
O
+

+
+
Figure 2-11 The extended chain pleated sheet structures. (A) Stereoscopic drawing without atomic symbols. (B) Drawing
with atomic symbols. At the left is the antiparallel structure. The 0.70 nm spacing is slightly decreased from the fully extended
length. The amino acid side chains (R) extend alternately above and below the plane of the accordion pleated sheet. The
pairs of linear hydrogen bonds between the chains impart great strength to the structure. The chain can fold back on itself
using a turn perpendicular to the plane of the pleated sheet. The parallel chain structure (right side) is similar but with a
less favorable hydrogen bonding arrangement. Arrows indicate chain directions.
A
B
63 D. The Architecture of Folded Proteins
Figure 2-13 (A) Stereoscopic view of the nucleotide binding domain of glyceraldehyde
phosphate dehydrogenase. The enzyme is from Bacillus stearothermophilus but is homologous
to the enzyme from animal sources. Residues are numbered 0148. In this wire model all of
the main chain C, O, and N atoms are shown but side chains have been omitted. The large
central twisted sheet, with strands roughly perpendicular to the page, is seen clearly;
hydrogen bonds are indicated by dashed lines. Helices are visible on both sides of the sheet.
The coenzyme NAD
+
is bound at the end of the sheet toward the viewer. Note that the two
phosphate groups in the center of the NAD
+
are H-bonded to the N terminus of the helix
beginning with R10. From Skarzynski et al.
111a
(B) Structural formula for NAD
+
.
Figure 2-12 Straight
(left) and twisted (right)
peptide chains in extended
conformations. From
Chothia.
110
A
B
parallel and antiparallel strands are often present in a
single sheet within a protein.
The structure is one of the most important sec-
ondary structures in proteins. It occurs in about 80%
of the soluble globular proteins whose structures have
been determined. In many cases almost the entire
protein is made up of structure. Single strands of
extended polypeptide chain are sometimes present
within globular proteins but more often a chain folds
back on itself to form a hairpin loop. A second fold
may be added to form an antiparallel meander
102
and additional folds to form sheets. Beta structures
are found in silk fibers (Box 2-B) as well as in soluble
proteins.
Twisted sheets. X-ray diffraction studies have
shown that pleated sheets are usually not flat but are
twisted. In a twisted sheet the individual polypeptide
chains make a shallow left-handed helix. However,
when successive carbonyl groups are viewed along the
direction of the chain, a right-handed twist is seen
(Fig. 2-12).
110
Such twisted sheets are often found in
the globular proteins. An example (Fig. 2-13) is the
nucleotide-binding domain of a dehydrogenase
enzyme. The twist of the sheet is seen clearly in this
stereoscopic view. When such chains are associated
into sheets, whether parallel or antiparallel, and are
viewed in a direction perpendicular to the chains and
looking down the edge of the sheet, a left-handed
propeller is seen. Such a propeller is visible in the
O
OH
HO
O
P
O
O
O
P
O
O
O
O N
N NH
2
HO
OH
N
C O
NH
2
+

NAD
+
64 Chapter 2. Amino Acids, Peptides, and Proteins
Figure 2-15 A stereoscopic alpha-carbon
plot showing the three-dimensional structure
of favin, a sugar-binding lectin from the
broad bean (Vicia faba). In this plot only the
-carbon atoms are shown at the vertices.
The planar peptide units are represented as
straight line segments. Side chains are not
shown. The protein consists of two identical
subunits, each composed of a 20-kDa chain
and a 20-kDa chain. The view is down
the twofold rotational axis of the molecule.
In the upper subunit the residues involved
in the front sheet are connected by double
lines, while those in the back sheet are
connected by heavy solid lines. In the lower
subunit the chain is emphasized. Notice
how the back sheet (not the chain) is con-
tinuous between the two subunits. Sites for
bound Mn
2+
(MN), Ca
2+
(CA), and sugar
(CHO) are marked by larger circles. From
Reeke and Becker.
112
drawing of carboxypeptidase A (Fig. 2-14).
The cause of the twist in sheets appears to lie
in noncovalent interactions between hydrogen atoms
on the -carbon atoms of side chains and the peptide
backbone atoms. For side chains of most L- amino
acids these interactions provide a small tendency
Figure 2-14 A ribbon drawing of the 307- residue protein-
hydrolyzing enzyme carboxypeptidase A. In this type of
drawing wide ribbons are used to show strands and helical
turns while narrower ribbons are used for bends and loops
of the peptide chains. The direction from the N terminus to
C terminus is indicated by the arrowheads on the strands.
No individual atoms are shown and side chains are omitted.
Courtesy of Jane Richardson.
117
towards the observed right-handed twist.
111
Nonpla-
narity in the amide groups (Fig. 2-5) may also contrib-
ute. Interstrand interactions seem to be important.
111b
Properties of sheets. In antiparallel sheets,
nonpolar residues are often present on one side of the
sheet and polar residues on the other. The nonpolar
side may be buried in the protein, perhaps backed up
against another sheet of similar structure as in the
carbohydrate-binding lectin shown in Fig 2-15 to give
a sandwich. To accommodate this packing arrange-
ment, nonpolar and polar residues tend to alternate in
the amino acid sequence. The facing nonpolar surfaces
of the two sheets are essentially smooth. The strands
in one sheet lie at an angle of 30 to those in the other
sheet. The twist of the strands allows pairs of nonpolar
side chains from the two sheets to maintain good van
der Waals contact with each other for a considerable
distance along the strands. Beta sheets of silks (Box
2-B) and of immunoglobulin domains (Fig 2-16B) are
also thought to be associated in a back-to-back fashion.
Parallel structures have been found only when there
are five or more strands, some of which may be anti-
parallel to the others. The parallel structure is appar-
ently less stable than the antiparallel structure. Parallel
strands are usually buried in the protein, being sur-
rounded by either other extended strands or helices.
Chains of hydrogen bonds and amide linkages
pass across sheets perpendicular to the chain direc-
tion. There are partial positive and negative charges
along the outside edges of the sheet where these hydro-
gen bond chains terminate (Fig. 2-11). The polarity of
these chains alternates and the positive end of one
peptide bond is relatively near to the negative end of
the next along the edges of the sheets. These unsatis-
fied ends of hydrogen bond chains are often sites of
65 D. The Architecture of Folded Proteins
Figure 2-16 Beta cylinders. (A) Stereo-
scopic -carbon plot of plastocyanin, a
copper-containing electron-transferring
protein of chloroplasts. The copper atom
at the top is also shown coordinated by the
nitrogen atoms of two histidine side chains.
The side chains of the aromatic residues
phenylalanines 19, 29, 35, 41, 70, and 82
and tyrosines 80 and 83 are also shown.
Most of these form an internal cluster.
From Guss and Freeman.
116
(B) Ribbon
drawing of immunoglobulin fold. This
is a common structure in domains of the
immunoglobulins and in many other
extracellular proteins. Two layers of anti-
parallel sheet are stacked face to face to
form a flattened barrel. One disulfide bridge
is always present and is represented as a
thick rod. From J. Richardson.
117
(C) Five
tandem fibronectin type III domains.
These domains, which are found in the
muscle protein titin as well as in
fibronectin, resemble immunoglobulin
domains but lack disulfide bridges. From
Erickson.
117a
Figure courtesy of Harold P.
Erickson.
A
B
60
40
50
90
70
80
30
10
20
60
40
50
70
90
80
30
10
20
C
interaction of polar groups from side chains. Thus,
OH groups of serine or threonine residues, amide
groups of asparagine and glutamine residues, etc.,
often fold back and hydrogen-bond to these ends.
Edges of sheets can also serve as binding sites
for other polar molecules. For example, substrates
bind to an edge of a sheet in the active sites of trypsin
and other proteases (Chapter 12). Some proteins,
e.g. the lectin shown in Fig. 2-15, form dimers by
joining identical edges of a sheet in antiparallel
orientation.
112
Cylinders and barrels. The twisted sheets of
proteins are often curved to form structures known
as cylinders or barrels (Fig. 2-16).
113,114
Simple
cylinders formed by parallel strands form the back-
bones of the electron transport protein plastocyanin,
the enzyme superoxide dismutase, the oxygen carrier
hemerythin (Fig. 16-20), transporter proteins that carry
hydrophobic ligands,
115
and the immunoglobulins in
which each domain contains a barrel.
The eight-stranded cylinder of plastocyanin (Fig.
2-16A) is somewhat flattened and can also be regarded
as a sandwich.
116,118
However, the barrel of triose
phosphate isomerase (see Fig. 2-28) is surrounded by
eight helices which provide additional stability and
a high symmetry. Bacterial outer membranes contain
pores created by very large cylinders within proteins
called porins.
119,120
The one shown in Fig. 8-20 has 16
strands.
A single strand can also be wound into a cylin-
der with the hydrogen bonds running parallel to the
helix axis. A right-handed parallel helix of this
type has been found in the bacterial enzyme pectate
lyase.
121,122
The polypeptide chains of the 353-residue
protein contain seven complete turns of about 22
66 Chapter 2. Amino Acids, Peptides, and Proteins
residues each which form a cylindrical parallel sheet.
The cylinder is folded inward into roughly an L-shape
with packed side chains filling the remaining inside
space. The tailspikes of bacteriophage P22, a virus
of Salmonella, are formed from 666-residue protein
subunits which contain 13 turns of parallel helix
(Fig. 2-17).
123
The tendency for a sheet to fold into a cylinder
is encouraged in antiparallel structures by the exist-
ence of a common irregularity called the bulge.
124,125
As illustrated in Fig. 2-18, a bulge contains an extra
residue inserted into one of the chains. In the second
type of bulge shown in Fig. 2-18, the extra residue is
glycine with torsion angles of about = 85, = 0,
which are possible only for glycine. In the two cylin-
ders of trypsin, chymotrypsin, and elastase (Fig. 12-9),
there are seven bulges.
Beta structures are found in many small peptides.
The hormone oxytocin (Fig. 2-4), the antibiotics grami-
cidin S (Fig. 2-4) and valinomycin (Fig. 8-22), and the
mushroom peptide antamanide (Box 28-B) are among
these. The cyclic structures of these compounds favor
formation of antiparallel strands with sharp turns at
the ends. Polypeptide antibiotics that have alternating
N
C
N
C
Figure 2-17 Wire model of the tailspike protein of
bacteriophage P22 of Salmonella. Three of these fish-
shaped molecules associate as a trimer to form the
spike. From Steinbacher et al.
123
Figure 2-18 Typical bulges in antiparallel pleated sheets. The residues R
1
, R
2
, and R
x
identify the bulges. (A) A classic
bulge, in which
1
and
1
are nearly those of an helix while other torsion angles are approximately those of regular
structures. (B) The G1 bulge in which the first residue is glycine with
1
= 85,
1
= 0. It is attached to a type II turn of
which the glycine (labeled 1) is the third residue.
A B
O C
N R
3
H
C
H
O
N H
C
N
O
H
R
2
H N
C O
N H
R
H
C
O N
H
O
H
R
1
H
H
R
x
H N
C O
H
R
2
N
H
O
H
H
N
H N
C O
C
H
R
x
N
H
H
O
O
N
C
O
H
R
H
turn
1
67 D. The Architecture of Folded Proteins
sequences of D and L residues can be coiled into a
helix
126,127
or a pair of polypeptide chains arranged in
an antiparallel fashion can form a double stranded
helix with an 0.3 to 0.4-nm hydrophilic pore through
the center. These peptide antibiotics appear to exert
their antibacterial action by creating pores through cell
membranes and allowing ions to pass through without
control.
Beta propellers. Another major folding pattern is
a circular array of four to eight blades that form a
propeller-like structure. Each blade is a small, roughly
triangular four-stranded antiparallel sheet (See Figs.
11-7 and 15-23). Sequences that fold into these blades
can often, but not always, be recognized as WD repeats.
These are typically 44- to 60-residue sequences that
have the sequence GH (Gly-His) about 11 24 resi-
dues from the N terminus and WD (Trp-Asp) at the
C terminus.
127a,127b
This repeat sequence encodes the
BOX 2-B SILKS
The green lacewing fly Chrysopa
flava lays its eggs on 1 cm silk stalks
glued to the undersides of leaves.
It has been proposed, as for other
silks, that the peptide chains in the
stalks are aligned perpendicular to
the long dimension of the fiber and
are folded back on themselves many
times to form a sheet with only ~ 8
residues between folds.
a
The chains
of silk fibroin, the major protein of
silkworm silk, contain 50 repeats of
the sequence:
b
GAGAGSGAAG(SGAGAG)
8
Y
All of the alanine and serine side
chains of this sequence presumably
protrude on one side of a sheet while the other
side has only the hydrogen atoms of the glycine.
This permits an efficient stacking of the sheets with
interdigitation of the side chains of the alanine and
serine residues of adjacent sheets.
However, DNA sequences of silk genes have
revealed a greater complexity. The silk fibroin
sequence suggests that at bends between strands
there are often S S bridges and the crystalline sheet
domains are interspersed with 100 200 residue
segments of amorphous protein
b d
whose coiled
chains can be stretched greatly. The protein from
Bombyx mori consists of 350-kDa heavy chains linked
by a disulfide bond to 25-kDa light chains.
e
A silk-
wormhousing silk contains polyalanine sequences,
e.g. (Ala)
10 14
. These evidently form -helical
regions.
f
Silk molecules are synthesized and stored
in glands as a concentrated solution of apparently
globular molecules. The silk is extruded through a
spinneret, whose diameter is ~ 10 m, after which
the silkworm stretches the silk, causing it to form a
stiff fiber. Apparently the stretching of the folded
polypeptide chains permits the -sheet-forming
sequences to find each other and
form oriented crystalline regions.
d
Spiders may produce silk from
as many as seven different types of
glands. Dragline silk, by which a
spider itself may descend, is stron-
ger than steel but because of the
coiled amorphous regions may be
stretched by 35%.
g l
The most elastic
silks are those of the catching spirals
of orb-webs.
i,j,l
They can be stretched
200% and contain a variety of repeat-
ed sequences, including GPCC(X)
n
.
The latter is similar to sequences in
elastin (Section 4), and is able to form
type II bends, which may have
proline in position 2 and must have
glycine in position 3 (Fig. 2-24).
A chain of repeated bend motifs can form a flexible
spring, a spiral as proposed for elastin. Genes for
spider silk have been cloned and are being used to
engineer new proteins with commercial uses, e.g., to
help anchor cells in regenerating body tissues.
d,h,j
a
Geddes, A. J., Parker, K. D., Atkins, E. D. T., and Beighton, E.
(1968) J. Mol. Biol. 32, 343 368
b
Garel, J.-P. (1982) Trends Biochem. Sci. 7, 105 108
c
Vollrath, R. (1992) Sci. Am. 266(Mar), 70 76
d
Calvert, P. (1998) Nature (London) 393, 309 311
e
Mori, K., Tanaka, K., Kikuchi, Y., Waga, M., Waga, S., and
Mizuno, S. (1995) J. Mol. Biol. 251, 217 228
f
van Beek, J. D., Beaulieu, L., Schfer, H., Demura, M., Asakura,
T., and Meier, B. H. (2000) Nature (London) 405, 1077 1079
g
Simmons, A. H., Michal, C. A., and Jelinski, L. W. (1996) Science
271, 84 87
h
Hinman, M. B., and Lewis, R. V. (1992) J. Biol. Chem. 267, 19320
19324
i
Hayashi, C. Y., and Lewis, R. V. (1998) J. Mol. Biol. 275, 773 784
j
Guerette, P. A., Ginzinger, D. G., Weber, B. H. F., and Gosline, J.
M. (1996) Science 272, 112 115
k
Spek, E. J., Wu, H.-C., and Kallenbach, N. R. (1997) J. Am. Chem.
Soc. 119, 5053 5054
l
Hayashi, C. Y., and Lewis, R. V. (2000) Science 287, 1477 1479
Courtesy of Ralph Buchsbaum
68 Chapter 2. Amino Acids, Peptides, and Proteins
fourth strand of one blade followed by the first three
strands of the next blade. This overlap snaps the
blades together to form the propeller. At least one
tight turn is present in each blade. A seven-bladed
propeller is present in the subunits of the regulatory
GTP-hydrolyzing G proteins, which couple extracel-
lular signals to intracellular enzymes and ion channels
(Fig. 11-7). A similar propeller is present in clathrin,
which forms cage-like enclosures around endocytic
vesicles (Chapter 8).
127c
Six-bladed propellers are
predicted to occur in many extracellular proteins.
127d
A propeller binds the coenzyme PQQ in bacterial
dehydrogenases (Fig. 15-23).
3. Helices
The alpha helix represents the second major struc-
tural element of soluble proteins
108,128
and is also found
in many fibrous proteins, including those of muscle
and hair. In the helix both and are about 50 to
60 and except at the N-terminal helix end, each NH is
hydrogen bonded to the fourth C=O further down the chain.
All of the NH and C=O groups of the peptide linkages
lie roughly parallel to the helix axis; the NH groups
point toward the N terminus of the chain and the
carbonyl groups toward the C terminus (Fig. 2-19A).
The number of amino acid units per turn of the
helix is 3.6, with five turns of the helix containing
18 residues. The pitch (repeat distance) of the helix,
which can be determined experimentally from X-ray
diffraction data, is 0.54 nm. Polar coordinates for the
helix have been tabulated.
130
With L-amino acids,
the right-handed helix, is more stable than the left-
handed helix which has so far not been found in pro-
teins. Frequently, however, a few residues have the ,
angles of this helix. The , angles of the helix
are given in Table 2-3 as 57, 47, but are much more
variable in real helices. In erythrocruorin, for which
an accurate structure determination has been made,
131
Figure 2-19 The helix. (A) The right-handed helix with vertical hydrogen bonds indicated by dotted lines. The positions
of the amino acid side chains are indicated by the numbers. (B) The conformation of the peptide backbone of myoglobin.
129
Five long helices are indicated as rods. Several shorter helices can also be seen. The overall size of the molecule is approxi-
mately 4.4 4.4 2.5 nm.
Heme group with
Fe atom on center
Approximate outer
surface when side
chains are included
The pitch of a helix is the length of one complete
turn. The translation per residue is the pitch
divided by the number of residues per turn,
0.54/3.6 = 0.15 nm for the helix.
A
B
69
O
H
H
N
C
O
H
O
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
O
N
19
15
12
11
8
7
5
4
1
2
3
6
10
14
18
17
9
13
16

+
4
1
5
2
6 3
7
E
H
C
Q
N
T
L
L
L
L
I
I
V
T
Q
Q
E
D
E
E
T
S
g
d
a
e
b
f
c
Figure 2-20 (A) The helix showing the three chains
of interconnected amide groups and hydrogen bonds
with partial net positive and negative charges at the
ends. These chains run across the turns of the helical
polypeptide backbone. (B) Scheme illustrating hydro-
gen-bonding pattern for 2
7
ribbon, 3
10
helix, helix,
and helix. (C) The helix represented as a helical
wheel. Imagine viewing the helix from the N-terminal
end of the segment with the lines corresponding to the
backbone of the peptide. Residues, which are desig-
nated by single letters, are spaced 100 apart since there
are 3.6 residues/turn. The peptide shown is a 22-residue
sequence from a leucine-zipper domain of a protein
that participates in gene regulation. From Fathallah-
Shaykh et al.
145a
A B
N
N
H
H
C
O
C
C
C
H
N
C
O
H
H
N
C
H
O
C
C
N
C
C
N
C
O
O
4
3
2
2
7
6
5
1
2
3
7
6
10

9
1

3
10
10
12
15
5
C
the average values are = 64, = 40. More impor-
tant is the observation that + = 104 within about
10 for most residues in helices of this and other
proteins.
132,133
The deviation from the ideal dimen-
sions results in part from the hydrogen bonding of
water molecules or polar protein side chains to the
carbonyl oxygen atoms in the helix.
131
Helix formation is spontaneous for peptides as
short as 13 residues in water.
134,135
Although the
difference in thermodynamic stability between an
helix and an unfolded random coil conformation
is small, poly-L-alanine peptides form helices in water.
Glycine destabilizes helices, presumably
because of the increased entropy of the un-
folded chain which results from the wider
range of the conformational angles and for
glycyl residues. Proline destabilizes helices
even more because its restricted and an-
gles cause the helix to be kinked.
136,137
How-
ever, prolyl residues are often present at ends
of helices. Other amino acids all fit into helices
but may stabilize or destabilize the helix
depending upon immediately neighboring
groups.
138140
Bulky side chain groups with
a low dielectric constant stabilize helices by
strengthening the hydrogen bonds within the
helices.
141
Helices of smaller and larger diameter
than that of the helix are possible. The
most important is the 3
10
helix (or 3.0
10
helix), which has exactly three residues per
turn.
140,142144
Each NH forms a hydrogen
bond to the third C=O on down the chain;
thus, the 3
10
helix is tighter than the helix.
Although long 3
10
helices are seldom found
in proteins, a single turn of this tighter helix
frequently occurs as a defect at the end of
an helix. A polymer of -aminoisobutyric
acid forms long 3
10
helices because the -
dimethyl side chains constrain and to
appropriate values.
145
Short helical peptide
chains in water may exist as a mixture of and 3
10
forms in equilibrium.
146
The helix, with 4.4 residues
per turn is of a larger diameter than the helix and has
only been found in proteins as a single turn, usually at
a C terminus.
132,132a
Properties of helices. The dipoles of the back-
bone amide linkages of an helix are all oriented in
the same direction. The positive end of each dipole is
associated through hydrogen bonding with the nega-
tive end of another. This leaves three partial positive
charges at the N terminus of the helix and three partial
D. The Architecture of Folded Proteins
70 Chapter 2. Amino Acids, Peptides, and Proteins
to account for the fact that the X-ray diffraction pattern
of the keratins of skin and hair indicated a pitch of
0.51 nm rather than the 0.54 nm expected for an helix.
Figure 2-21 (A) Ribbon drawing of the transcription factor
called Max in a complex with DNA. The C termini of the
peptide chains are at the top.
169
Courtesy of S. K. Burley.
(B) Helical wheel representation of residues 231 of the
coiled coil portion of the leucine zipper (residues 249281)
of the related transcription factor GCN4 from yeast. The
view is from the N terminus and the residues in the first two
turns are circled. Heptad positions are labeled ag. Leucine
side chains at positions d interact with residues d and e of
the second subunit which is parallel to the first. However,
several residues were altered to give a coiled coil that mimics
the structure of the well-known heterodimeric oncoproteins
Fos and Jun (see Chapter 11). This dimer is stabilized by ion
pairs which are connected by dashed lines. See John et al.
172
GCN4 Jun GCN4 Fos
E
Q
L
E
Y
A
G
SNK
E
R
H
E
E
E
E
L
L
L
R
V
Q
T
Q
L
L
L
R
V
N
V
V
K
A
R
E
Y
A
G
KNS
E
E
R
H
Q
K
L
M
E
K
D
Q
M
K
D
Q
E
L
d
a
e
b
f
c
g
a
e
b
f
c
g
N
V
V
d
A
B
negative charges at the C terminus and creates a macro-
dipole. It has been estimated that each end of the
helix carries one-half an elementary unit of charge.
147148b
However, this may be an overestimate. The partial
charges are connected by three chains of hydrogen
bonds that run across the turns of the helix as is indi-
cated in Fig. 2-20A. These chains are polarized and
also possess a large hyperpolarizability,
149
i.e., the
polarization of the helix increases more rapidly than
in direct proportion to an applied electrical field.
In a 3
10
helix, there are just two chains of hydro-
gen bonds across the polypeptide chain and running
the length of the helix. The characteristic hydrogen
bonding of the helix and that of the 3
10
helix are
often both present within a helix. Irregularities with
3
10
type hydrogen bonds, arising from interactions
with amino acid side chains or with solvent molecules,
may cause a helix to be kinked or curved.
132
Side
chains of polar residues, including those of Asn, Asp,
Thr, and Ser (and less often Glu, Gln, or His), frequently
fold back and hydrogen bond to the NH and CO groups
that carry the partial charges at the helix ends (Fig.
2-20). The side chain of the third residue in the helix
may also hydrogen bond to the NH of the first resi-
due.
133,150157
The hydrogen bonding of a negatively
charged side chain group to the N-terminal end of the
helix or of a positively charged group to the C-termi-
nal end provides an N-cap or a C-cap which helps
to stabilize the helix by strengthening its hydrogen
bonds.
158
However, the most frequent residue at the
C-terminal end is glycine.
151,155,159
Helices often point
toward active sites of enzymes and interactions of the
helix dipoles with substrates undergoing reaction may
be important to the mechanism of action of these
catalysts.
148,149,160
Stacking of helices in proteins. Many proteins
are made up almost entirely of helices. One of these,
myoglobin, was the first protein for which the com-
plete three-dimensional structure was worked out by
X-ray crystallography.
129
Myoglobin is a small oxygen-
carrying protein of muscle. Its structure is closely related
to that of hemoglobin of blood. Its 153 amino acid
residues are arranged in eight different -helical seg-
ments containing from 7 to 26 residues each. These
rodlike helices are stacked together in an irregular
fashion as shown in Fig. 2-19. Serum albumin (Box 2-A)
has 28 helices organized into three homologous domains.
In contrast, the filamentous bacterial viruses have pro-
tein coats made up of small subunits, each coiled as a
single helix. These are packed in a regular manner
to form the rodlike virus coats (Fig. 7-7).
Coiled coils. In a large family of proteins, two
right-handed helices are coiled around each other in
a left-handed superhelix (Fig. 2-21).
161167
This coiled
coil structure was first suggested by Francis Crick
166
71
Crick suggested that two supercoiled helices inclined
at an angle of 20 to each other would produce the
apparent shortening in the helix pitch and would also
permit nonpolar side chains from one strand to fit into
gaps in the surface of the adjacent strand, a knobs-
into-holes bonding arrangement. Helical strands
tend to coil into ropes because a favorable interstrand
contact can be repeated along the length of the strands
only if the strands coil about each other. A coiled coil
can often be recognized by heptad repeats:
Here, residues a, d, and g (1, 4, and 7) often carry non-
polar side chains. These come together in the coiled
coil as is illustrated in the helical wheel representations
in Fig. 2-21B and provide a longitudinal hydrophobic
strip along the helix.
167
Charged groups are often
present in other locations and in such a way as to
provide electrostatic stabilization through interactions
between residues within a single helix
170- 171a
or
between the pair of helices. The latter type of inter-
action also determines whether the coiled-coil consists
of a parallel or antiparallel pair and whether the two
helices are of identical or of differing amino acid
sequence.
171b
Recent attention has been focused on a DNA-bind-
ing structure called the leucine zipper. A pair of
parallel helices are joined as a coiled coil at one end
but flare out at the other end to bind to DNA. In the
yeast transcription factor GCN4, whose three-dimen-
sional structure has been determined to high resolu-
tion (Fig. 2-21B),
172
the d position of the coiled coil is
occupied by leucine and the e and g positions are often
occupied by charged groups that form stabilizing ion
pairs. Residues at positions b, c, and f are generally on
the outside and exposed to solvent.
168,171,173
The coiled
coil flares out at the C-terminal ends and carries DNA-
binding groups. The structure of a related transcription
factor is shown in Fig. 2-21A.
169
The muscle proteins myosin
174
and tropomyosin
also both consist of pairs of identical chains oriented
in the same direction. The two 284-residue tropo-
myosin chains each contain 40 heptads and are linked
by a single disulfide bridge. X-ray crystallographic
studies
175,176
and electron microscopy
177
show that the
molecule is a rod of 2.0 nm diameter and 41 nm length,
the dimensions expected for the coiled coil. However,
as with other regular protein structures, there are
some irregularities. Myosin chains (Chapter 19) contain
156 heptads.
Another group of proteins have parallel coiled
coils flanked by nonhelical domains in subunits that
associate as filaments. These include the keratins of
skin as well as the intermediate fiber proteins of the
cytoskeleton (Chapter 7).
164,178
Natural coiled coils
often have a parallel orientation, but synthetic pep-
tides have been designed to form antiparallel coiled
coils.
179,179a
Helix bundles. A third peptide chain can be
added to a coiled coil to form a triple-stranded bun-
dle.
180183
An example is the glycoprotein laminin
found in basement membranes. It consists of three
peptide chains which, for ~600 residues at their C-
terminal ends, form a three-stranded coil with heptad
repeats.
182,184
Numerous proteins are folded into four
helical segments that associate as four-helix bundles
(Fig. 2-22).
185188
These include electron carriers, hor-
mones, and structural proteins. The four-helix bundle
not only is a simple packing arrangement, but also
allows interactions between the + and ends of the
macro-dipoles of the helices.
Membranes contain many largely -helical proteins.
Cell surface receptors often appear to have one, two,
or several membrane-spanning helices (see Chapter 8).
The single peptide chain of the bacterial light-operated
ion pump bacteriorhodopsin (Fig. 23-45) folds back
upon itself to form seven helical rods just long enough
to span the bacterial membrane in which it functions.
189
Photosynthetic reaction centers contain an helix
bundle which is formed from two different protein
subunits (Fig. 23-31).
190
A recently discovered ,
barrel contains 12 helices. Six parallel helices form
an inner barrel and 6 helices antiparallel to the first 6
form an outer layer (see Fig. 2-29).
191193
1 2 3 4 5 6 7
a b c d e f g - - - - - - ) (
n
Figure 2-22 Ribbon drawing of an up-and-down four-helix
bundle in myohemerythrin. The two spheres represent the
two iron atoms which carry an O
2
molecule. They are coordi-
nated by histidine and aspartate side chains. Courtesy of
J. Richardson.
117
D. The Architecture of Folded Proteins
72 Chapter 2. Amino Acids, Peptides, and Proteins
4. Polyglycine II and Collagen
In a second form of polyglycine each amino acid
residue is rotated 120 from the preceding one about
a 3-fold screw axis as is shown in the end view of Fig.
2-23A. The angle is about 150 while is about 80
for each residue. The distance along the axis is 0.31
nm/residue and the repeat distance is 0.93 nm. The
molecules can coil into either a right-handed or a left-
handed helix. In this structure, the NH and C=O
groups protrude perpendicular to the axis of the helix
and, as in the structure, can form H-bonds with
adjacent chains.
Poly (L-proline) assumes a similar helical structure.
However, the presence of the bulky side chain groups
induces a left-handed helical twist. Collagen, the
principal protein of connective tissue, basement mem-
branes, and other structures, is the most abundant
protein in the animal body. Its fundamental unit of
structure is a triple helix of overall dimensions 1.4 x 300
nm which resembles the structure of polyglycine II but
contains only three chains.
195
The left-handed helices
of the individual chains are further wound into a
right-handed superhelix (Fig. 2-23B,C). Collagen
contains 33% glycine and 21% (proline + hydroxypro-
lines). The reason for the high glycine content is that
bulky side chain groups cannot fit inside the triple
helix. A hydrogen-bonded cylinder of hydration
surrounds each triple helix and is anchored by hydro-
gen bonds to the OH groups of the hydroxyproline
residues.
196
Sequences of several collagen chains have
been established. One of these contains 1050 residues,
1020 of which consist of 340 triplets of sequence
GlyXY where Y is usually proline and X is often hydroxy-
proline (Hyp).
197,198
The commonest triplet is Gly-Hyp-
Pro.
There are several types of collagen. In one type,
two identical chains of one kind are coiled together
with a third dissimilar chain to form the triple helix.
Several of these triple helices associate to form 8-nm
microfibrils (Fig 2-23D).
199
Once synthesized, collagen
is extensively modified and crosslinked. See Chapter 8.
Collagen-like triple helices also occur within other
proteins. One of these is protein C1q, a component of
the complement system of blood (Chapter 31). This
protein interacts with antibodies to trigger a major
aspect of the immune response. C1q has six subunits,
each made up of three different polypeptide chains of
about 200 residues apiece. Beginning a few residues
from the N termini, there are over 80 residues in each
chain with collagen-like sequences. The three chains
apparently form a triple helix within each subunit.
However, the C-terminal portions are globular in
nature.
200
Collagen-like tails also are present on some
forms of the enzyme acetylcholinesterase (see Chapter
12C,10). The extensins of plant cell walls contain
4-hydroxyproline and evidently have a structure
related to that of collagen.
201,202
Shorter 4- to 8-residue
segments of left-handed polyproline helix are found in
many proteins.
203
5. Turns and Bends
To form a globular protein, a polypeptide chain
must repeatedly fold back on itself. The turns or bends
by which this is accomplished can be regarded as a
third major secondary structural element in proteins.
Turns often have precise structures, a few of which are
illustrated in Fig. 2-24. As components of the loops of
polypeptide chains in active sites, turns have a special
importance for the functioning of enzymes and other
proteins. In addition, tight turns are often sites for
modification of proteins after their initial synthesis
(Section F).
The turn (Fig. 2-24) is often found in hairpin or
reverse turns at the edges of sheets (Fig. 2-11) and at
other locations.
204212
If all four residues that contribute
to bends are counted, they constitute about one-third
of the amino acid residues in most proteins.
124
In many
turns, the C=O of the first residue hydrogen bonds
to the NH of the fourth residue. This hydrogen bond
may be part of the hydrogen bond network of a
pleated sheet. The peptide unit between -carbon
atoms 2 and 3 of the turn is perpendicular to the sheet.
There are two possibilities for the orientation of this
peptide unit. In a type I turn, the C=O is down when
the turn is viewed as in Fig. 2-24, while the side chains
of residues 2 and 3 point upwards or outward on the
opposite side of the bend. In a type II turn, the C=O is
up and the NH down. Residue 3 is always glycine in a
type II turn because the side chain would collide with
the C=O group if any other amino acid were present.
As is seen in Fig. 2-24, a trans-proline can fit at position
2 in a type II turn
206
as well as in type I turns. A cis-
proline residue can fit at position 2 or position 3 in a
type I turn.
137,213
The type III turn is similar to a type I turn but has
the , angles of a 3
10
helix and the two chains emerg-
ing from the turn are not as nearly parallel as they are
in type I turns. Beta turns of the less common types I,
II, and III have a left-handed twist. As can be seen
in Fig. 2-24, this permits a better match to the twist of
strands in a sheet. Unless glycine is present, these
bends are less stable because of steric hindrance.
214,215
Polar side chain groups such as those of aspartate or
asparagine often form hydrogen bonds to the central
peptide units of turns.
214
The tight turn
215
and the proline-containing
turn shown in Fig. 2-24 are thought to be major com-
ponents of the secondary structure of elastin.
216218
This stretchable polymer, which consists largely of
nonpolar amino acids, is the most abundant protein
of the elastic fibers of skin, lungs, and arteries. The
73
0 0.5 nm
H
H
H
H
H
H
H
H
N
N
N
C C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C
C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C
C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C C
C
C
C
C
O
O
O
H
H
H
H
H
H
H
H
H
N
N
N
C
C
C
C
C
C
O
O
O
H
X
Y
Figure 2-23 (A) The structure of polyglycine if
viewed down the axis of a helix chain. Note the
threefold symmetry within the structure. Polyproline
forms a similar structure and collagen is thought to be
a triple-stranded rope of three peptide chains having
essentially the same structure but in addition contain-
ing a right-handed supercoil. This is illustrated by the crystal structure of a collagen-like peptide shown in B and C. (B) View
similar to that in (A), but showing how three chains form the triple-stranded ropes separated by a cylinder of hydration. The
structure is shown as a wire model. The xs are water molecules. They form an extensive network of H-bonds to one another
and to peptide groups. From Bella et al.
194
C) Electron micrograph of collagen fibrils, each of which consists of many triple-
helical units. Deposited from suspension and shadow cast with chromium. Courtesy of Jerome Gross, M.D. (D) Stereoscopic
view of a collagen-like model peptide. Each of the three parallel 30-residue chains contains a (Pro-Hyp-Gly)
3
cap at each
end and the 12-residue sequence Ile-Thr-Gly-Ala-Arg-Gly-Leu-Ala-Gly-Pro-Hyp-Gly in the center. This sequence is found in
human type III collagen and includes a site (green asterisk) of known mutations (see Chapter 8). From Kramer et al.
194a
Courtesy of Helen Berman.
A B
C
D. The Architecture of Folded Proteins
* *
D
500 nm
74 Chapter 2. Amino Acids, Peptides, and Proteins
6. Domains, Subunits, and Interfaces
Many proteins are organized into tightly folded
globular domains consisting of 50150 amino acid
residues.
117,222227
Smaller proteins may have 2 or 3
domains but large proteins may have more. For exam-
ple, the immunoglobulin heavy chains (Chapter 31)
have four or five domains similar to that shown in
Fig. 2-16B. The enormous 3000-kDa muscle protein
titin contains 260 domains, many of which are of the
immunoglobulin typeFig. 2-16,C.
228,117a
In most pro-
teins the polypeptide chain folds to form one domain,
then passes through a hinge to the next. In others,
the C terminus or the N terminus of the polypeptide
folds back across two or more domains as a kind of
strap that helps to hold the protein together. Even
when a protein contains only one domain, it often
consists of two distinct lobes with a cleft between
them. Many proteins, e.g. hemoglobin (Fig. 7-25),
consist of subunits about the size of the globular
domains in larger proteins.
Structural domains of proteins are sometimes
encoded by a single coding segment of DNA i.e., by a
single exon in a split gene. Domains of this type may
have served as evolutionarily mobile modules that
have spread to new proteins and multiplied during
evolution. For example, the immunoglobulin structural
domain is found not only in antibodies but also in a
variety of cell surface proteins.
229232
Whether we deal with domains connected by a
flexible hinge or with subunits, there are interfaces
between the different parts of the protein. These inter-
faces are often formed largely of nonpolar groups.
However, they frequently contain a small number of
hydrogen bonds that bridge between one domain and
another or between one subunit and another. In the
case of hemoglobin, important changes occur in this
hydrogen bonding and movement occurs along one
of the interfaces between two subunits. Likewise the
active sites of enzymes are often located at interfaces
between domains. During catalysis, movement and
reorganization of the hydrogen bonds and side chain
packing in the interfaces may take place.
232a
70-kDa chains are thought to have 70- to 75-residue
regions in which the polypeptide folds back on itself
repeatedly with a large number of bends in a broad
left-handed spiral.
216,218a
The consensus sequence
VPGVP, which tends to form a type II turn with
proline in position 2 (Fig. 2-24) is present in long tan-
dem repeats e.g., (VPGVG)
11
. These extensible regions
alternate with short helices which are crosslinked
to other chains. Similar structures are present in silks
(Box 2-B) and in proteins of wheat gluten.
217a,218b
Besides hairpin turns and broader U-turns, a protein
chain may turn out and fold back to reenter a sheet
from the opposite side. Such crossover connections,
which are necessarily quite long, often contain helices.
Like turns, crossover connections have a handedness
and are nearly always right-handed (Fig. 2-25).
117,219
Most proteins also contain poorly organized loops on
their surfaces. Despite their random appearance, these
loops may be critical for the functioning of a protein.
220
In spite of the complexity of the folding patterns,
peptide chains are rarely found to be knotted.
221
Left Right
Figure 2-25 Right- and left-handed crossover connections
in proteins. These connections are nearly always right-
handed. The broad arrows represent strands. The
crossover often contains a helix. Units of two adjacent
strands ( units) with an helix between are found
frequently in globular proteins.
Figure 2-24 Tight turns found in polypeptide chains. Two
types of turn are shown. A third variant, the type III or 3
10
turn resembles the type I turn but has the , angles of a 3
10
helix. Type II turns containing proline and tighter turns
are thought to be major structural components of elastin.
Another turn, lacking the hydrogen bond has a cis-proline
residue at position 3.
C
N
C
C
C
H R
2
N
O
C
C
H
R
3
H
H
R
1
O H N
C
H
R
4
O
Turn: Type I
H
C
N
C
C
C
R
2
N C
C
H
X
H
H
R
1
O H N
C
H
R
4
O
Turn: Type II
H
O
H
Turn: Type II
with proline Turn
C
N
C
C
C
N C
C
H
H
H
H
R
1
O H N
C
H
R
4
O
O
H
2
3
N H O C
C C H
H
N C
N C
C
O H
O
CH
CH
3
CH
3
H
2
3 1
H
H
75
7. Packing of Side Chains
In Fig. 2-19B myoglobin is pictured as a cluster of
-helical rods surrounding the heme core. This picture
is incomplete because the space between the rods and
inside the molecule is tightly packed with amino acid
side chains almost all of which are hydrophobic. The
same is true for the barrels of Fig. 2-16, which are
filled largely with nonpolar side chains. As the struc-
tures of more and more proteins have been determined,
a consistent pattern has emerged. Within the interior
of proteins the side chain groups are packed together
remarkably well.
99,100,222,233236
Although occasional
holes are present, they are often filled by water mole-
cules.
99,237a
The packing density, the volume enclosed
by the van der Waals envelope divided by the total
volume, is 0.75 for the interior of the lysozyme and
ribonuclease molecules compared with the theoretical
value of 0.74 for close-packed spheres. However,
regions with many hydrogen bonds may be less tightly
packed.
238
The interiors of proteins often contain large numbers
of aromatic side chains which are frequently associated
as pairs or as aromatic clusters.
239,240
For example,
see the structure of plastocyanin in Fig. 2-16A. Rings
may lie perpendicular one to another or be stacked
face to face but offset. Oxygen atoms often lie in con-
tact with the edges of aromatic rings.
241
The planar
guanidinium groups of arginine side chains often
stack against aromatic rings and amide groups may
sometimes do the same.
242
It has been suggested that
both aromatic: aromatic and aromatic:oxygen interac-
tions may be associated with additional stabilization
of the protein by 4- to 8- kJ/mol. Tyrosine side chain
OH groups often stabilize ends of strands by form-
ing H-bonds to backbone atoms.
243
Most polar groups are on the surfaces of proteins,
and those that are not are almost always hydrogen
bonded to other groups in the interior.
244
While most
nonpolar groups are inside proteins, they are also
present in the outer surfaces where they are often
clustered into hydrophobic regions or patches.
The latter may be sites of interaction with other pro-
teins or with lipid portions of membranes.
8. The Network of Internal Hydrogen Bonds
The fact that nonpolar groups tend to be buried
in the interior of proteins suggests that the inside of
a protein might be a flexible blob of oily material. In
fact, the nonpolar groups tend to be densely packed
and aromatic rings often impart considerable rigidity
to the hydrophobic cores. Buried polar groups, which
are invariably hydrogen bonded to other side chain
groups, to amide groups of the polypeptide back-
bone,
244247
or to buried water molecules, form a well-
defined internal network. When a series of closely
related proteins, for example, those having the same
function in several different species, are compared, the
hydrogen bonded network is often nearly invariant.
This suggests a functional significance. The hydrogen
bonding possibilities for some of the side chain groups
in proteins are indicated in Fig. 2-26.
Charged side chains are sometimes buried in the
interior of proteins, usually together with an ion of
the opposite charge to form a hydrogen-bonded ion
pair.
248,248a,248b
However, there is sometimes a single
charge that is neutralized by the interaction with
dipoles of polar groups.
217
Sometimes an undissociated
carboxyl group is hydrogen bonded to a carboxylate
ion.
249
Figure 2-26 Some of the possibilities for hydrogen bonding
of side chain groups in proteins. Oxygen atoms can and
frequently do form up to three hydrogen bonds at once.
Open arrows point from H-atoms and toward electron donor
pairs.
His
N
N
H
N
N
H
O

Tyr
H
O
N
H
Trp Arg
H H
N
N
H
H
+
N
H
C
O
N H
Asn
Gln
H
Lys N H
H
N
H
H
H
+
Asp
Glu
C
O
O
C
O
O
H

Ser
Thr
Tyr
O
H
C
O
O
H

O
C
O
D. The Architecture of Folded Proteins
76 Chapter 2. Amino Acids, Peptides, and Proteins
A buried carboxyl group of this type will display a pK
a
value far higher than the normal value.
Arginine side chains are large and able to form
multiple hydrogen bonds
247,250
as well as salt link-
ages
251
to different parts of a folded peptide. Cations
of arginine and lysine are never buried unless in ion
pairs. Protons from strong acids have long been
known to bind to centers of benzene rings and water
can form weak hydrogen bonds to the centers of aro-
matic rings.
252,253,253a
Such bonds also occur within
proteins and often involve the binding of guanidinium
groups or of inorganic cations to indole rings of tryp-
tophans.
253b,253c
Protonated imidazole groups may
also bind to aromatic rings.
253d
Another important aspect of the structure of pro-
teins is the presence of hydrogen-bonded water
molecules in pockets and cracks. These molecules,
as well as a much larger number of water molecules
bound at the outer surface, are clearly visible from
X-ray studies. They often occur singly, bonded to the
ends of amide groups, especially to the carbonyl ends.
Internal bends of the peptide chain are almost always
hydrated.
254
These water molecules often make two or
more hydrogen bonds to different parts of the protein
or to other water molecules. Clusters of water mole-
cules,
49,255,255a
sometimes in the form of pentagonal
rings,
256
are often present. NMR spectroscopy has
shown that water molecules bound to protein surfaces
exchange rapidly with the bulk water in which the
protein is dissolved; the residence time on the
protein is typically less than a nanosecond. Interior
water molecules have much longer residence times
of 10
2
to 10
8
s for a small protein.
257
They may be
regarded as part of the protein structure.
E. Folding Patterns and Protein Families
Proteins are folded in many ways. We have already
considered several simple patterns: the antiparallel
cylinder (Fig. 2-16), the 2-helix coiled coil (Fig. 2-21) and
the 3- and 4-helix bundles (Fig. 2-22). Another simple
motif that has been found repeatedly is the helixturn
helix or helixloophelix in which two helices at
variable angles, one to another and with a turn or short
loop between them, form a structural unit. DNA-
binding repressors and transcription factors (see Fig.
2-21 and also Chapter 5) often contain this motif as do
many Ca
2+
-binding proteins. Proteins containing 36
helical segments, often fold into a roughly polyhedral
shape.
258,259
An example is myoglobin (Fig. 2-19B).
1. Complex Folding Patterns
Proteins often contain elements of both and
structure. One of the first of the complex folding
motifs to be recognized was a nucleotide-binding
domain identified by Rossmann and associates.
260262
This Rossman fold contains six parallel strands
which alternate with six helices. The result of the
parallel structure is that the helices are also parallel
and that their amino-terminal ends, which carry
partial positive charges, are aligned in approximately
the same directions. The positive end of one of the
helices lies behind the negatively charged phosphate
groups
148
which characteristically bind at the edge of
the sheet containing the C termini of the strands.
This can be seen in Fig. 2-13, which shows binding of
the coenzyme NAD to the nucleotide binding domain
of glyceraldehyde phosphate dehydrogenase.
Similar nucleotide-binding domains are found in many
other dehydrogenases whose members constitute a
protein family.
Figure 2-27 depicts topology diagrams for the
Rossman fold and for two related families of proteins.
These families bind the nucleotides called GTP and
ATP, respectively. Both are structural relatives of
NAD. A major part of the structure of all of the pro-
teins in these families consist of units, each one
containing a strand followed by a helix. They are
Figure 2-27 Topologies of the folds of three families of
nucleotide binding / proteins. Cylinders represent
helices and arrows strands. (A) The ATPase fold for the
clathrin-uncoating ATPase; (B) The G-protein fold that binds
GTP and is found in ras proteins; (C) The Rossmann fold
that binds NAD in several dehydrogenases. From Brndn.
262
1 2 3 4 5 6
N
C
1 3 4 5
N
C
2
1 2 4 5 6 3
N
C
A
B
C
H
2
O benzene
hydrogen bond
H O
H
77
H4
H6
H7
H8
H10
H13
H2
H1
H11
H12
H9
H5
H3
classified as / proteins. The Rossman fold is com-
posed of six units. Recently a ribonuclease inhibi-
tor protein with 15 consecutive units has been
characterized.
263
Each unit contains several
residues of leucine. This leucine-rich repeat occurs
in many other proteins as well.
264,264a,264b
The / barrel shown in Fig. 2-28 consists of 8
consecutive units in a symmetric array.
265,266
By
1995 over 40 of these barrels had been identified in a
diverse group of enzymes. One bifunctional enzyme
contains two / barrels. Although the nature of the
reaction catalyzed varies, the active site is always
found in the center of the barrel at the C-terminal ends
of the 8 parallel strands and therefore between the N
termini of the surrounding helices. The enzyme se-
quences show no homology and frequent occurrence
of the 8-stranded barrel may reflect the fact that it is a
natural packing arrangement of units. However,
a 10-stranded barrel of this type has also been
found.
267
Figure 2-29 Structure of the , barrel of a fungal enzyme glucoamylase. (A) side view (stereoscopic); (B) top view. The
active site, which cleaves glucose units from the ends of starch chains, is in the depression in the center of the barrel. Here it is
occupied by an inhibitor. See Aleshin et al.
192
Courtesy of Alexander Aleshin.
Figure 2-28 The eight-fold / barrel structure of triose phosphate isomerase. From Richardson. (A) Stereoscopic view.
(B) Ribbon drawing. Courtesy of Jane Richardson.
117
A B
This barrel can be compared with that of the 12-
helix , barrel of a fungal glucoamylase whose struc-
ture is shown in Fig. 2-29. Numerous more complex
folding patterns have been discovered. They have
been classified by Jane Richardson.
117,122
Many of the
proteins described by these folding patterns can be
grouped into families and superfamilies.
227
Chothia
suggested that there may be about 1,000 families in
nature;
268
over 700, with over 360 distinct folds have
been identified.
268a
2. Symmetry
A sometimes puzzling feature of protein structure
is the widespread occurrence of an approximate two-
fold axis of symmetry. This often arises as a natural
result of association of a pair of irregular subunits
(Chapter 7). The association is such that rotation
A B
E. Folding Patterns and Protein Families
78 Chapter 2. Amino Acids, Peptides, and Proteins
about the twofold axis will cause the two subunits to
exchange positions and to remain in an identical
chemical environment. Approximate symmetry is
often observed also within single peptide chains. For
example, in the Rossman fold (Figs. 2-13, 2-27), an
approximate twofold axis passes between the center
strands of the sheet residues and relates the two flank-
ing helices, which begin with residues R10 and T100,
respectively. The bound NAD
+
also possesses an approx-
imate twofold axis, but it is not quite symmetrically
placed at the end of the sheet. Both phospho groups
are seen to interact with the N terminus of the helix
beginning at residue 10. The small bacterial protein
ferredoxin (Fig. 16-16B) contains two iron-sulfur clus-
ters related by an approximate 2-fold axis. The two
cylinders of elastase (Fig. 12-9) as well as the two sides
of the flattened barrel of copperzinc superoxide
dismutase
269
are approximately related by twofold axes.
The enzyme thiosulfate: cyanide sulfurtransferase (Eq.
24-46) is remarkably symmetric but the active site is
located in just one half. The widespread existence of
this approximate symmetry suggests a biological
significance that remains to be discovered.
3. Effects of Sequence on Folding
Studies of synthetic polypeptides as well as exami-
nation of known protein structures reveal that some
amino acids, e.g., Glu, Ala, Leu, tend to promote
helix formation. Others, such as Tyr, Val, and Ile, are
more often present in structure, while Gly, Pro, and
Asn are likely to be found in bends.
270,270a,270b
The
frequencies with which particular amino acids appear
in helices, structure, or turns were first compiled by
TABLE 2-4
Classification of Protein Residues According to
Their Tendencies to Form Helix, Structure, and
Turns
a
a
The conformational parameters P

, P

, and P
t
( turn) are the
frequencies of finding a particular amino acid in an helix,
structure, or turn (in 29 proteins of known structure) divided by
the average frequency of residues in those regions. Residues are
arranged in order of decreasing tendency toward helix formation.
From Chou, P. V. and Fasman, G. D (1974) Biochemistry 13, 222
245.
i = indifferent
br = breaker
br+ = strong breaker
++ = strong former
+ = former
w = weak former
Glu
-
1.51 ++ 0.37 br+ 0.44
Met 1.45 ++ 1.05 + 0.67
Ala 1.42 ++ 0.83 i 0.57
Leu 1.21 ++ 1.30 + 0.53
Lys+ 1.16 + 0.74 br 1.01
Phe 1.13 + 1.38 + 0.71
Gln 1.11 + 1.10 + 0.56
Trp 1.08 + 1.37 + 1.11
Ile 1.08 + 1.60 ++ 0.58
Val 1.06 + 1.70 ++ 0.30
Asp
-
1.01 w 0.54 br+ 1.26
His
+
1.00 w 0.87 i 0.69
Arg
+
0.98 i 0.93 i 1.00
Thr 0.83 i 1.19 + 1.00
Ser 0.77 i 0.75 br 1.56
Cys 0.70 i 1.19 + 1.17
Tyr 0.69 br 1.47 ++ 1.25
Asn 0.67 br 0.89 i 1.68
Pro 0.57 br+ 0.55 br+ 1.54
Gly 0.57 br+ 0.75 br 1.68
P
t
P

Helix-
forming
tendency P

Amino
acid
structure-
forming
tendency
Figure 2-30 Plot of hydropathy index versus sequence number for bovine chymotrypsinogen. The indices for individual residues
have been averaged nine at a time. The solid bars at the top of the plot mark interior regions as determined by crystallography.
The solid bars below the plot indicate regions that are on the outside of the molecule. From Kyte and Doolittle.
280
40
20
0
-20
-40
H
y
d
r
o
p
a
t
h
i
c

i
n
d
e
x
20 40 60 80 100 120 140 160 180 200 220 240
Sequence number
0
79
F. Chemical Modification and Crosslinking
Some newly synthesized proteins, upon folding,
are ready to go to work immediately but others
must be modified. Pieces are frequently cut from the
ends or out of the center of a folded peptide chain.
Sometimes the amino and carboxyl termini are con-
verted to nonionic groups, e.g.,
Side chains may be modified in a very large num-
ber of different ways.
282284
These include acetylation
and other kinds of acylation (Eq. 2-12),
285287
methyl-
ation (Eqs. 2-14, 2-15), phosphorylation (Eq. 2-16),
phosphoadenylation,
288
formation of sulfate esters
(Eq. 2-17),
289,290
and hydrolysis (Eqs. 2-18, 2-19).
291
In
at least a few proteins some L-amino acid residues are
converted to D-.
292294
Chou and Fasman
270
(Table 2-4). These frequencies
also differ significantly between parallel and antiparal-
lel sheets. Some of the preferences are readily under-
standable. For example, glycine is too flexible to stabilize
a helix, but it can occur in helices between residues
that are better helix formers. Because the peptide link-
age to its nitrogen lacks an NH group, proline can fit
into only one of three positions at the N terminus of a
helix. For the strong helix formers, the -CH
2
groups
fit into the helix well and provide stabilization through
van der Waals interactions. However, the -methyl
groups of valine and isoleucine cause crowding. For
these residues, a structure is more favorable than a
helix.
271
Side chains of Asp, Asn, Ser, Thr, and Cys can
hydrogen bond to backbone amide groups and can
either stabilize or destabilize a helix or sheet depend-
ing upon their location.
It appears that the folding pattern of a peptide
is encoded in the sequence itself.
272274
Thus, when
several residues that favor helix formation are clus-
tered together, a helix may form. Chou and Fasman
suggested that when four helix formers out of six
residues are clustered, nucleation of a helix takes
place. The helix can then be elongated in both direc-
tions until terminated by a proline or other helix
breaker; additional folding can then occur. If three
out of five formers are clustered, a strand may
form. If random folding brings two or more of these
strands together they may associate to form the nucleus
for a sheet. Some success has been achieved using
this approach in predicting whether a given residue
will be found in a helix, a strand or a loop.
275
However, prediction of complete folding patterns is
much more difficult. Many new approaches are being
explored.
275279d
The problem is an important one.
Although three-dimensional structures have been
determined for thousands of proteins, sequences are
known for hundreds of thousands. The number is
growing rapidly. Being able to predict correctly a
three-dimensional protein structure will be a major
scientific accomplishment with many practical conse-
quences.
A way of examining the entire sequence for polar
or nonpolar character of the side chains was introduced
by Kyte and Doolittle.
280,281
A hydropathy index
based on the polarity of the side chains of a given
residue and of its neighbors in the sequence is plotted
against residue number (Fig. 2-30). Helices are often
found to have an amphipathic character, hydrophobic
on one side and hydrophilic on the other. Such helices
can be characterized by a plot such as that in Fig. 2-30.
The hydrophobic side of an amphipathic helix can
pack against a hydrophobic core of a protein, can lie
against a membrane, or can be aligned with other
helices to form coiled coils (Fig. 2-21) or to give a
hydrophilic channel with an outer hydrophobic sur-
face that can fit into a cell membrane.
(2-12)
(2-13)
NH
3
NH C R
O
R=CH
3
or long alkyl
+
C
O
N
H
CH
2
COO C
O
NH
2

(2-14)
(2-15)
(2-16)
(2-17)
(2-18)
(2-19)
COO C
OCH
3
O

NH
3
N CH
3
CH
3
CH
3
+ +
CH
2
OH CH
2
O P O
O
O

CH
2
OH CH
2
O SO
3

C
O
NH
2
C
O
O

+ H
2
O + NH
4
Asn, Gln
side chain
+
N C
NH
NH
2
H
N C
O
NH
2
H
Arginine
+ H
2
O + NH
4
Citrulline
+
+
F. Chemical Modification and Crosslinking
80 Chapter 2. Amino Acids, Peptides, and Proteins
Usually, only one or a small number of side chains
of a protein is modified. However, glycoproteins may
contain many different attached sugar molecules. An
example is the glucoamylase shown in Fig. 2-29. In
several proteins involved in blood clotting (Fig. 12-17),
as many as 10 glutamic acid side chains are carboxylated
(Eq. 2-20).
The 216-residue hen egg yolk storage protein
phosvitin contains 123 serine residues, most of which
have been phosphorylated (Eq. 2-16).
295
A basic protein
of the myelin sheath of neurons contains as many as 6
specific residues of citrulline (Eq. 2-19).
296
An adhesive
protein from the foot of a marine mollusk contains 80
repeated sequences containing hydroxy-proline 2,3-
dihydroxyproline and 3,4-dihydroxyphenylalanine
(Dopa).
297,298
Some modification reactions alter the electrical
charge on a side chain and, as a consequence, can
affect the ways in which the protein interacts with
other molecules. Negative charges added in formation
of clusters of -carboxyglutamates (Eq. 2-20) create
strong calcium ion-binding centers in the modified
proteins. Acylation of N termini or of Ser, Thr, or Cys
side chains by long-chain fatty acids provides hydro-
phobic tails able to anchor proteins to membrane
surfaces.
285,286
Addition of polyprenyl groups to cys-
teine side chains near the C-termini has a similar
effect.
299,300
These and more complex membrane
anchors are considered in Chapter 8. While modified
amino acids can be found at many places in a protein,
they are often located at turns. For example, serine or
threonine residues in turns are often phosphorylated
or glycosylated. Modification of proteins is dealt with
further in Chapter 10 and at other points in the book.
Glycosylation of proteins is considered in Chapter 20.
In addition to deliberate enzyme-catalyzed pro-
cesses, there are nonenzymatic processes that alter
proteins. These include the degradative reactions
described in Section 5 and also reversible reactions that
may be physiologically important. For example, the
N-terminal amino groups of peptides, and other amino
groups of low pK
a
can form carbamates with bicar-
bonate (Eq. 2-21).
301303
This provides an important
mechanism of carbon dioxide transport in red blood
cells (Chapter 7) and a way by which CO
2
pressure
can control some metabolic processes.
1. Disulfide Crosslinkages
The linking together of two different parts of a
peptide chain or of two different peptide chains is
extremely important to living beings.
304,305
One of
the most widespread of crosslinkages is the disulfide
bridge. It forms spontaneously when two SH groups
of cysteine side chains are close together and are oxi-
dized by O
2
or some other reagent (Eq. 2-22).
The disulfide group is inherently chiral, with the
preferred torsion angle about the SS bond being 90.
Both right- and left-handed disulfide bridges occur in
proteins.
305,306
Disulfide linkages are frequently present in pro-
teins that are secreted from cells but are less common
in enzymes that stay within cells. Perhaps because the
latter are in a protected environment, the additional
stabilization provided by disulfide bridges is not
needed. Disulfide bridges are not only found within
single-subunit proteins but they also link different
peptide chains. For example, the four chains of each
immunoglobulin molecule (Chapter 31) are joined by
disulfide bridges and each domain within the chains
is stabilized by a disulfide bridge. One of the most
highly crosslinked proteins known occurs in the kera-
tin matrix of hair. Breakage of the SS linkages of
this protein is an essential step in the chemical per-
manent wave process. A thiol compound is used to
reductively cleave the crosslinks and after the hair is
reset new crosslinks are formed by air oxidation.
2. Other Crosslinkages
Another common crosslink is an amide formed
between the -carboxyl group of a glutamic acid side
chain and an amino group from a lysine residue.
307
This isopeptide linkage is formed from a residue of
gluta-mine through the action of the enzyme trans-
glutaminase (Eq. 2-23). Isopeptide crosslinks are
found in hair, skin, connective tissue, and blood clots.
S CH
2
CH
2
Two S atoms viewed
down SS axis
(2-20)
CH
2
CH
2
COO CH
2
C
COO
COO
H

(2-21)
NH
3
+
NH C
O
OH
+ CO
2
+ H
+
(2-22)
CH
2
SH H
2
C
S S
CH
2
2
81
Occasionally an isopeptide linkage joins amino
groups of lysine side chains to the C-terminal carboxyl
groups of other peptide chains to give branched chains,
e.g. see ubiquitin (Box 10-C). Elaborate crosslinks
derived from lysine are found in collagen and elastin
(Chapter 8). Dityrosine linkages formed by oxidative
joining of the aromatic rings of tyrosine are found in
insect cuticle and in the plant cell wall extensins
202
(Chapters 20, 25).
G. Dynamic Properties of Proteins
Sometimes using energy from the cleavage of
chemical bonds, sometimes depending only upon
energy provided by the random bombardment by
solvent molecules, proteins perform their specific
functions with amazing speed. A question that has
long intrigued biochemists is to what extent do pro-
teins stretch or flex, unfold and refold, or undergo
other conformational movements during their action?
To answer this and related questions, many techniques
are being applied to study the dynamic properties of
proteins.
308,309
1. Motion of Backbone and Side Chains
Even in the crystalline state there is evidence of
movement. In the images constructed from X-ray or
neutron diffraction experiments side chains on the
surfaces of protein molecules are often not clearly
visible because of rapid rotational movement. Some
segments of the polypeptide chain may be missing
from the image. However, side chain groups within
the core of a domain are usually seen clearly. They
probably move only in discrete steps. However, they
may sometimes shift rapidly between different con-
formations, all of which maintain a close-packed
interior.
310312
Studies of nuclear magnetic resonance spectra
(Chapter 3) and of polarization of fluorescence (Chapter
23), have shown that there is rapid though restricted
rotational movement of side chains of proteins in
solution. Even buried phenylalanine and tyrosine side
chains often rotate rapidly whereas movement of the
bulkier tryptophan rings is more limited. Peptide NH
protons in unfolded polypeptide chains undergo rapid
acid or base catalyzed exchange with H
2
O.
313
However,
in globular proteins, the rate of exchange of buried
NH protons is often orders of magnitude slower.
314316
For most proteins there appears to be little tendency to
unfold completely and then refold; the major domains
hold together tightly. However, there may be local
unfolding, for example, of a helix at the surface of a
protein, which will allow more rapid exchange. Using
NMR techniques (Chapter 3), rates of exchange of all
of the individual peptide NH protons within small
proteins can be measured.
315
Cracks may open up in
proteins. This is suggested by the fact that O
2
, I
2
, and
certain other small molecules are apparently able to
penetrate the protein freely and to quench the fluores-
cence of buried tryptophan side chains (Chapter 23).
Since packing density tends to be lower at active
sites than in the bulk of the protein, it is probable that
more conformational alterations occur near active sites
than elsewhere.
317
Lumry and Rosenberg
318
suggested
that the defects of poor packing and poor hydrogen
bonding of some regions of a folded peptide chain
provide a store of potential energy that can be impor-
tant to the functioning of a protein. Even in a very
well-packed protein domain there are defects. Some
atoms are compressed by the folding of the peptide
chain and are closer together by over 0.04 nm than
predicted by the van der Waals radii. These packing
defects have been estimated to destablize the protein
by as much as 250 kJ/mol.
319
Details of structural
heterogeneity within several proteins for which very
precise structural data are available have been de-
scribed.
311
2. Conformational Changes
We have seen that some polypeptides assume an
extended conformation while others form helices.
In some cases, the same protein can do both. For
example, hair can be stretched greatly, the helices
of the keratin molecules uncoiling and assuming a
conformation with hydrogen bonds between chains
instead of within a single chain. Thus, a polymer may
have more than one conformational state in which the
folding and hydrogen bonding are different.
320,321
With soluble proteins more than one folded conforma-
tion is possible with different sets of hydrogen bonds
and internal hydrophobic interactions. Some of the
conformations of a globular protein are more stable
than others, and a protein will ordinarily assume one
of the energetically most favorable conformations.
However, there may be other conformations of almost
equal energy.
A large body of evidence suggests that many pro-
teins do exist in two or more different but well-defined
(2-23)
C

C
O
NH
2
C

H
3
N
C
N
H
C

O
Transglutaminase
NH
4
+
+
G. Dynamic Properties of Proteins
82 Chapter 2. Amino Acids, Peptides, and Proteins
conformational states and that the ability of a protein
to undergo easy conversion from one to another is of
great biological significance. In some cases such as
that of hemoglobin (Chapter 7), there are changes in
the interactions between subunits. Alterations in the
hinge regions between domains of immunoglobulins
have been seen. In many enzymes, including the
dehydrogenases (Chapter 15), kinases (Chapter 12), and
aspartate aminotransferase (Fig. 2-6), a cleft between
two domains appears to open and close.
322
In other
cases, more subtle alterations in conformational state,
involving mainly changes in the internal hydrogen
bonded network together with small localized changes
in chain folding, have been observed.
Conformational alterations in proteins are probably
facilitated by the fact that some hydrogen-bonded
groups are found within the hydrophobic interior. All
of the buried hydrogen atoms suitable for hydrogen bond
formation are ordinarily hydrogen bonded to an electron
donor group. However, because oxygen atoms in proteins
each have two unshared electron pairs, there are, in general,
more electron donor groups than there are hydrogen atoms
to which they can bind. This sets the stage for a competition
between electronegative centers for particular proton suitable
for hydrogen bonding and provides a molecular basis for the
easy triggering of conformational changes.
323
3. Denaturation and Refolding
An extreme conformational alteration is the dena-
turation of proteins, which may be caused by heating
or by treatment with reagents such as strong acids and
bases, urea, guanidinium chloride, and sodium
dodecyl sulfate (SDS).
Denaturation leads to unfolding of a protein to a
more random conformation. In the denatured state
the amide groups of the peptide chain form hydrogen
bonds with surrounding water molecules and with
denaturants such as urea or the guanidinium ion
rather than with each other.
324,325
Denaturants also
diminish the strength of the hydrophobic interactions
that promote folding.
326
Characteristic biochemical
activities are lost and physical properties such as sedi-
mentation constant, viscosity, and light absorption are
altered. The ease of denaturation of proteins and the
fact that denaturation is sometimes reversible show
that the energy differences between the folded confor-
mations and the open random coil conformation are
usually not great.
274,327,328
However, it has been diffi-
cult to establish the amount of stabilization of a folded
polypeptide chain provided by buried hydrogen
bonds
328a,328b,328c
or the role of cooperatively formed
hydrogen-bonded chains
328c,328d
or clusters.
328c,328d,328e
Complete denaturation of a protein was generally
regarded as an irreversible process prior to 1956 when
Anfinsen showed that denatured ribonuclease (Chapter
12) could refold spontaneously.
273
This 124-residue
protein contains four disulfide (SS) bridges and
thus is tied firmly together. When these bridges are
broken reductively in the presence of a denaturing
agent, the enzyme becomes inactive. Anfinsen found
that upon reoxidation under appropriate conditions,
complete activity reappeared. The molecules had
folded spontaneously into the correct conformation,
the one in which the correct one of 105 (7 x 5 x 3 x 1)
possible pairings of the eight SH groups present
needed to reform the four disulfide bridges had taken
place. This observation has had an important influence
on thinking about protein synthesis and folding of
polypeptide chains into biologically active molecules.
A puzzling problem was posed by Levinthal many
years ago.
329
We usually assume that the peptide
chain folds into one of the most stable conformations
possible. However, proteins fold very rapidly. Even
today, no computer would be able, in our lifetime, to
find by systematic examination the thermodynamically
most stable conformation.
328
It would likewise be
impossible for a folding protein to try out more than
a tiny fraction of all possible conformations. Yet folded
and unfolded proteins often appear to be in a thermo-
dynamic equilibrium! Experimental results indicate
that denatured proteins are frequently in equilibrium
with a compact denatured state or molten globule
in which hydrophobic groups have become clustered
and some secondary structures exists.
330336
From this
state the polypeptide may rearrange more slowly
through other folding intermediates to the final
native conformation.
336a,336b
It is generally assumed that within cells the folding
of the peptide chain commences while the chain is still
being synthesized on a ribosome. The growing chain
probably folds rapidly in a random way until it finds
one or more stable conformations which serve as
folding intermediates for slower conversion to the
finished protein.
337,338
However, any process within
a cell is affected by the complex intracellular environ-
ment.
339
Folding can be catalyzed or inhibited by
proteins known as molecular chaperones. Folding
may sometimes require isomerization of one or a few
proline residues from trans to cis.
340,341
For example,
during the refolding of ribonuclease the isomerization
of Pro 93 appears to be a rate-limiting step.
342,343
Such
C
H
2
N
H
2
N
O C
NH
2
H
2
N NH
2
Na
Urea
Guanidinium
chloride
CH
3
(CH
2
)
11
OSO
3
+
Sodium dodecyl sulfate (SDS)
+
83
BOX 2-C THE NOBEL PRIZES
Many young scientists dream of one day winning a Nobel Prize. Although denounced by some, the much sought and
highly publicized award has, since 1901, been given to an outstanding group of scientists. Many of these have made major
contributions to biochemistry or to techniques important to biochemists. Here is a partial list.
isomerizations may be assisted by peptidyl-prolyl-
(cistrans) isomerases (Box 9-F). Disulfide linkages
are sometimes formed incorrectly.
344,345
A protein-
disulfide isomerase catalyzes cleavage and reformation
of these bridges, allowing the protein to find the most
stable crosslinking arrangement. The actions of these
enzymes and of molecular chaperones are considered
further in Chapters 10 and 12.
4. Effects of pH and Solvent
Because polypeptide chains contain many acidic
and basic amino acid side chains, the properties of
proteins are greatly influenced by pH. At low pH
carboxylates, S

, and imidazole groups accept protons


causing the overall net charge on the macromolecule
to be strongly positive. At high pH protons are lost
and the protein becomes negatively charged. Electro-
static repulsion between like charges may cause pro-
teins to denature at low or high pH. More stable
proteins may be very soluble at low or high pH. Pro-
teins often have a minimum solubility and a maximum
stability near the isoelectric point, the pH at which
the net charge is zero.
336,346,347
Activities of enzymes,
abilities to bind specifically to other proteins, and
Year Name Prize
a
Discovery or subject of study
1901 Wilhelm C. Rntgen Physics Discovery of X-rays
1902 Emil H. Fischer Chemistry Synthesis of sugars and purines
1903 Svante A. Arrhenius Chemistry Electrolytic dissociation; a founder of physical chemistry
1903 Antoine Henri Becquerel; Marie S. Curie, and Pierre Curie Physics Discovery and study of radioactivity
1906 Camillo Golgi and S. Ramon y Cajal Physiology/Medicine Discovery of Golgi apparatus
1907 Edward Buchner Chemistry Biochemistry, cell-free fermentation
1910 Albrecht Kossel Physiology/Medicine Isolation of nucleic acid bases
1914 Max von Laue Physics Discovery of X-ray diffraction by crystals
1915 Richard M. Willsttter Chemistry Plant pigments, especially chlorophyll
1915 William H. and William L. Bragg Physics Analysis of crystal structure by X-rays
1919 Jules Bordet Physiology/Medicine Discovery of blood complement; complement fixation test
1920 Walther H. Nernst Chemistry Thermochemistry
1922 Archibald V. Hill and Otto F. Meyerhof Physiology/Medicine Chemistry of muscle contraction
1923 Frederick G. Banting and John J. R. Macleod Physiology/Medicine Discovery of insulin and treatment of diabetes
1926 Theodor Svedburg Chemistry Study of high M
r
compounds, development of ultracentrifuge
1927 Heinrich O. Wieland Chemistry Bile acids
1928 Adolf O. R. Windaus Chemistry Sterols and vitamins
1929 Frederick G. Hopkins and Christiaan Eijkman Physiology/Medicine Discovery of vitamins, tryptophan, vitamin B
1
1929 Arthur Harden and Hans A. S. von Euler-Chelpin Chemistry Fermenting enzymes, fermentation of sugars
1930 Karl Landsteiner Physiology/Medicine Blood groups A, B, O, Rh
1930 Hans Fischer Chemistry Structures and chemistry of porphyrins, chlorophyll
1931 Otto H. Warburg Physiology/Medicine Respiratory enzymes
1933 Thomas H. Morgan Physiology/Medicine Chromosome theory and chromosome maps
1934 George R. Minot, William P. Murphy, and George H. Whipple Physiology/Medicine Treatment of pernicious anemia
1936 Henry H. Dale and Otto Loewi Physiology/Medicine Acetylcholine release at nerve endings
1937 Albert von Szent-Gyrgyi Physiology/Medicine Vitamin C
1937 Walter N. Haworth and Paul Karrer Chemistry Carbohydrate structures, structures of carotenoids, flavins, vitamin A
1938 Richard Kuhn Chemistry Carotenoids and vitamins
1939 Gerhard Domagk Physiology/Medicine Prontosil, first antibacterial sulfa drug
1939 Adolf F. J. Butenandt and L. Ruzicka Chemistry Isolation and study of sex hormones, study of polymethylenes, terpenes
1943 E. A. Doisy and Carl Henrik Dam Physiology/Medicine Isolation and study of vitamin K
1945 Alexander Fleming, Ernst B. Chain, and Howard W. Florey Physiology/Medicine Discovery and structure of penicillin
1945 A. J. Virtanen Chemistry Nutritional chemistry
1946 James B. Sumner, J. H. Northrop, and W. M. Stanley Chemistry Crystallization of enzymes and virus proteins
1947 Carl F. Cori and Gerty T. Cori Physiology/Medicine Glycogen metabolism, the Cori cycle
1947 B. A. Houssay and Robert Robinson Chemistry Investigation of plant alkaloids
1948 Arne W. K. Tiselius Chemistry Electrophoresis, study of serum proteins
1950 Phillip S. Hench, Edward C. Kendall, and Tadeus Reichstein Physiology/Medicine ACTH
1952 Selman A. Waksman Physiology/Medicine Discovery of streptomycin
1952 A. J. P. Martin and Richard L. M. Synge Chemistry Paper chromatography
1953 Fritz A. Lipmann and Hans A. Krebs Physiology/Medicine Coenzyme A, citric acid cycle
1954 Linus C. Pauling
b
Chemistry The nature of the chemical bond
1955 A. H. T. Theorell Physiology/Medicine Oxidative enzymes
1955 Vincent du Vigneaud Chemistry Synthesis of biotin and oxytocin
1957 Daniel Bovet Physiology/Medicine First antihistamine drug
1957 Alexander R. Todd Chemistry Work on nucleotides coenzymes
1958 Joshua Lederberg, George W. Beadle, and Edward L. Tatum Physiology/Medicine One gene-one enzyme hypothesis from genetic studies of neurospora
1958 Frederick Sanger
b
Chemistry Protein sequencing, insulin
G. Dynamic Properties of Proteins
84 Chapter 2. Amino Acids, Peptides, and Proteins
numerous physiological processes are controlled by
effects of pH on proteins.
The solvent for most proteins in nature is water.
However, many enzymes function well in organic
solvents if they retain only a small amount of essential
structural and catalytic water.
348
5. Irreversible Damage to Proteins
Every protein in an organism has its own charac-
teristic lifetime. No sooner is it synthesized than degra-
dation begins. Enzyme-catalyzed cleavage of the
peptide linkages leads to turnover of proteins but
before this occurs a number of spontaneous damaging
reactions may alter the protein. Prevalent among these
is deamidation of asparaginyl residues to aspartyl or
BOX 2-C THE NOBEL PRIZES (continued)
a
Prizes are given in Physics, Chemistry, Physiology or Medicine, Literature and Peace.
b
Sanger and Pauling each have been awarded two Nobel Prizes.
Year Name Prize
a
Discovery or subject of study
1959 Arthur Kornberg and Severo Ochoa Physiology/Medicine Enzymatic synthesis of DNA
1960 F. M. Burnet and P. Medawar Physiology/Medicine Immunological tolerance in animals
1961 Melvin Calvin Chemistry Photosynthesis using
14
CO
2
1962 Francis H. C. Crick, James D. Watson, and Maurice H. F. Wilkins Physiology/Medicine Molecular structure of DNA
1962 John C. Kendrew and Max F. Perutz Chemistry Structures of protein by X-ray diffraction
1962 Linus C. Pauling
b
Peace Ending atmospheric testing of nuclear weapons
1964 Konrod E. Bloch and Feodor Lynen Physiology/Medicine Pathways of cholesterol biosynthesis
1964 Dorothy M. Crowfoot Hodgkin Chemistry X-ray structures, vitamin B
12
1965 Andre Lwolf, Jacques Monod, and Francois Jacob Physiology/Medicine Messenger RNA, regulation of transcription
1967 Ragnar Granit, Haldan Keffer Hartline, and George Wald Physiology/Medicine Chemistry of vision
1967 Manfred Eigen and Norrish Porter Chemistry Study of high-speed chemical reactions
1968 Robert W. Holley Physiology/Medicine RNA sequence
1968 H. Gobind Khorana and Marshall W. Nirenberg Physiology/Medicine The genetic code
1969 Max Delbrck, Alfred D. Hershey, and Salvador E. Luria Physiology/Medicine Replication and genetic structures of viruses
1970 Julius Axelrod, Bernard Katz, and Ulf von Euler Physiology/Medicine Transmission of nerve impulses, noradrenaline
1970 Luis F. Leloir Chemistry Role of sugar nucleotides in biosynthesis of carbohydrates
1971 Earl W. Sutherland, Jr. Physiology/Medicine Cyclic AMP
1972 Gerald M. Edelman and Rodney R. Porter Physiology/Medicine Structure of antibodies
1972 Christian B. Anfinsen, Sanford Moore, and William H. Stein Chemistry Ribonuclease, structure and activity
1974 Albert Claude, Christian R. de Duve, and George E. Palade Physiology/Medicine Cell structure
1975 David Baltimore, Renato Dulbecco, and Howard M. Temin Physiology/Medicine Reverse transscriptase
1975 John W. Cornforth and Vladimir Prelog Chemistry Stereochemistry of organic molecules and of enzymatic reactions
1977 Rosalyn S. Yalow, Roger C. L. Guillemin, and Andrew V. Schally Physiology/Medicine Radioimmunoassay thyrotropin-releasing hormone
1978 Daniel Nathans, Werner Arber, and Hamilton O. Smith Physiology/Medicine Restriction enzymes
1978 Peter Mitchell Chemistry Biological energy transfer
1980 Paul Berg, Walter Gilbert, and Frederick Sanger
b
Chemistry Recombinant DNA, methods of sequence determination for DNA
1982 Sune K. Bergstrom, Bergt I. Samuelsson, and John R. Vane Physiology/Medicine Isolation and study of prostaglandins
1982 Aaron Klug Chemistry Development of crystallographic electron microscopy
1983 Barbara McClintock Physiology/Medicine Gene transposition
1984 Niels K. Jerne, Georges J. F. Khler, and Cesar Milstein Physiology/Medicine Cellular basis of immunology
1984 R. Bruce Merrifield Chemistry Solid-phase synthesis of peptides
1985 Joseph L. Goldstein and Michael S. Brown Physiology/Medicine Control of cholesterol metabolism
1986 Rita Levi-Montalcini and Stanley Cohen Physiology/Medicine Nerve growth factor
1987 Susumu Tonegawa Physiology/Medicine Genetics of antibody formation
1988 Johan Diesenhofer, Robert Huber, and Hartmut Michel Chemistry Three-dimensional structure of a photosynthetic reaction center
1988 Gertrude Elion, George Hitchings, and James Black Physiology/Medicine Principles of drug treatment and design of many important drugs
1989 J. Michael Bishop, Harold E. Varmus, and Joseph E. Murray Physiology/Medicine Origin of retroviral oncogenes
1989 Sidney Altman and Thomas R. Cech Chemistry Catalytic RNA
1991 Erwin Neher and Bert Sakmann Physiology/Medicine Functioning of single ion channels in cells
1991 Richard R. Ernst Chemistry High-resolution NMR
1992 Edmond H. Fischer and Edwin G. Krebs Physiology/Medicine Reversible protein phosphorylation in biological regulation
1992 Rudolph A. Marcus Chemistry Theory of electron transfer reactions
1993 Richard J. Roberts and Phillip A. Sharp Physiology/Medicine Discovery of split genes
1993 Michael Smith and Kary B. Mullis Chemistry Oligonucleotide-directed mutagenesis and polymerase chain reaction
1994 Alfred Gilman and Martin Rodbell Physiology/Medicine G-proteins
1995 Edwin B. Lewis, Christiane Nusslein-Volhard, and Eric Wieschaus Physiology/Medicine Homeotic mutations in Drosophila
1995 Paul Crutzen, Sherwood Rowland, and Mario Molina Chemistry Damage to the stratospheric ozone layer
1996 Peter C. Doherty and Rolf M. Zinkernagel Physiology/Medicine Specificity of cell-mediated immune response
1997 Paul D. Boyer and John E. Walker Chemistry Mechanism of ATP synthesis
1997 Jens C. Skou Chemistry Discovery of Na
+
, K
+
-ATPase
1997 Stanley B. Prusiner Physiology/Medicine Discovery of prions
1998 Robert F. Furchgott, Louis J. Ignarro, and Ferid Murad Physiology/Medicine Nitric oxide as a signaling molecule
1999 Gnter Blobel Physiology/Medicine Intrinsic signals that govern transport and localization of proteins
2000 Arvid Carlsson Physiology/Medicine Identification of dopamine as signaling molecule in brain
Paul Greengard Physiology/Medicine Discovery of the dopamine signaling cascade
Eric R. Kandel Physiology/Medicine Molecular basis of learning
85
isoaspartyl groups (Eq. 2-24).
349,350
Aspartyl residues
can undergo the same type of cyclization at low pH.
The sequence Asn-Gly is especially susceptible to
rearrangement according to Eq. 2-24. However, the
peptide torsion angles may be more important in
determining whether deamidation occurs. The inter-
mediate succinimide may be racemized easily at the
chiral center marked by the asterisk in Eq. 2-24. Thus,
the Asn-Gly sequence represents a weak linkage,
which is nevertheless present in many proteins.
Cystine residues in disulfide bridges are also not
completely stable but undergo elimination
305,351
(Chapter 13) at slightly alkaline pH values according
to Eq. 2-25. The free thiol group formed (HSR) is still
attached to the protein but may sometimes be in a
position to enter into thiol-disulfide exchange reactions
with other SS bridges causing further degradation.
Methionine side chains in proteins can be oxidized to
sulfoxides: and hydroxyl groups
can be introduced into aromatic rings by oxidation.
Hydrogen peroxide and other oxidants may be respon-
sible for such oxidation within cells (see Chapter 18).
The very long-lived proteins of the lens of the eye are
especially susceptible to deamidation, racemization,
oxidation, and accumulation of covalently attached
blue fluorescent compounds.
352354
H. Design and Engineering of Proteins
H. Design and Engineering of Proteins
Methods of chemical synthesis of polypeptides and
of cloning and mutating genes now allow us to alter
peptide sequences at will and to design completely
new proteins.
355356b
The methods are discussed in
Chapters 3, 5, and 26. The following are examples.
Peptides that form helices that associate as
coiled coils,
357
or as three- or four-helix tetrameric
bundles
179a,358360
or amphipathic helices that associate
with lipid bilayers have been made.
355,361
More difficult
has been the design of proteins that form sheets.
362364a
Many efforts are being made to understand protein
stability
365367
by systematic substitutions of one residue
for another. Addition of new disulfide linkages at
positions selected by study of three-dimensional struc-
tures sometimes stabilizes enzymes.
368371
On the other
hand elimination of unnecessary cysteine residues can
enhance stability by preventing elimination
351
and
replacement of asparagine by threonine can improve
the thermostability of enzymes by preventing deami-
dation.
372,373
Artificial mutants of subtilisin
373
and of many
other enzymes are helping us to understand the mech-
anisms of catalysis. Artificially prepared temperature-
sensitive mutants (Chapter 26) and naturally occurring
mutants are providing new insight into pathways of
folding of proteins whose three-dimensional structures
are known. For these studies it is necessary to follow a
rational strategy in deciding which of the astronomical
number of possible mutant forms may be of interest.
For example, residues within active sites may be
changed. Introduction of mutations at random locations
can also be useful in finding regions of interest.
374
When mutant proteins can be crystallized, an exact
understanding of the effect of the mutation on the
structure is possible. Sometimes two or more mutations
may have to be introduced to obtain the desired modi-
fication.
It is also possible to incorporate a range of un-
natural amino acids in specific sites in a polypeptide
sequence and to observe resulting effects on a proteins
properties.
375
A quite different approach is to design
polypeptides that mimic a natural peptide but consist
of D-amino acids. The peptide chain is reversed, i.e., the
N terminus becomes the C terminus and every peptide
linkage is also reversed. The amino acid side chains
preserve their relationships one to another and the back-
bone atoms tend to preserve their hydrogen bonding
pattern. Peptides made in this way tend to be resistant
to cleavage by enzymes. Some may be useful as
drugs.
376,377
(2-24)
(2-25)
N C
H
CH
2
H O
S S R
N C
H O
CH
2
HS SR
HS R + S
0

CH
2
CH
2
SCH
3
O
N
H
C
H
N
H
2
C
C
NH
2
H
O
H
O
N
H
C N
H
2
C
C
H
O
O
N
H
C
O
H
2
C
C
N
H
O
H
O
l-asparaginyl
OH
l-succinimidyl
l-isoaspartyl
*
+

86 Chapter 2. Amino Acids, Peptides, and Proteins


References
1. Glendening, E. D., Faust, R., Streitwieser, A.,
Vollhardt, K. P. C., and Weinhold, F. (1993) J.
Am. Chem. Soc. 115, 1095210957
2. Gao, Q., Weber, H.-P., Craven, B. M., and
McMullan, R. K. (1994) Acta Crystallogr. B50,
695703
3. Hegstrom, R. A., and Kondepudi, D. K. (1990)
Sci. Am. 262(Jan), 108115
4. Cahn, R. S., Ingold, C., and Prelog, V. (1966)
Angew. Chem. Int. Ed. Engl. 5, 385415
5. Eliel, E. L., Wilen, S. H., and Mander, L. N.
(1994) Stereochemistry of Organic Compounds,
Wiley, New York
6. Bentley, R. (1969) Molecular Asymmetry in
Biology, Vol. 1, Academic Press, New York
(pp. 4956)
7. Blackwood, J. E., Gladys, C. L., Loening, K. L.,
Petrarca, A. E., and Rush, J. E.
(1968) J. Am. Chem. Soc. 90, 509510
8. Hanson, K. R. (1966) J. Am. Chem. Soc. 88,
27312742
9. Lambert, J. B. (1970) Sci. Am. 222(Jan), 5870
10. Klyne, W., and Prelog, V. (1960) Experientia 16,
521523
11. Libecq, C., ed. (1992) Biochemical Nomencla-
ture, Portland Press, London and Chapel Hill,
North Carolina (for the International Union of
Biochemistry and Molecular Biology)
12. Chiang, Y., Kresge, A. J., and Tang, Y. S. (1984)
J. Am. Chem. Soc. 106, 460462
13. Beak, P., Fry, F. S., Jr., Lee, J., and Steele, F.
(1976) J. Am. Chem. Soc. 98, 171179
14. Beak, P. (1977) Acc. Chem. Res. 10, 186192
15. Metzler, D. E., Harris, C. M., Johnson, R. J.,
Siano, D. B., and Thomson, J. A. (1973)
Biochemistry 12, 53775392
16. Katritzky, A. R., and Karelson, M. (1991) J.
Am. Chem. Soc. 113, 15611566
17. Blomberg, F., Maurer, W., and Rterjans, H.
(1977) J. Am. Chem. Soc. 99, 81498159
18. Walters, D. E., and Allerhand, A. (1980) J. Biol.
Chem. 255, 62006204
19. Fersht, A. (1977) Enzyme Structure and
Mechanism, Freeman, San Francisco,
California
20. Gabler, R. (1978) Electrical Interactions in
Molecular Biophysics, Academic Press, New
York (p. 188)
21. Israelachvili, J. N., and McGuiggan, P. M.
(1988) Science 241, 795800
22. Salem, L. (1962) Can. J. Biochem. Physiol. 40,
12871298
23. Tsao, Y.-H., Fennell Evans, D., and
Wennerstrm, H. (1993) Science 262, 547550
23a. Kurihara, K., and Kunitake, T. (1992) J. Am.
Chem. Soc. 114, 1092710933
23b. Yang, L., Valdeavella, C. V., Blatt, H. D., and
Pettitt, B. M. (1996) Biophys. J. 71, 3022 3029
24. Warshel, A. (1987) Nature (London) 330, 1516
25. Honig, B., and Nicholls, A. (1995) Science 268,
11441149
26. Meot-Ner, M. M. (1988) J. Am. Chem. Soc. 110,
30753080
27. Loewenthal, R., Sancho, J., Reinikainen, T.,
and Fersht, A. R. (1993) J. Mol. Biol. 232, 574
583
28. Gristina, A. G., Oga, M., Webb, L. X., and
Hobgood, C. D. (1985) Science 228, 990993
29. Jeffrey, G. A., and Saenger, W. (1991) Hydrogen
Bonding in Biological Structures, Springer-
Verlag, Berlin
30. Gilli, P., Bertolasi, V., Ferretti, V., and Gastone,
G. (1994) J. Am. Chem. Soc. 116, 909915
30a. Jeffrey, G. A. (1997) An Introduction to Hydrogen
Bonding, Oxford Univ. Press, New York
31. Derewenda, Z. S., Derewenda, U., and Kobos,
P. M. (1994) J. Mol. Biol. 241, 8393
31a. Wahl, M. C., and Sundaralingam, M. (1997)
Trends Biochem. Sci. 22, 97102
31b. Bella, J., and Berman, H. M. (1996) J. Mol. Biol.
264, 734742
32. Schulz, G. E., and Schirmer, R. H. (1979)
Principles of Protein Structure, Springer, New
York (p. 36)
32a. Platts, J. A., Howard, S. T., and Bracke, B. R. F.
(1996) J. Am. Chem. Soc. 118, 27262733
33. Taylor, R., Kennard, O., and Versichel, W.
(1983) J. Am. Chem. Soc. 105, 57615766
34. Murray-Rust, P., and Glusker, J. P. (1984) J.
Am. Chem. Soc. 106, 10181025
35. Grbitz, C. H., and Etter, M. C. (1992) J. Am.
Chem. Soc. 114, 627631
36. Nelson, D. D., Jr., Fraser, G. T., and
Klemperer, W. (1987) Science 238, 16701674
36a. Ghanty, T. K., Staroverov, V. N., Koren, P. R.,
and Davidson, E. R. (2000) J. Am. Chem. Soc.
122, 1210 1214
36b. Cornilescu, G., Ramirez, B. E., Frank, M. K.,
Clore, G. M., Gronenborn, A. M., and Bax, A.
(1999) J. Am. Chem. Soc. 121, 6275 6279
36c. Benedict, H., Shenderovich, I. G., Malkina, O.
L., Malkin, V. G., Denisov, G. S., Golubev, N.
S., and Limbach, H.-H. (2000) J. Am. Chem.
Soc. 122, 1979 1988
37. Fersht, A. R., Shi, J., Knill-Jones, J., Lowe, D.
M., Witkinson, A. J., Blow, D. M., Brick, P.,
Carter, P., Woye, M. M. Y., and Winter, G.
(1985) Nature (London) 314, 235 238
38. Eriksson, A. E., Baase, W. A., Zhang, X.-J.,
Heinz, D. W., Blaber, M., Baldwin, E. P., and
Matthews, B. W. (1992) Science 255, 178183
38a. Takano, K., Yamagata, Y., Kubota, M.,
Funahashi, J., Fujii, S., and Yutani, K. (1999)
Biochemistry 38, 6623 6629
39. Lim, W. A., Hodel, A., Sauer, R. T., and
Richards, F. M. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 423427
40. Schuster, P., Zundel, G., and Sandorfy, C.
(1976) The Hydrogen Bond, North Holland
Publ. Co., Amsterdam (3 vols.)
41. Luck, W. A. P., and Kleeberg, H. (1978) in
Photosynthetic Oxygen Evolution (Metzner, H.,
ed), pp. 129, Academic Press, London
42. Edsall, J. T. (1983) Trends Biochem. Sci. 8, 2931
43. Grunwald, E. (1986) J. Am. Chem. Soc. 108,
57195726
44. Westhof, E. (1993) Water and Biological
Macromolecules, CRC Press, Boca Raton, Florida
45. Kusalik, P. G., and Svishchev, I. M. (1994)
Science 265, 12191221
46. Libnau, F. O., Toft, J., Christy, A. A., and
Kvalheim, O. M. (1994) J. Am. Chem. Soc. 116,
83118316
47. Benson, S. W., and Siebert, E. D. (1992) J. Am.
Chem. Soc. 114, 42694276
48. Liu, K., Cruzan, J. D., and Saykally, R. J. (1996)
Science 271, 929933
49. Pauling, L. (1935) J. Am. Chem. Soc. 57, 2680
50. Wittebort, R. J., Usha, M. G., Ruben, D. J.,
Wemmer, D. E., and Pines, A. (1988) J. Am.
Chem. Soc. 110, 56685671
51. Li, J., and Ross, D. K. (1993) Nature (London)
365, 327329
52. Dyke, T. R., Mack, K. M., and Muenter, J. S.
(1977) J. Chem. Phys. 66, 498510
53. Saenger, W. (1979) Nature (London) 279, 280,
343344, 848
54. Saenger, W. (1987) Ann. Rev. Biophys. Biophys.
Chem. 16, 93114
55. Pribble, R. N., and Zwier, T. S. (1994) Science
265, 7579
56. Gregory, J. K., Clary, D. C., Liu, K., Brown, M.
G., and Saykally, R. J. (1997) Science 275, 814
817
57. Quiocho, F. A., Sack, J. S., and Vyas, N. K.
(1987) Nature (London) 329, 561564
58. Pflugrath, J. W., and Quiocho, F. A. (1988) J.
Mol. Biol. 200, 163180
59. Kauzmann, W. (1959) Adv. Prot. Chem. 14, 163
60. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York (p. 393)
61. Tanford, C. (1980) The Hydrophobic Effect:
Formation of Micelles and Biological Membranes,
2nd ed., Wiley, New York
62. Abraham, M. H. (1980) J. Am. Chem. Soc. 102,
59105912
63. Ben-Nim, A. (1980) Hydrophobic Interactions,
Plenum, New York
64. Murphy, K. P., Privalov, P. L., and Gill, S. J.
(1990) Science 247, 559561
65. Muller, N. (1992) Trends Biochem. Sci. 17, 459
463
66. Spolar, R. S., Livingstone, J. R., and Record, J.,
MT. (1992) Biochemistry 31, 39473955
67. Stites, W. E., Meeker, A. K., and Shortle, D.
(1994) J. Mol. Biol. 235, 2732
68. Hecht, D., Tadesse, L., and Walters, L. (1993)
J. Am. Chem. Soc. 115, 33363337
69. Gill, S. J., and Wads, I. (1976) Proc. Natl.
Acad. Sci. U.S.A. 73, 29552958
70. Sharp, K. A., Nicholls, A., Fine, R. F., and
Honig, B. (1991) Science 252, 106109
71. Van Oss, C. J. (1994) Interfacial Forces in
Aqueous Media, Dekker, New York
71a. Hummer, G., Garde, S., Garca, A. E.,
Pohorille, A., and Pratt, L. R. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 89518955
72. Israelachvili, J., and Wennerstrm, H. (1996)
Nature (London) 379, 219225
73. Radzicka, A., and Wolfenden, R. (1988)
Biochemistry 27, 16641670
74. Kellis, J. T., Jr., Nyberg, K., Sali, D., and Fersht,
A. R. (1988) Nature (London) 333, 784786
75. Sanger, F. (1988) Ann. Rev. Biochem. 57, 128
76. Creighton, T. E. (1993) Proteins, Structures and
Molecular Principles, 2nd ed., Freeman, New
York
77. Kyte, J. (1995) Structure in Protein Chemistry,
Garland Publ., New York
78. Guillemin, R., and Burgus, R. (1972) Sci. Am.
227(Nov), 2433
79. Hochster, R. M., Kates, M., and Quastel, J. H.,
eds. (1973) Metabolic Inhibitors, Vol. 3,
Academic Press, New York (p. 312)
80. Ramachandran, G. N., and Kolaskar, A. S.
(1973) Biochim. Biophys. Acta. 303, 385388
81. MacArthur, M. W., and Thornton, J. M. (1996)
J. Mol. Biol. 264, 1180-1195
81a. Milner-White, E. J. (1997) Protein Sci. 6, 2477
2482
82. Moffat, J. B. (1973) J. Theor. Biol. 40, 247258
83. Fersht, A. R. (1971) J. Am. Chem. Soc. 93, 3504
3515
84. Karle, I. L. (1974) Biochemistry 13, 21552162
85. Ovchinnikov, Y. A. (1973) FEBS Lett. 29, 3133
86. Doonan, S. (1974) FEBS Lett. 38, 229233
87. Bhown, A. S. (1990) Handbook of Proteins, A & M
Publ., Birmingham, Alabama
88. Boguski, M., and McEntyre, J. (1994) Trends
Biochem. Sci. 19, 71
88a. Branden, C., and Tooze, J. (1991) Introduction
to Protein Structure, Garland Publ., New York
89. Clements, S., Mehansho, H., and Carlson, D.
M. (1975) J. Biol. Chem. 260, 1347113477
90. Condit, C. M., and Meagher, R. B. (1986)
Nature (London) 323, 178181
91. Reynolds, P., Weber, S., and Prakash, L. (1985)
Proc. Natl. Acad. Sci. U.S.A. 82, 168172
92. Rodakis, G. C., and Kafatos, F. C. (1982) Proc.
Natl. Acad. Sci. U.S.A. 79, 35513555
93. Young, J. F., Hockmeyer, W. T., Gross, M.,
Ballou, W. R., Wirtz, R. A., Trosper, J. H.,
Beaudoin, R. L., Hollingdale, M. R., Miller, L.
H., Diggs, C. L., and Rosenberg, M. (1985)
Science 228, 958962
87
152. Nicholson, H., Anderson, D. E., Dao-pin, S.,
and Matthews, B. W. (1991) Biochemistry 30,
98169828
153. qvist, J., Luecke, H., Quiocho, F. A., and
Warshel, A. (1991) Proc. Natl. Acad. Sci. U.S.A.
88, 20262030
154. Lyu, P. C., Wemmer, D. E., Zhou, H. X.,
Pinker, R. J., and Kallenbach, N. R. (1993)
Biochemistry 32, 421425
155. Viguera, A. R., and Serrano, L. (1995) J. Mol.
Biol. 251, 150160
156. Doig, A. J., Chakrabartty, A., Klingler, T. M.,
and Baldwin, R. L. (1994) Biochemistry 33,
33963403
157. Doig, A. J., Macarthur, M. W., Stapley, B. J., and
Thornton, J. M. (1997) Protein Sci. 6, 147155
158. Sali, D., Bycroft, M., and Fersht, A. R. (1988)
Nature (London) 335, 740743
159. Aurora, R., and Rose, G. D. (1998) Protein Sci.
7, 2138
160. Metzler, D. E. (1979) Adv. Enzymol. 50, 140
161. Lupas, A. (1996) Trends Biochem. Sci. 21,
375 382
161. Lupas, A. (1996) Trends Biochem. Sci. 21, 375
382
162. Kohn, W. D., Mant, C. T., and Hodges, R. S.
(1997) J. Biol. Chem. 272, 25832586
163. Fraser, R. D. B., MacRae, T. P., Parry, D. A. D.,
and Suzuki, E. (1986) Proc. Natl. Acad. Sci.
U.S.A. 83, 11791183
164. Johnson, L. D., Idler, W. W., Zhou, X.-M.,
Roop, D. R., and Steinert, P. M. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 18961900
165. Cooper, D., and Sun, T.-T. (1986) J. Biol. Chem.
261, 46464654
166. Crick, F. H. C. (1953) Acta Crystallogr. 6, 689
697
167. Vazquez, S. R., Kuo, D. Z., Bositis, C. M.,
Hardy, L. W., Lew, R. A., and Humphreys, R.
E. (1992) J. Biol. Chem. 267, 74067410
168. OShea, E. K., Llemm, J. D., Kim, P. S., and
Alber, T. (1991) Science 254, 539544
169. Ferr-DAmar, A. R., Prendergast, G. C., Ziff,
E. B., and Burley, S. K. (1993) Nature (London)
363, 3844
170. Lyu, P. C., Marky, L. A., and Kallenbach, N. R.
(1989) J. Am. Chem. Soc. 111, 27332734
171. Graddis, T. J., Myszka, D. G., and Chaiken, I.
M. (1993) Biochemistry 32, 1266412671
171a. Fairman, R., Chao, H.-G., Lavoie, T. B.,
Villafranca, J. J., Matsueda, G. R., and
Novotny, J. (1996) Biochemistry 35, 28242829
171b. Kohn, W. D., Kay, C. M., and Hodges, R. S.
(1998) J. Mol. Biol. 283, 9931012
172. John, M., Briand, J.-P., Granger-Schnarr, M.,
and Schnarr, M. (1994) J. Biol. Chem. 269,
1624716253
173. Zhou, N. E., Kay, C. M., and Hodges, R. S.
(1992) Biochemistry 31, 57395746
174. Cohen, C., and Parry, D. A. D. (1986) Trends
Biochem. Sci. 11, 245248
175. Phillips, G. N., Jr., Fillers, J. P., and Cohen, C.
(1986) J. Mol. Biol. 192, 111131
176. Mo, J., Holtzer, M. E., and Holtzer, A. (1993)
Protein Sci. 2, 128130
177. Xie, X., Rao, S., Walian, P., Hatch, V., Phillips,
G. N., Jr., and Cohen, C. (1994) J. Mol. Biol.
236, 12121226
178. Eckert, R. L. (1988) Proc. Natl. Acad. Sci. U.S.A.
85, 11141118
179. Gernert, K. M., Surles, M. C., Labean, T. H.,
Richardson, J. S., and Richardson, D. C. (1995)
Protein Sci. 4, 2252 2260
179a. Monera, O. D., Zhou, N. E., Lavigne, P., Kay,
C. M., and Hodges, R. S. (1996) J. Biol. Chem.
271, 3995 4001
180. Lovejoy, B., Choe, S., Cascio, D., McRorie, D.
K., DeGrado, W. F., and Eisenberg, D. (1993)
Science 259, 12881293
References
94. Doolittle, R. F. (1987) Of URFS and ORFS: A
Primer on How to Analyze Derived Amino Acid
Sequences, Univ. Science Books, Mill Valley,
California
95. Doolittle, R. F. (1989) Trends Biochem. Sci. 14,
244245
96. Pearson, W. R., and Lipman, D. J. (1988) Proc.
Natl. Acad. Sci. U.S.A. 85, 24442448
97. Holm, L., and Sander, C. (1993) J. Mol. Biol.
233, 123138
98. Lewin, R. (1987) Science 237, 1570
99. Fleming, P. J., and Richards, F. M. (2000) J.
Mol. Biol. 299, 487498
100. Gekko, K., and Hasegawa, Y. (1986)
Biochemistry 25, 65636571
101. Goodsell, D. S., and Olson, A. J. (1993) Trends
Biochem. Sci. 18, 6568
102. Schultz, G. E., and Schirmer, R. H. (1979)
Principles of Protein Structure, Springer-Verlag,
New York
102a. Ramachandran, G. N., Ramakrishnam, C.,
and Sasisekharan, V. (1963) J. Mol. Biol., 7, 95
99
103. Ramachandran, G. N., Venkatachalam, C. M.,
and Krimm, S. (1966) Biophys. J. 6, 849872
103a. Karplus, P. A. (1996) Protein Sci. 5, 1406 1420
104. Sasisekharian, V., Lakshminarayanan, A. V.,
and Ramachandran, G. N. (1967) in Conformation
of Biopolymers, Vol. 2 (Ramachandran, G. N.,
ed), p. 641654, Academic Press, New York
104a. Gunasekaran, K., Ramakrishnan, C., and
Balaram, P. (1996) J. Mol. Biol. 264, 191198
104b. Herzberg, O., and Moult, J. (1991) Proteins 11,
223229
105. Milner-White, E. J., Bell, L. H., and Maccallum,
P. H. (1992) J. Mol. Biol. 228, 725 734
106. Rhee, S. (1990) High resolution X-ray diffraction
studies of cytosolic aspartate aminotransferase
[Ph.D. Dissertation], University of Iowa, Iowa
City, Iowa
107. Dunbrack, R. L., Jr., and Karplus, M. (1993) J.
Mol. Biol. 230, 543574
108. Pauling, L., and Corey, R. B. (1951) Proc. Natl.
Acad. Sci. U.S.A. 37, 729740
109. Rich, A. (1994) Nature (London) 371, 285
110. Chothia, C. (1973) J. Mol. Biol. 75, 295302
111. Chou, K.-C., Nmethy, G., and Scheraga, H.
A. (1983) Biochemistry 22, 62136221
111a. Skarzynski, T., Moody, P. C. E., and Wonacott,
A. J. (1987) J. Mol. Biol. 193, 171187
111b. Wang, L., OConnell, T., Tropsha, A., and
Hermans, J. (1996) J. Mol. Biol. 262, 283293
112. Reeke, G. N., Jr., and Becker, J. W. (1986)
Science 234, 11081111
113. Murzin, A. G., Lesk, A. M., and Chothia, C.
(1994) J. Mol. Biol. 236, 13691381
114. Murzin, A. G., Lesk, A. M., and Chothia, C.
(1994) J. Mol. Biol. 236, 13821400
115. LaLonde, J. M., Bernlohr, D. A., and
Ranaszak, L. J. (1994) FASEB J. 8, 12401241
116. Guss, J. M., and Freeman, H. C. (1983) J. Mol.
Biol. 169, 521563
117. Richardson, J. S. (1981) Adv. Prot. Chem. 34,
167339
117a. Erickson, H. P. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 1011410118
118. Redinbo, M. R., Cascio, D., Choukair, M. K.,
Rice, D., Merchant, S., and Yeates, T. O. (1993)
Biochemistry 32, 1056010567
119. Weiss, M. S., and Schulz, G. E. (1993) J. Mol.
Biol. 231, 817824
120. Kreusch, A., and Schulz, G. E. (1994) J. Mol.
Biol. 243, 891905
121. Yoder, M. D., and Jurnak, F. (1995) FASEB J. 9,
335342
122. Richardson, J. S. (1994) FASEB J. 8, 12371239
123. Steinbacher, S., Seckler, R., Miller, S., Steipe,
B., Huber, R., and Reinemer, P. (1994) Science
265, 383385
124. Richardson, J. S., Getzoff, E. D., and
Richardson, D. C. (1978) Proc. Natl. Acad. Sci.
U.S.A. 75, 25742578
125. Chan, A. W. E., Hutchinson, E. G., Harris, D.,
and Thornton, J. M. (1993) Protein Sci. 2,
15741590
126. Urry, D. W. (1972) Proc. Natl. Acad. Sci. U.S.A.
69, 16101614
127. Nicholson, L. K., and Cross, T. A. (1989)
Biochemistry 28, 93799385
127a. Smith, T. F., Gaitatzes, C., Saxena, K., and Neer,
E. J. (1999) Trends Biochem. Sci. 24, 181185
127b. Garcia-Higuera, I., Fenoglio, J., Li, Y., Lewis,
C., Panchenko, M. P., Reiner, O., Smith, T. F.,
and Neer, E. J. (1996) Biochemistry 35, 13985
13994
127c. ter Naar, E., Harrison, S. C., and Kirchhausen,
T. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 1096
1100
127d. Springer, T. A. (1998) J. Mol. Biol. 283, 837862
128. Pauling, L. (1993) Protein Sci. 2, 10601063
129. Kendrew, J. C. (1969) Sci. Am. 205(Dec), 96110
130. Arnott, S., and Dover, S. D. (1967) J. Mol. Biol.
30, 209212
131. Steigemann, W., and Weber, E. (1979) J. Mol.
Biol. 127, 309338
132. Barlow, D. J., and Thornton, J. M. (1988) J. Mol.
Biol. 201, 601619
132a. Rajashankar, K. R., and Ramakumar, S. (1996)
Protein Sci. 5, 932 946
133. Harper, E. T., and Rose, G. D. (1993)
Biochemistry 32, 76057609
134. Chakrabartty, A., Schellman, J. A., and
Baldwin, R. L. (1991) Nature (London) 351,
586588
135. Creamer, T. P., and Rose, G. D. (1995) Protein
Sci. U.S.A. 4, 13051314
136. Dempsey, C. E. (1992) Biochemistry 31, 4705
4712
137. Ramachandran, G. N., and Mitra, A. K. (1976)
J. Mol. Biol. 107, 8592
138. Blaber, M., Zhang, X.-j, and Matthews, B. W.
(1993) Science 260, 16371639
139. Chakrabartty, A., Kortemme, T., and Baldwin,
R. L. (1994) Protein Sci. 3, 843852
140. Toniolo, C., and Benedetti, E. (1991) Trends
Biochem. Sci. 16, 350353
141. Avbelj, F., and Moult, J. (1995) Biochemistry 34,
755764
142. Karpen, M. E., De Haseth, P. L., and Neet, K.
E. (1992) Protein Sci. 1, 13331342
143. Fiori, W. R., Miick, S. M., and Millhauser, G.
L. (1993) Biochemistry 32, 1195711962
144. Smythe, M. L., Huston, S. E., and Marshall, G.
R. (1993) J. Am. Chem. Soc. 115, 1159411595
145. Bavosa, A., Benedetti, E., Di Blasio, B.,
Pavone, V., Pedone, C., Toniolo, C., and
Bonora, G. M. (1986) Proc. Natl. Acad. Sci.
U.S.A. 83, 19881992
145a. Fathallah-Shaykh, H., Wolf, S., Wong, E.,
Posner, J. B., and Furneaux, H. M. (1991) Proc.
Natl. Acad. Sci. U.S.A 88, 34513454
146. Miick, S. M., Martinez, G. V., Flori, W. R.,
Todd, A. P., and Millhauser, G. L. (1992)
Nature (London) 359, 653655
147. Wada, A. (1976) Adv. Biophys. 9, 163
148. Hol, W. G. J., van Duijnen, P. T., and Berendsen,
H. J. C. (1978) Nature (London) 273, 443446
148a. Hol, W. G. J., Halie, L. M., and Sander, C.
(1981) Nature (London) 294, 532536
148b. Lockhart, D. J., and Kim, P. S. (1992) Science
257, 947951
149. Levine, B. F., and Bethea, C. G. (1976) J. Chem.
Phys. 65, 19891993
150. Gray, T. M., and Matthews, B. W. (1984) J. Mol.
Biol. 175, 7581
151. Richardson, J. S., and Richardson, D. C. (1988)
Science 240, 16481652
88 Chapter 2. Amino Acids, Peptides, and Proteins
References
181. Harbury, P. B., Kim, P. S., and Alber, T. (1994)
Nature (London) 371, 8083
182. Efimov, V. P., Nepluev, I. V., Sobolev, B. N.,
Zurabishvili, T. G., Schulthess, T., Lustig, A.,
Engel, J., Haener, M., Aebi, U., Venyaminov, S.
Y., Potekhin, S. A., and Mesyanzhinov, V. V.
(1994) J. Mol. Biol. 242, 470486
183. Lieberman, M., Tabet, M., and Sasaki, T.
(1994) J. Am. Chem. Soc. 116, 50355044
184. Utani, A., Nomizu, M., Timpl, R., Roller, P. P.,
and Yamada, Y. (1994) J. Biol. Chem. 269,
1916719175
185. Robinson, C. R., and Sligar, S. G. (1993)
Protein Sci. 2, 826837
186. Steif, C., Weber, P., Hinz, H.-J., Flossdorf, J.,
Cesareni, G., and Kokkinidis, M. (1993)
Biochemistry 32, 38673876
187. Redfield, C., Boyd, J., Smith, L. J., Smith, R. A.
G., and Dobson, C. M. (1992) Biochemistry 31,
1043110437
188. Harris, N. L., Presnell, S. R., and Cohen, F. E.
(1994) J. Mol. Biol. 236, 13561368
189. Chou, K.-C., Carlacci, L., Maggiora, G. M.,
Parodi, L. A., and Schulz, M. W. (1992) Protein
Sci. 1, 810827
190. Rees, D. C., Komiya, H., Yeates, T. O., Allen, J.
P., and Feher, G. (1989) Ann. Rev. Biochem. 58,
607633
191. Aleshin, A. E., Hoffman, C., Firsov, L. M., and
Honzatko, R. B. (1994) J. Mol. Biol. 238, 575
591
192. Aleshin, A. E., Firsov, L. M., and Honzatko,
R. B. (1994) J. Biol. Chem. 269, 1563115639
193. Harris, E. M. S., Aleshin, A. E., Firsov, L. M.,
and Honzatko, R. B. (1993) Biochemistry 32,
16181626
194. Bella, J., Eaton, M., Brodsky, B., and Berman,
H. M. (1994) Science 266, 7581
194a. Kramer, R. Z., Bella, J., Mayville, P., Brodsky,
B., and Berman, H. M. (1999) Nature Struct.
Biol. 6, 454 457
195. Brodsky, B., and Shah, N. K. (1995) FASEB J. 9,
15371546
196. Redeker, V., Levilliers, N., Schmitter, J.-M., Le
Caer, J.-P., Rossier, J., Adoutte, A., and Br,
M.-H. (1994) Science 266, 16881691
197. Miller, A. (1982) Trends Biochem. Sci. 7, 1318
198. Eyre, D. R. (1980) Science 207, 13151322
199. Parry, D. A. D., and Craig, A. S. (1979) Nature
(London) 282, 213215
200. Porter, R. R., and Reid, K. B. M. (1979) Adv.
Prot. Chem. 33, 171
201. Ashford, D., and Neuberger, A. (1980) Trends
Biochem. Sci. 5, 245248
202. Chen, J., and Varner, J. E. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 43994403
203. Adzhubei, A. A., and Sternberg, M. J. E. (1994)
Protein Sci. 3, 23952410
204. Richardson, J. S. (1985) Nature (London) 316,
102103
205. Hutchinson, E. G., and Thornton, J. M. (1994)
Protein Sci. 3, 22072216
206. Venkatachalam, C. M. (1968) Biopolymers 6,
14251436
207. Perczel, A., Foxman, B. M., and Fasman, G. D.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89, 82108214
208. Yan, Y., Tropsha, A., Hermans, J., and Erickson,
B. W. (1993) Proc. Natl. Acad. Sci. U.S.A. 90,
78987902
209. Perczel, A., McAllister, M. A., Csszr, P., and
Csizmadia, I. G. (1993) J. Am. Chem. Soc. 115,
48494858
210. Mattos, C., Petsko, G. A., and Karplus, M.
(1994) J. Mol. Biol. 238, 733747
211. Hynes, T. R., Hodel, A., and Fox, R. O. (1994)
Biochemistry 33, 50215030
212. Haque, T. S., Little, J. C., and Gellman, S. H.
(1994) J. Am. Chem. Soc. 116, 41054106
213. Gierasch, L. M., Deber, C. M., Madison, V.,
Niu, C.-H., and Blout, E. R. (1981) Biochemistry
20, 47304738
214. Rees, D. C., Lewis, M., and Lipscomb, W. N.
(1983) J. Mol. Biol. 168, 367387
215. Sapse, A.-M., Mallah-Levy, L., Daniels, S. B.,
and Erickson, B. W. (1987) J. Am. Chem. Soc.
109, 35263529
216. Urry, D. W. (1993) Angew. Chem. Int. Ed. Engl.
32, 819841
217. Sandberg, L. B., Soskel, N. T., and Leslie, J. G.
(1981) N. Engl. J. Med. 304, 566579
217a. Tatham, A. S., and Shewry, P. R. (2000) Trends
Biochem. Sci. 25, 567571
218. Gotte, L., Volpin, D., Horne, R. W., and
Mammi, M. (1976) Micron 7, 95102
218a. Urry, D., Luan, C.-H., and Peng, S. (1995)
Ciba. Found. Symp. 192, 430
218b. van Dijk, A. A., de Boef, E., Bekkers, A., van
Wijk, L. L., van Swieten, E., Hamer, R. J., and
Robillard, G. T. (1997) Protein Sci. 6, 649656
219. Sternberg, M. J. E., and Thornton, J. M. (1976)
J. Mol. Biol. 105, 367382
220. Leszczynski, J. F., and Rose, G. D. (1986)
Science 234, 849855
221. Liang, C., and Mislow, K. (1994) J. Am. Chem.
Soc. 116, 1118911190
222. Chothia, C. (1984) Ann. Rev. Biochem. 53, 537
572
223. Resnick, D., Pearson, A., and Krieger, M.
(1994) Trends Biochem. Sci. 19, 58
224. Shoyab, M., Plowman, G. D., McDonald, V. L.,
Bradley, J. G., and Todaro, G. J. (1989) Science
243, 10741076
225. Doolittle, R. F. (1995) Ann. Rev. Biochem. 64,
287 314
226. Berman, A. L., Kolker, E., and Trifanov, E. N.
(1994) Proc. Natl. Acad. Sci. U.S.A. 91, 40444047
227. Orengo, C. A., Jones, D. T., and Thornton, J.
M. (1994) Nature (London) 372, 631634
228. Pan, K.-M., Damodaran, S., and Greaser, M. L.
(1994) Biochemistry 33, 82558261
229. Doolittle, R. F., and Bork, P. (1993) Sci. Am.
269(Oct), 5056
230. Baron, M., Norman, D. G., and Campbell, I.
D. (1991) Trends Biochem. Sci. 16, 1317
231. Mulichak, A. M., Tulinsky, A., and
Ravichandran, K. G. (1991) Biochemistry 30,
1057610588
232. Bork, P., Holm, L., and Sander, C. (1994) J.
Mol. Biol. 242, 309320
232a. Bogan, A. A., and Thorn, K. S. (1998) J. Mol.
Biol. 280, 19
233. Tsai, J., Taylor, R., Chothia, C., and Gerstein,
M. (1999) J. Mol. Biol. 290, 253266
233a. Gerstein, M., and Chothia, C. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1016710172
234. Varadarajan, R., and Richards, F. M. (1992)
Biochemistry 31, 1231512327
235. Allewell, N. (1987) Trends Biochem. Sci. 12,
417418
236. Lim, W. A., Farruggio, D. C., and Sauer, R. T.
(1992) Biochemistry 31, 43244333
237. Hubbard, S. J., and Argos, P. (1994) Protein Sci.
3, 21942206
237a. Hubbard, S. J., and Argos, P. (1996) J. Mol.
Biol. 261, 289 300
238. Hunt, N. G., Gregoret, L. M., and Cohen, F. E.
(1994) J. Mol. Biol. 241, 214225
239. Hunter, C. A., and Sanders, K. M. (1990) J.
Am. Chem. Soc. 112, 55255534
240. Burley, S. K., and Petsko, G. A. (1985) Science
229, 2328
241. Thomas, K. A., Smith, G. M., Thomas, T. B.,
and Feldmann, R. J. (1982) Proc. Natl. Acad.
Sci. U.S.A. 79, 48434847
242. Flocco, M. M., and Mowbray, S. L. (1994) J.
Mol. Biol. 235, 709717
243. Hemmingsen, J. M., Gernert, K. M.,
Richardson, J. S., and Richardson, D. C. (1994)
Protein Sci. 3, 19271937
244. Teeter, M. M. (1991) Ann. Rev. Biophys.
Biophys. Chem. 20, 577600
245. McDonald, I. K., and Thornton, J. M. (1994) J.
Mol. Biol. 238, 777793
246. Bordo, D., and Argos, P. (1994) J. Mol. Biol.
243, 504519
247. Shimoni, L., and Glusker, J. P. (1995) Protein
Sci. 4, 6574
248. Dao-pin, S., Anderson, D. E., Baase, W. A.,
Dahlquist, F. W., and Matthews, B. W. (1991)
Biochemistry 30, 1152111529
248a. Kumar, S., and Nussinov, R. (1999) J. Mol. Biol.
293, 12411255
248b. Strop, P., and Mayo, S. L. (2000) Biochemistry
39, 12511255
249. Sawyer, L., and James, M. N. G. (1982) Nature
(London) 295, 7980
250. Borders, C. L., Jr., Broadwater, J. A., Bekeny, P.
A., Salmon, J. E., Lee, A. S., Eldridge, A. M.,
and Pett, V. B. (1994) Protein Sci. 3, 541548
251. Mrabet, N. T., Van den Broeck, A., Van den
brande, I., Stanssens, P., Laroche, Y., Lambeir,
A.-M., Matthiissens, G., Jenkins, J., Chiadmi,
M., van Tilbeurgh, H., Rey, F., Janin, J., Quax,
W. J., Lasters, I., De Maeyer, M., and Wodak, S.
J. (1992) Biochemistry 31, 22392253
252. Dougherty, D. A. (1996) Science 271, 163168
253. Mitchell, J. B. O., Nandi, C. L., McDonald, I.
K., Thornton, J. M., and Price, S. (1994) J. Mol.
Biol. 239, 315331
253a. De Wall, S. L., Meadows, E. S., Barbour, L. J.,
and Gokel, G. W. (1999) J. Am. Chem. Soc. 121,
5613 5614
253b. Gallivan, J. P., and Dougherty, D. A. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 9459 9464
253c. Wouters, J. (1998) Protein Sci. 7, 2472 2475
253d. Fernndez-Recio, J., Romero, A., and Sancho,
J. (1999) J. Mol. Biol. 290, 319 330
254. Rose, G. D., Young, W. B., and Gierasch, L. M.
(1983) Nature (London) 304, 654657
255. Watenpaugh, K. D., Margulis, T. N., Sieker, L.
C., and Jensen, L. H. (1978) J. Mol. Biol. 122,
175190
255a. Nakasako, M. (1999) J. Mol. Biol. 289, 547564
256. Teeter, M. M. (1984) Proc. Natl. Acad. Sci.
U.S.A. 81, 60146018
257. Otting, G., Liepinsh, E., and Wthrich, K.
(1991) Science 254, 974980
258. Murzin, A. G., and Finkelstein, A. V. (1988) J.
Mol. Biol. 204, 749770
259. Chothia, C. (1989) Nature (London) 337, 204205
260. Buehner, M., Ford, G. C., Moras, D., Olsen, K.
W., and Rossmann, M. G. (1974) J. Mol. Biol.
90, 2549
261. Rossmann, M. G., Liljas, A., Branden, C.I.,
and Banaszak, L. J. (1975) The Enzymes, 3rd ed.,
Vol. 11, Academic Press, New York (p. 61)
262. Brndn, C. (1990) Nature (London) 346, 607608
263. Kobe, B., and Deisenhofer, J. (1995) Nature
(London) 374, 183186
264. Kobe, B., and Deisenhofer, J. (1994) Trends
Biochem. Sci. 19, 415421
264a. Kajava, A. V. (1998) J. Mol. Biol. 277, 519 527
264b. Kobe, B., and Kajava, A. V. (2000) Trends
Biochem. Sci. 25, 509515
265. Farber, G. K., and Petsko, G. A. (1990) Trends
Biochem. Sci. 15, 228234
266. Reardon, D., and Farber, G. K. (1995) FASEB J.
9, 497503
267. Uhlin, U., and Eklund, H. (1994) Nature
(London) 370, 533539
268. Chothia, C. (1992) Nature (London) 357, 543544
268a. Zhang, C., and DeLisi, C. (1998) J. Mol. Biol.
284, 13011305
269. McLachlan, A. D. (1980) Nature (London) 285,
267268

89
References
270. Chou, P. Y., and Fasman, G. D. (1974)
Biochemistry 13, 222245
270a. Myers, J. K., Pace, C. N., and Scholtz, J. M.
(1997) Biochemistry 36, 10923 10929
270b. Spek, E. J., Olson, C. A., Shi, Z., and
Kallenbach, N. R. (1999) J. Am. Chem. Soc. 121,
5571 5572
271. Minor, D. L., Jr., and Kim, P. S. (1994) Nature
(London) 367, 660663
272. Richards, F. M. (1991) Sci. Am. 264(Jan), 5463
273. Anfinsen, C. B. (1973) Science 181, 223230
274. Dill, K. A., Bromberg, S., Yue, K., Fiebig, K.
M., Yee, D. P., Thomas, P. D., and Chan, H. S.
(1995) Protein Sci. 4, 561 602
275. Rost, B., Schneider, R., and Sander, C. (1993)
Trends Biochem. Sci. 18, 120123
276. Wako, H., and Blundell, T. L. (1994) J. Mol.
Biol. 238, 693708
277. Yi, T.-M., and Lander, E. S. (1993) J. Mol. Biol.
232, 11171129
278. Monge, A., Friesner, R. A., and Honig, B. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 50275029
279. Smith, C. K., Withka, J. M., and Regan, L.
(1994) Biochemistry 33, 55105517
279a. Lacroix, E., Viguera, A. R., and Serrano, L.
(1998) J. Mol. Biol. 284, 173 191
279b. Gotoh, O. (1996) J. Mol. Biol. 264, 823 838
279c. Dandekar, T., and Argos, P. (1996) J. Mol. Biol.
256, 645 660
279d. Jones, D. T. (1999) J. Mol. Biol. 292, 195 202
280. Kyte, J., and Doolittle, R. F. (1982) J. Mol. Biol.
157, 105132
281. Reyes, V. E., Phillips, L., Humphreys, R. E.,
and Lew, R. A. (1989) J. Biol. Chem. 264,
1285412858
282. Wold, F. (1986) Trends Biochem. Sci. 11, 5859
283. Graves, D. J., Martin, B. L., and Wang, J. H.
(1994) Co-and post-translational modification of
proteins, Oxford Univ. Press, New York
283a. Angeletti, R. H., ed. (1998) Protein, Analysis
and Design, Academic Press, San Diego,
California
284. Yan, S. C. B., Grinnell, B. W., and Wold, F.
(1989) Trends Biochem. Sci. 14, 264268
285. Berthiaume, L., Deichaite, I., Peseckis, S., and
Resh, M. D. (1994) J. Biol. Chem. 269, 64986505
286. McIlhinney, R. A. J. (1990) Trends Biochem. Sci.
15, 387391
287. Violand, B. N., Schlittler, M. R., Lawson, C.
Q., Kane, J. F., Siegel, N. R., Smith, C. E.,
Kolodziej, E. W., and Duffin, K. L. (1994)
Protein Sci. 3, 10891097
288. Hilz, H., Fanick, N., and Klapproth, K. (1986)
Proc. Natl. Acad. Sci. U.S.A. 83, 62676271
289. Hortin, G., Fok, K. F., Toren, P. C., and Strauss,
A. W. (1987) J. Biol. Chem. 262, 30823085
290. Bateman, A., Solomon, S., and Bennett, H. P. J.
(1990) J. Biol. Chem. 265, 2213022136
291. Takahara, H., Tsuchida, M., Kusubata, M.,
Akutsu, K., Tagami, S., and Sugawara, K.
(1989) J. Biol. Chem. 264, 1336113368
292. Kreil, G. (1994) J. Biol. Chem. 269, 1096710970
293. Mor, A., Amiche, M., and Nicolas, P. (1992)
Trends Biochem. Sci. 17, 481485
294. Kreil, G. (1994) Science 266, 996997
295. Prescott, B., Renugopalakrishnan, V.,
Glimcher, M. J., Bhushan, A., and Thomas, G.
J., Jr. (1986) Biochemistry 25, 27922798
296. Wood, D. D., and Moscarello, M. A. (1989) J.
Biol. Chem. 264, 51215127
297. Waite, J. H., Housley, T. J., and Tanzer, M. L.
(1985) Biochemistry 24, 50105014
298. Taylor, S. W., Waite, J. H., Ross, M. M.,
Shabanowitz, J., and Hunt, D. F. (1994) J. Am.
Chem. Soc. 116, 1080310804
299. Clarke, S. (1992) Ann. Rev. Biochem. 61, 355386
300. Marshall, C. J. (1993) Science 259, 18651866
301. Mroz, E. A. (1989) Science 243, 1615
302. Rothgeb, T. M., England, R. D., Jones, B. N.,
and Gurd, R. S. (1978) Biochemistry 17, 4564
4571
303. Lorimer, G. H. (1983) Trends Biochem. Sci. 8,
6568
304. Friedman, M. (1977) Protein Crosslinking, Vol. 2,
Plenum, New York
305. Torchinsky, Y. M. (1981) Sulfur in Proteins,
Pergamon, Oxford
306. Harrison, P. M., and Sternberg, M. J. E. (1994)
J. Mol. Biol. 244, 448463
307. Folk, J. E., and Finlayson, J. S. (1977) Adv. Prot.
Chem. 31, 1133
308. Gurd, F. R. N., and Rothgeb, T. M. (1979) Adv.
Prot. Chem. 33, 73165
309. Karplus, M., and McCammon, J. A. (1983)
Ann. Rev. Biochem. 53, 263300
310. Caspar, D. L. D., Clarage, J., Salunke, D. M.,
and Clarage, M. (1988) Nature (London) 332,
659662
311. Smith, J. L., Hendrickson, W. A., Honzatko,
R. B., and Sheriff, S. (1986) Biochemistry 25,
50185027
312. Frauenfelder, H., Sligar, S. G., and Wolynes,
P. G. (1991) Science 254, 15981603
313. Buck, M., Radford, S. E., and Dobson, C. M.
(1994) J. Mol. Biol. 237, 247254
314. Kim, K.-S., Fuchs, J. A., and Woodward, C. K.
(1993) Biochemistry 32, 96009608
315. Rohl, C. A., and Baldwin, R. L. (1994)
Biochemistry 33, 77607767
316. Spyracopoulos, L., and ONeil, J. D. J. (1994)
J. Am. Chem. Soc. 116, 13951402
317. Huber, R. (1979) Trends Biochem. Sci. 4, 271276
318. Lumry, R., and Rosenberg, A. (1974) Colloques
Internationaux du C.N.R.S. 246, 53
319. Mao, B., Pear, M. R., McCammon, J. A., and
Northrup, S. H. (1981) J. Mol. Biol. 151, 199202
320. Brack, A., and Spach, G. (1981) J. Am. Chem.
Soc. 103, 63196323
321. Frauenfelder, H., Parak, F., and Young, R. D.
(1988) Ann. Rev. Biophys. Biophys. Chem. 17,
451479
322. Gerstein, M., Lesk, A. M., and Chothia, C.
(1994) Biochemistry 33, 67396749
323. Kretsinger, R. H., and Nockolds, C. E. (1973)
J. Biol. Chem. 248, 33133326
324. Liepinsh, E., and Otting, G. (1994) J. Am.
Chem. Soc. 116, 96709674
325. Scholtz, J. M., Barrick, D., York, E. J., Stewart,
J. M., and Baldwin, R. L. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 185189
326. Kamoun, P. P. (1988) Trends Biochem. Sci. 13,
424425
327. Dill, K. A., and Shortle, D. (1991) Ann. Rev.
Biochem. 60, 795825
328. Baker, D., and Agard, D. A. (1994) Biochemistry
33, 75057509
328a. Sharp, K. A., and Englander, S. W. (1994)
Trends Biochem. Sci. 19, 526 529
328b. Pace, C. N., Shirley, B. A., McNutt, M., and
Gajiwala, K. (1996) FASEB J. 10, 75 83
328c. Sippl, M. J. (1996) J. Mol. Biol. 260, 644 648
328d. Meyer, E. (1992) Protein Sci. 1, 1543 1562
328e. Hill, R. B., Hong, J.-K., and DeGrado, W. F.
(2000) J. Am. Chem. Soc. 122, 746 747
329. Levinthal, C. (1968) J. Chim. Phys.-Chim. Biol.
65, 4445
330. Lattman, E. E., Fiebig, K. M., and Dill, K. A.
(1994) Biochemistry 33, 61586166
331. Mark, A. E., and van Gunsteren, W. F. (1992)
Biochemistry 31, 77457748
332. Peng, Z., and Kim, P. S. (1994) Biochemistry 33,
21362141
333. Hagihara, Y., Tan, Y., and Goto, Y. (1994) J. Mol.
Biol. 237, 336348
334. Yu, Y., Makhatadze, G. I., Pace, C. N., and
Privalov, P. L. (1994) Biochemistry 33, 33123319
335. Dobson, C. M., Evans, P. A., and Radford, S.
E. (1994) Trends Biochem. Sci. 19, 3137
336. Yang, A.-S., and Honig, B. (1994) J. Mol. Biol.
237, 602614
336a. Baker, D. (2000) Nature (London) 405, 39 42
336b. Dinner, A. R., Sali, A., Smith, L. J., Dobson, C.
M., and Karplus, M. (2000) Trends Biochem. Sci.
25, 331339
337. Udgaonkar, J. B., and Baldwin, R. L. (1988)
Nature (London) 335, 694699
338. Roder, H., Elve, G. A., and Englander, S. W.
(1988) Nature (London) 335, 700704
339. Gething, M.-J., and Sambrook, J. (1992) Science
355, 3345
340. Odefey, C., Mayr, L. M., and Schmid, F. X.
(1995) J. Mol. Biol. 245, 69 78
341. Chazin, W. J., Krdel, J., Drakenberg, T.,
Thulin, E., Brodin, P., Grundstrm, T., and
Forsn, S. (1989) Proc. Natl. Acad. Sci. U.S.A.
86, 21952198
342. Lin, L.-N., and Brandts, J. F. (1987) Biochemistry
26, 35373543
343. Herning, T., Yutani, K., Taniyama, Y., and
Kikuchi, M. (1991) Biochemistry 30, 98829891
344. Chatrenet, B., and Chang, J.-Y. (1993) J. Biol.
Chem. 268, 2098820996
345. Talluri, S., Rothwarf, D. M., and Scheraga, H.
A. (1994) Biochemistry 33, 1043710449
346. Pace, C. N. (1990) Trends Biochem. Sci. 15, 1417
347. Yang, A.-S., and Honig, B. (1993) J. Mol. Biol.
231, 459474
348. Hartsough, D. S., and Merz, K. M., Jr. (1993)
J. Am. Chem. Soc. 115, 65296537
349. Aswad, D. W., and Johnson, B. A. (1987)
Trends Biochem. Sci. 12, 155158
350. Brennan, T. V., and Clarke, S. (1993) Protein
Sci. 2, 331338
351. Volkin, D. B., and Klibanov, A. M. (1987)
J. Biol. Chem. 262, 29452950
352. Wells-Knecht, M. C., Huggins, T. G., Dyer, D.
G., Thorpe, S. R., and Baynes, J. W. (1993)
J. Biol. Chem. 268, 1234812352
353. Luthra, M., Ranganathan, D., Ranganathan,
S., and Balasubramanian, D. (1994) J. Biol.
Chem. 269, 2267822682
354. Lubec, G., Weninger, M., and Anderson, S. R.
(1994) FASEB J. 8, 11661169
355. Kaiser, E. T. (1987) Trends Biochem. Sci. 12,
305309
355a. Beasley, J. R., and Hecht, M. H. (1997) J. Biol.
Chem. 272, 2031 2034
356. Oxender, D. E., and Fox, C. F., eds. (1987)
Protein Engineering, Liss, New York
356a. Dahiyat, B. I., and Mayo, S. L. (1997) Science
278, 82 87
356b. DeGrado, W. F., Summa, C. M., Pavone, V.,
Nastri, F., and Lombardi, A. (1999) Ann. Rev.
Biochem. 68, 779819
357. Bryson, J. W., Betz, S. F., Lu, H. S., Suich, D. J.,
Zhou, H. X., ONeil, K. T., and DeGrado, W. F.
(1995) Science 270, 935941
358. Nautiyal, S., Woolfson, D. N., King, D. S., and
Alber, T. (1995) Biochemistry 34, 11645 11651
359. Betz, S. F., Liebman, P. A., and DeGrado, W. F.
(1997) Biochemistry 36, 2450 2458
360. Handel, T. M., Williams, S. A., and DeGrado,
W. F. (1993) Science 261, 879885
361. Struthers, M. D., Cheng, R. P., and Imperiali,
B. (1996) Science 271, 342345
362. Yan, Y., and Erickson, B. W. (1994) Protein Sci.
3, 10691073
363. Quinn, T. P., Tweedy, N. B., Williams, R. W.,
Richardson, J. S., and Richardson, D. C. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 87478751
364. Hecht, M. H. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 87298730
364a. Kortemme, T., Ramrez-Alvarado, M., and
Serrano, L. (1998) Science 281, 253256
90 Chapter 2. Amino Acids, Peptides, and Proteins
365. Kellis, J. T., Jr., Nyberg, K., and Fersht, A. R.
(1989) Biochemistry 28, 49144922
366. Goldenberg, D. P. (1988) Ann. Rev. Biophys.
Biophys. Chem. 17, 481507
367. Alber, T. (1989) Ann. Rev. Biochem. 58, 765798
368. Katz, B. A., and Kossiakoff, A. (1986) J. Biol.
Chem. 261, 1548015485
369. Pantoliano, M. W., Ladner, R. C., Bryan, P. N.,
Rollence, M. L., Wood, J. F., and Poulos, T. L.
(1987) Biochemistry 26, 20772082
370. Wetzel, R. (1987) Trends Biochem. Sci. 12, 478
482
371. Mitchinson, C., and Wells, J. A. (1989)
Biochemistry 28, 48074815
372. Ahern, T. J., Casal, J. I., Petsko, G. A., and
Klibanov, A. M. (1987) Proc. Natl. Acad. Sci.
U.S.A. 84, 675679
373. Estell, D. A., Graycar, T. P., Miller, J. V.,
Powers, D. B., Burnier, J. P., Ng, P. G., and
Wells, J. A. (1986) Science 233, 659663
374. Loeb, D. D., Swanstrom, R., Everitt, L.,
Manchester, M., Stamper, S. E., and
Hutchison, C. A., III. (1989) Nature (London)
340, 397440
375. Mendel, D., Ellman, J. A., Chang, Z., Veenstra,
D. L., Kollman, P. A., and Schultz, P. G. (1992)
Science 266, 17981802
376. Brady, L., and Dodson, G. (1994) Nature
(London) 368, 692694
377. Liu, N., Deillon, C., Klauser, S., Gutte, B., and
Thomas, R. M. (1998) Protein Sci. 7, 12141220
References
91
Study Questions
1. Name all of the isometric tripeptides which could
be formed from one molecule each of tyrosine,
alanine, and valine.
2. What functional groups are found in protein side
chains? Of what importance to protein structure
and function are (a) hydrophobic groups, (b)
acidic and basic groups, (c) sulfyhydryl groups?
3. If placed in water and adjusted to a pH of 7, will
the following migrate toward the anode or the
cathode if placed in an electrical field?
(a) Aspartic acid, (b) alanine, (c) tyrosine, (d)
lysine, (e) arginine, (f) glutamine.
4. Draw the following hydrogen-bonded structures:
(a) A dimer of acetic acid.
(b) A tyrosinecarboxylate bond in the interior of
a protein.
(c) A phosphateguanidinium ion pair in an
enzymesubstrate complex.
5. Contrast the properties of the amino acids with
those of the saturated fatty acids with respect to
solubility in water and in ether and to physical
state.
6. Describe in as much detail as you can the charac-
teristic properties of (a) sheets, (b) helices, (c)
turns in peptide chains, and (d) collagen.
7. Predict whether the following peptide segments
will be likely to exist as an helix or as part of a
structure within a protein:
(a) Poly-l-leucine
(b) Poly-l-valine
(c) Pro-Glu-Met-Val-Phe-Asp-Ile
(d) Pro-Glu-Ala-Leu-Phe-Ala-Ala
8. Describe three ways in which a side chain of a
serine residue can fold back and hydrogen bond to
a C=O or NH group of the backbone and two
ways by which an asparagine side chain can do
the same. There are yet other possibilities.
9. Compare structural features and properties of the
following proteins: silk fibroin, -keratin, col-
lagen, and bovine serum albumin.
10. In what way do the solubilities of proteins usually
vary with pH? Why?
11. Compare the following: the diameters of (a) a
carbon atom in an organic molecule (b) a bacterial
cell, e.g. of E. coli (c) a human red blood cell (d) a
ribosome (e) the length of a peptide unit in an
extended polypeptide chain (f) the length of the
carbon atom chain in an 18-carbon fatty acid.
12. Compare: (a) the length of a peptide unit (residue)
in a polypeptide in an extented () conformation.
(b) the length by which an helix is extended by
the addition of one amino acid unit (c) the length
of one turn of an helix. (d) the diameter of an
helix (both using atom centers in the backbones
and using van der Waals radii) for a poly-l-alanine
helix.
13. What are: albumins, globulins, protamines,
scleroproteins, glycoproteins, lipoproteins?
14. Where are the following found and what are their
functions? Gamma globulin, hemocyanin, pepsin,
glucagon, ferritin, phosphorylase.
15. List the nutritionally essential amino acids for
human beings. Compare these needs with those
of other species, including lactic acid bacteria,
malaria parasites, green plants, etc.
16. Define: chiral, enantomer, diastereomer, epimer,
anomer (see Chapter 4), prochiral (see Chapter 9).
What is meant by the statement that biochemical
reactions are stereochemically specific? Why is
such stereospecificity to be expected in organisms
(which are constructed from asymmetric units)?
See Chapter 9 for further discussion.
17. What are disulfide bridges and of what signifi-
cance are they in protein structure?
18. What is meant by denaturation of a protein?
Mention several ways in which denaturation can
be brought about. How is denaturation explained
in terms of structure?
19. A chain of l-amino acids can form either a right-
handed or a left-handed helix. From the
Ramachandran diagram in Fig. 2-9, can you say
anything about the relative stabilities of right and
left-handed helices? What do you predict for
polyglycine?
20. What similarities and differences would you
predict for two proteins of identical amino acid
sequence but one made from all l-amino acids and
the other from all d-amino acids?
92 Chapter 2. Amino Acids, Peptides, and Proteins
21. Complete the following peptide structure for l-
seryl-l-valyl-l-asparaginyletc. Extend the chain
in the C-terminal direction to form a beta turn
with the chain coming back to form a beta sheet.
Add a third segment of peptide parallel to the
folded back chain to form a 3-stranded beta sheet.
Indicate all hydrogen bonds correctly. Draw the
side chains of the seryl and asparaginyl residues in
positions 1 and 3 so that they form proper hydro-
gen bonds to groups in the peptide backbone.
22. Complete the following structure to form a short
segment of alpha helix. Extend the chain by at
least three residues. Form all hydrogen bonds
correctly. Add at least three side chains with the
correct chirality at the alpha carbon positions.
Add one electrically charged side chain and show
how it interacts with the peptide backbone at
either the C- or N-terminus to help stabilize the
helix.
23. What is the relationship between ngstrom units
() and nanometers (nm)? Give the indicated
distance in or nm.
Study Questions
CH
2
Phenylanine
side chain
C
C
C
C
CH
2
van der
Waals
surface
H
2
C
H
2
C
CH
2
Lysine
side chain
C
H
H
H
H
H
N
+
N
H
O
C
H
H
H
C
C
H
H
N
C
N
H O
C
H
H
C
H
O
H
van der Waals surface
H
3
N
N
N
H
H
+
1
2
3
N
N
C
C
C
N
C
C
C
N
C
C
C
N
93
Study Questions
H
3
N
N
N
O
H O
H O
N
N
N
H
O H
O
+
Polypeptide
1
H
3
O
O
1
2
3
2
Retro-inverso
polypeptide

24. Draw three residues of a polypeptide chain


constructed of l-amino acids using the top tem-
plate of the two below. Now, using the second
template, whose polypeptide chain begins at the
right, use the same three residues, numbering from
the C-terminus using d-amino acids. This is know
as a retro-inverso polypeptide.
Compare the outer surface of of the standard
polypeptide and its retro-inverso analog. How
would your answer be affected by the presence of
threonine or isoleucine in the peptides?
If the two polypeptides were folded into a hairpin
loop of structure how would the exterior sur-
faces compare? How would the hydrogen bond-
ing compare?
What possible value can you imagine for retro-
inverso polypeptides in design of drugs? See
Brady and Dodson, Nature 368, 692694 (1994) or
Guichard et al., Proc. Natl. Acad. Sci. USA 91, 9765
9769 (1994)
94 Chapter 3. Determining Structures and Analyzing Cells
Contents
G. Microscopy
H. X-ray and Neutron Diffraction
I. Nuclear Magnetic Resonance (NMR)
1. Basic Principles of NMR Spectroscopy
Band widths
The chemical shift
Scalar coupling (J coupling)
2. Nuclei Other Than Hydrogen
Deuterium (2H)
Carbon - 13
Nitrogen - 15
Phosphorus - 31
Fluorine - 19
Some Other Nuclei
3. Fourier Transform Spectrometers and
Two-Dimensional NMR
Free induction decay
Relaxation times T1 and T2
Two-dimensional and multidimensional
NMR spectra
4. Three-Dimensional Structures and Dynamics
of Proteins
Assignment of resonances
NOEs and distance constraints
5. Other Information from NMR Spectra
NMR titrations
Observing exchangable protons
Exchange rates of amide protons
Solid-state NMR and other topics
J. The Protein Data Bank, Three-Dimensional
Structures, and Computation
References
Study Questions
A. Understanding pH and Electrical Charges on
Macromolecules
1. Strengths of Acids and Bases: the pKa's
2. Titration Curves
3. Buffers
B. Isolating Compounds
1. Fractionation of Cells and Tissues
2. Separations Based on Molecular Size, Shape, and
Density
Dialysis, ultrafiltration, and perfusion
chromatography
Centrifugation
3. Separations Based on Solubility
4. Separation by Partition
5. Ion Exchange Chromatography
6. Affinity Chromatography
7. Electrophoresis and Isoelectric Focusing
C. Determining the Relative Molecular Mass, M
r
1. Ultracentrifugation
Sedimentation velocity
2. Gel Filtration and Gel Electrophoresis
3. Mass Spectrometry
D. Determining Amino Acid Composition and
Sequence
1. Cleavage of Disulfide Bridges
2. Hydrolysis and Other Chain Cleavage Reactions
Selective enzymatic hydrolysis
Nonenzymatic cleavages
Separating the peptides
3. Determining Amino Acid Sequence
The Edman degradation
Protein sequences from the genes
Mass spectrometry in sequencing
4. Locating Disulfide Bridges
5. Detecting Products
6. Absorption of Light
E. Quantitative Determinations and Modification
Reactions of Side Chain Groups
1. Reactions of Amino Groups
2. Reactions of SH Groups
3. Reactions of Other Side Chains
4. Affinity Labeling
F. Synthesis of Peptides
1. Solid-Phase Peptide Synthesis
2. Combinatorial Libraries
95
95
96
97
98
98
100
100
100
101
102
103
104
106
108
108
108
112
112
115
115
116
116
117
118
118
118
119
119
119
120
122
123
123
125
126
127
127
127
128
102
107
110
116
121
122
99
117
141
149
Boxes
Box 3-A Quantitative Estimation of Protein
Concentrations
Box 3-B Sorting and Analyzing Single Cells
Box 3-C Isotopes in Biochemical Investigations
Box 3-D The Proteome
Box 3-E Ninhydrin
Box 3-F Biosensors and Electronic Noses
Tables
Table 3-1 Practical pK
a
Values for Some Useful Buffer
Compounds at 25C and Ionic Strength 0.1
Table 3-2 Specifities of Commonly Used Protein-
Hydrolyzing Enzymes
Table 3-3 Approximate Chemical Shift Ranges in
1
H- and in
15
N- NMR Spectra
Table 3-4 Selected World Wide Web Servers
Related to Protein Structures and
Sequences
129
132
137
137
138
138
139
140
140
140
140
140
140
141
141
141
141
142
145
145
145
147
147
148
148
149
149
150
157
A Laue X-ray diffraction pattern from a protein crystal. A stationary
crystal is irradiated with very intense white, multiwavelength X rays
from a synchrotron source. The diffraction pattern is rich in infor-
mation. A single 0.1 ms X-ray pulse may provide a pattern with
enough information to determine a three-dimensional structure.
The pattern consists of thousands of diffraction spots arranged on
intersecting rings. The coordinates of the diffraction spots together
with their measured intensities provide the necessary information
for structure determination. Courtesy of Louise Johnson.
95
3
How have chemists deduced the thousands of
structural formulas that we write for the substances
found in nature? The answer is far too complex to
give here in detail. However, the separation of com-
pounds, the analysis of mixtures, and the unraveling
of structures remain essential parts of biochemistry.
A minireview of methods, with emphasis on proteins,
follows. Additional procedures having to do primarily
with carbohydrates, nucleic acids, or lipids are given
in Chapters 4, 5, and 8, respectively.
A. Understanding pH and Electrical Charges
on Macromolecules
Proteins, nucleic acids, and carbohydrates all con-
tain acidic or basic functional groups. The strengths
of the acidic groups vary over a broad range from that
of the strongly acidic phosphate and sulfate esters to
that of the very weakly acidic alcoholic OH group.
The net electrical charge, as well as the spatial distri-
bution of the charged groups, affects the properties of
a macromolecule greatly. Therefore, it will be worth-
while for us to consider some aspects of acidbase
chemistry before discussing other topics.
1. Strengths of Acids and Bases: the pK
a
s
The strength of an acid is usually described by the
acid dissociation constant K
a
HA = A

+ H
+
K
a
= [A

][H
+
] / [HA]
Determining Structures and Analyzing Cells
(3-1)
(3-2)
For strong acids K
a
is high and for weak acids it is low.
Since the values of K
a
vary by many orders of magni-
tude it is customary to use as a measure of the acid
strength pK
a.
This is the negative logarithm of K
a
(pK
a
= log K
a
) . For very strong acids pK
a
is less than zero,
while very weak acids have pK
a
values as high as 15 or
more.
In the biochemical literature the strength of a base is
nearly always given by the pK
a
of the conjugate acid. Thus,
A

in Eq. 3-1 is a base and HA its conjugate acid. The


base could equally well be uncharged A and its conju-
gate acid HA
+
. In both cases Eq. 3-2 would hold. This
defines the strength of both the acid HA and the base
A

. It follows that strong bases have weak conjugate


acids with high pK
a
values, while weak bases have
strong conjugate acids with low pK
a
values.
For a compound containing several acidic groups
we define a series of consecutive dissociation constants
K
1a
, K
2a
, K
3a
, etc. For the sake of simplicity we will omit
the as and call these K
1
, K
2
, K
3
,
...
.
If there are n consecutive dissociation constants there
will be n + 1 ionic species H
3
A, H
2
A, etc. Notice that
H
3
A could be a neutral molecule with H
2
A, HA, and A
bearing changes of 1, 2, and 3, respectively. Alter-
natively, H
3
A might carry 1, 2, or 3 positive changes.
In every case the mathematical expressions will be the
same. For this reason the charges have been deliber-
ately omitted from Eq. 3-3 and others that follow.
Each constant in Eq. 3-3 is defined as in Eq. 3-2, i.e.,
K
2
= [HA][H
+
] / [H
2
A], etc. Keep in mind that there
K
1
H
3
A
K
2
H
2
A
K
3
HA A (3-3)
96 Chapter 3. Determining Structures and Analyzing Cells
(3-4)
(3-5)
are significant differences between the apparent equi-
librium constants (concentration equilibrium constants)
that we ordinarily use and thermodynamic equilibri-
um constants that are obtained by extrapolation to
zero ionic strength.
1
A related complication is the
uncertainity associated with the measurement of pH.
This is often considered a measurement of hydrogen
ion activity but this is not a correct statement. (The
matter is considered briefly in Chapter 6). However,
for all practical purposes, in the range of about pH
410 the pH can be equated with log [H
+
].
Often only one of the ionic forms of Eq. 3-3 will be
important in a biochemical reaction. A particular ionic
species may be the substrate for an enzyme. Likewise,
an enzymesubstrate complex in only a certain state
of protonation may react to given products. In these
cases, and whenever pH affects an equilibrium, it is
useful to relate the concentration [A
i
] of a given ionic
form of a compound to the total of all ionic forms [A]
t
using Eqs. 3-4 and 3-5.
[A
i
] = [A]
t
/ F
i
[A]
t
= [A] + [HA] + [H
2
A] +
...
[H
n
A]
For Eq. 3-4, A
1
= H
n
A, A
2
= H
n-1
A, etc. and F
1
, F
2
,
etc. are the Michaelis pH functions,
2,3
which were proposed
by L. Michaelis in 1914. For the case represented by
Eq. 3-3 there are four ionic species and therefore four
Michaelis pH functions which have the following form
(Eq. 3 - 6). Here, K
1
, K
2
, etc. are the usual consecutive
acid dissociation constants.
F
1
= 1 + K
1
/ [H
+
] + K
1
K
2
/ [H
+
]
2
+ K
1
K
2
K
3
/ [H
+
]
3
F
2
= [H
+
]/K
1
+ 1 + K
2
/ [H
+
] + K
2
K
3
/ [H
+
]
2
F
3
= [H
+
]
2
/K
1
K
2
+ [H
+
]/K
2
+ 1 + K
3
/ [H
+
]
F
4
= [H
+
]
3
/K
1
K
2
K
3
+ [H
+
]
2
/K
2
K
3 +
[H
+
]/K
3
+ 1
If there are only three ionic forms the first three of
these equations will apply if the final term is dropped
from each. The student should be able to verify these
equations and to write the appropriate pH functions
for other cases. Since these relationships are met so
often in biochemistry it is worthwhile to program a
computer to evaluate the Michaelis pH functions and
to apply them as needed. From Eq. 3-4 it can be seen
that the reciprocal of the Michaelis pH function for a given
ionic form represents the fraction of the total compound in
that form and that the sum of these reciprocals for all
the ionic forms is equal to one. Examples of the use
of the Michaelis pH functions in this book are given in
Eq. 6-50, which relates the Gibbs energy of hydrolysis
of ATP to the pH, and in Eqs. 9-55 to 9-57, which de-
scribe the pH dependence of enzymatic action.
Notice that in Eq. 3-3 single arrows have been
used rather than the pairs ( ) that are often em-
ployed to indicate reversible equilibria. This is done
so that the direction of the arrow indicates whether we are
using a dissociation constant or an association constant.
The use of single arrows in this manner to indicate
how the equilibrium constants are to be written is a
good practice when dealing with complex equilibria.
2. Titration Curves
When a neutral amino acid is titrated with acid,
the carboxylate groups become protonated and acid is
taken up. Likewise titration with base removes protons
from the protonated amino groups and base is taken
up. If we plot the number of equivalents of acid or
base that have reacted with the neutral amino acid
versus pH, a titration curve such as that shown in Fig.
3-1 is generated. The curve for histidine contains three
steps; the first corresponds to the titration of the car-
boxylate group with acid, the second to the titration
of the protonated imidazole of the side chain, and the
third to the titration of the protonated amino group
with base. Each step is characterized by a midpoint
that is equal to the pK
a
value for the group being
titrated. The ends of the curve at low and high pH,
which are drawn with a dashed line, are obtained only
after corrections have been applied to the data. If only
the equivalents of acid or base added rather than the
number reacted are plotted, we obtain the solid line
shown in Fig. 3-1. We see that at the low pH end there
is no distinct end point. As we add more acid to try to
complete the titration, the correction that must be
applied to the data becomes increasingly greater. The
difference between the dashed and solid lines reflects
the fact that at the low pH end much of the acid added
is used to simply lower the pH. Therefore, we have a
large free [H
+
]. Similarly, at the high pH end we have
a high free [OH

].
The exact shapes of the ends of the titration curve
depend heavily on the total concentration. Likewise,
the magnitude of the correction required to obtain a
plot of equivalents of acid or base reacted varies with
the concentration and is smaller the higher the concen-
tration of the substance being titrated (see problems 2
and 3 at the end of this chapter). An important rule in
doing acid-base titrations, especially when very small
samples are available, is to use the highest possible
concentration of sample in the smallest possible volume
and to titrate with relatively concentrated acid or base.
Because of the difficulty of adequately correcting titra-
tion curves at low pH it is hard to estimate the pK
a
values
of the carboxyl groups of amino acids accurately from
titration. An additional experimental difficulty exists
at the high pH end because of the tendency for basic
(3-6)
97
6.0
9.2
0 2 4 6 8 10 12 14
pH
1
0
1
2
N
a
O
H
H
C
l
E
q
u
i
v
a
l
e
n
t
s

a
d
d
e
d

p
e
r

m
o
l
e

o
f

h
i
s
t
i
d
i
n
e
1.8
N
N
H3N
H
COOH
+
H
H
+
N
N
H3N
H
COO

+
H
H
+
N
N
H3N
H
COO

+
H
N
N
H2N
H
COO

H
Figure 3-1 Titration curve for
histidine. The solid line represents
the uncorrected titration curve for
3 mM histidine monohydrochloride
titrated with 0.2 M HCL to lower
pH or with 0.2 M NaOH to higher
pH assuming pK
a
values of 1.82,
6.00, and 9.17. The dashed line
represents the corrected curve
showing the number of protons
bound or lost per mole of histidine
monohydrochloride.
solutions to absorb CO
2
from the air. Sometimes for-
maldehyde is added to shift the apparent pK
a
of the
amino groups to lower values and make the titration
more satisfactory.
Despite the difficulties, real proteins can be titrated
successfully to estimate the numbers of carboxyl, histi-
dine, tyrosine, and other groups.
4
An example is
shown in Fig. 3-2. The experimental data have been
fitted with a theoretical curve based on the pK
a
s of
the carboxyl, histidine, tyrosine, and amino groups as
determined by NMR measurements.
5
Account has
been taken of the effect of the electrical field created by
the many charged groups in distorting the curve from
that obtained by summing the theoretical titration
curves of the component groups. However, when there
are multiple acidbase groups that are close together
in a protein, a more complex situation involving tauto-
merism arises. This is discussed in Chapter 6.
Titration curves based on plots of light absorption
versus pH or of NMR chemical shifts versus pH (see
Fig. 3-29) are often useful. They have the important
advantage that no special correction for free acid or
base is needed at low or high pH.
3. Buffers
A mixture of a weak acid HA and of its conjugate
base A constitutes a buffer which resists changes in
pH. This can be seen most readily by taking logarithms
of both sides of Eq. 3-2. By replacing log K with pK
a
and log [H
+
] with pH and rearranging we obtain Eq.
3-7 (the HendersonHasselbalch equation). It is some-
times useful to rewrite this as Eq. 3-8, where is the
fraction of the acid that is dissociated at a given pH.
Logarithms to the base 10 are used in both equations.
These equations are useful in preparing buffers and in
thinking about what fraction of a substance exists in a
given ionic form at a particular value of pH. From Eq.
3-7 it is easy to show that when the pH is near the pK
a
relatively large amounts of acid or base must be added
to change the pH if the concentrations of the buffer
pair A and HA are high.
Buffers are often added to maintain a constant
pH in biochemical research
6
and naturally occurring
buffer systems within body fluids and cells are very
important (Box 6-A). Among the most important
natural buffers are the proteins themselves, with the
imidazole groups of histidine side chains providing
much of the buffering capacity of cells around pH 7
(Figs. 3-1 and 3-2). Table 3-1 lists some useful bio-
chemical buffers and their pK
a
values. Here are a few
A. Understanding pH and Electrical Charges on Macromolecules
(3-7)
(3-8)
pH pK
a
= log ([A]/[HA])
pH pK
a
= log [/(1 )];
= [A] / [HA] + [A] =
10
(pH pK
a
)
1 + 10
(pH pK
a
)
98 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-2 Acidbase titration curve for hen lysozyme at 0.1 ionic strength
and 25

C. , initial titration from the pH attained after dialysis; , back


titration after exposure to pH 1.8; , back titration after exposure to pH 11.1.
The solid curve was constructed on the basis of intrinsic pK
a
values based
on NMR data. From Kuramitsu and Hamaguchi
5
0
H
+

d
i
s
s
o
c
i
a
t
e
d

p
e
r

p
r
o
t
e
i
n

m
o
l
e
c
u
l
e
5
10
15
20
0 1 2 3 4 5 6 7 8 9 10 11 12
pH
practical hints about buffer preparation. Buffers con-
taining monovalent ions tend to change pH with dilution
less than do those with multivalent ions such as HPO
4
2
or HP
2
O
7
3
. The pK
a
values of carboxylic acids and of
phosphoric acid, or of its organic derivatives, change
very little with change in temperature. The pH of a
buffer prepared with such components is nearly inde-
pendent of temperature (Table 3-1). However, the pK
a
of the NH
3
+
group changes greatly with temperature.
Buffer composition can be calculated readily from Eq.
3-8 and pK
a
values from Table 3-1. It is convenient
to keep in the laboratory standardized (to ~ 1% error)
1 M HCl and 1 M NaOH for use in buffer preparation.
Compositions calculated from Eq. 3-8 will usually
yield buffers of pH very close to those expected.
Final adjustment with HCl may be needed if the pH
is more than one unit away from a pK
a
value. When
two buffering materials are present, the composition
should be calculated independently for each. The
measurement of pH should always be done with great
care because it is easy to make errors. Everything
depends upon the reliability of the standard buffers
used to calibrate the pH meter.
7
Often, especially
during isolation of small compounds, it is desirable to
work in the neutral pH region with volatile buffers, e.g.,
trimethylamine and CO
2
or ammonium bicarbonate,
which can be removed by vacuum
evaporation or lyophilization.
8
B. Isolating Compounds
Before structural work can
begin, pure substances must be
separated from the complex mixtures
in which they occur in cells and
tissues.
4,13 24
Often, a substance
must be isolated from a tissue in
which it is present in a very low
concentration. After it is isolated in
pure form, if it is a large molecule,
it must often be cut up into smaller
pieces which are separated, purified,
and identified. Accurate quantita-
tive analysis is required to deter-
mine the ratios of these fragments.
Considerable ingenuity may then
have to be exercised in putting the
pieces of the jigsaw puzzle back
together to determine the structure
of the native molecule. Many
books, a few of which are cited
here,
4,13 23,25 45
provide instructions.
There are also journals and other
periodicals dedicated to biochemical
methods.
46 53
1. Fractionation of Cells and Tissues
A fresh tissue or a paste of packed cells of a micro-
organism, usually collected by centrifugation, may be
the starting material.
23,54 57
Tissue is often ground in
a kitchen-type blender or, for gentler treatment, in a
special homogenizer. The popular PotterElvehjem
homogenizer is a small apparatus in which a glass or
plastic pestle rotates inside a tight-fitting mortar tube
(see standard laboratory equipment catalogs for pic-
tures). Microbial cells are frequently broken with
supersonic oscillation (sonication) or in special pres-
sure cells. It is important to pay attention to the pH,
buffer composition, and, if subcellular organelles are
to be separated, the osmotic pressure. To preserve the
integrity of organelles, 0.25 M sucrose is frequently
used as the suspending medium, and MgCl
2
as well
as a metal complexing agent such as ethylenediamine-
tetraacetate (EDTA) (Table 6-10) may be added. Solu-
ble enzymes are often extracted without addition of
sucrose, but reducing compounds such as glutathione
(Box 11-B), mercaptoethanol, or dithiothreitol (Eq. 3-23)
may be added. The crude homogenate may be strained
and is usually centrifuged briefly to remove cell frag-
ments and other debris. Large-scale purification of
99
TABLE 3-1
Practical pK
a
Values for Some Useful Buffer Compounds at 25C and Ionic Strength 0.1
a
B. Isolating Compounds
Charge on conjugate base
a
Based on compilation by Ellis and Morrison
9
with additional data from Good et al.
10,11
and Dawson et al.
12
The Good buffers have dipolar
ionic constituents. Since no form is without electrically charged groups they are unlikely to enter and disrupt cells.
b
Abbreviations used:
BES N,N-Bis(2-hydroxyethyl)-2-aminoethanesulfonic acid
BICINE N,N-Bis(2-hydroxyethyl)glycine
BIS-TRIS Bis(2-hydroxyethyl)iminotris(hydroxymethyl)methane
HEPES N-2-Hydroxyethylpiperazine-N-2-ethanesulfonic acid
MES 2-(N-Morpholino)ethanesulfonic acid
PIPES Piperazine-N,N-bis(2-ethanesulfonic acid)
TES N-Tris(hydroxymethyl)methyl-2-aminoethanesulfonic acid
TRICINE N-Tris(hydroxymethyl)methylglycine
TRIS Tris (hydroxymethyl) aminomethane
c
For CO
2
(solid) + H
2
O H
+
+ HCO
3

, apparent pK
a
.
Compound pK
a
Grams per mole d(pK
a
) / dT
Citric acid (pK
1
) 192 1
Formic acid 3.7 0 1
Citric acid (pK
2
) 4.45 192 0.0016 2
Acetic acid 4.64 60 0.0002 1
Succinic acid (pK
2
) 5.28 118 0 2
Citric acid (pK
3
) 5.80 192 0 3
3,3-Dimethylglutaric acid 5.98 160 0.006 2
Piperazine (pK
1
) (5.68) 6.02 86 0
Cacodylic acid (dimethylarsinic acid) 6.1 138 1
MES
b
6.1 195 0.011
BIS-TRIS
b
6.41 209 .017
Carbonic acid (pK
1
) 6.4
c
Pyrophosphoric acid (pK
3
) 6.76 178 -2
Phosphoric acid (pK
2
) 6.84 98 .0028 2
PIPES
b
6.90 353 .0085 2
Imidazole 7.07 68 .020 0
BES
b
7.06 213 .016 1
Diethylmalonic acid 7.2 136
TES
b
7.37 279 .020 1
HEPES
b
7.46 238 .014 1
N-Ethylmorpholine 7.79 115 .022 0
Triethanolamine 7.88 149 .020 0
TRICINE
b
8.02 178 .021 1
TRIS
b
8.16 121 .031 0
Glycylglycine 8.23 132 .028 1
BICINE
b
8.26 163 .018 1
4-Phenolsulfonic acid 8.70 174 .013 2
Diethanolamine 9.00 105 .024 0
Ammonia 9.2 0.031 0
Boric acid 9.2 0.008 mixed
Pyrophosphoric acid (pK
4
) 9.41 178 -3
Ethanolamine 9.62 61 .029 0
Glycine (pK
2
) 9.8 75 0.025 1
Piperazine (pK
2
) 9.82 86 0
Carbonic acid (pK
2
) 10.0 0.009 2
Piperidine 11.1 85 0
100 Chapter 3. Determining Structures and Analyzing Cells
proteins is often initiated with such a crude homogenate.
Cell organelles are also often separated by centri-
fugation. In one procedure a homogenate in 0.25 M
sucrose (isotonic with most cells) is centrifuged for
10 min at a field of 600 1000 times the force of gravity
(600 1000 g) to sediment nuclei and whole cells. The
supernatant fluid is then centrifuged another 10 min
at ~ 10,000 g to sediment mitochondria and lysosomes.
Finally, centrifugation at ~ 100,000 g for about an hour
yields a pellet of microsomes (p. 14),
58
which contains
both membrane fragments and ribosomes. Each of the
separated components can be resuspended and recen-
trifuged to obtain cleaner preparations of the organelles.
The sedimented particles can often be solubilized by
chemical treatment, for example, by the addition of
either ionic or nonionic detergents. Membrane proteins
can be isolated following solubilization in this way
(Chapter 8). The soluble supernatant fluid remaining
after the highest speed centrifugation provides the
starting material for isolation of soluble enzymes and
many small molecules.
2. Separations Based on Molecular Size,
Shape, and Density
The simplest way to separate very large dissolved
molecules is to let the small ones pass through a suit-
able sieve which may be a membrane with holes or a
bed of gel particles. If the size of the particles approaches
that of the holes in the sieve, the rate of passage will
depend upon shape as well as size.
Dialysis, ultrafiltration, and perfusion chro-
matography. In dialysis
59
and ultrafiltration,
60
a thin
membrane, e.g., made of cellulose acetate (cellophane)
and containing holes 110 nm in diameter (typically
5 nm), is used as a semipermeable barrier. Small
molecules pass through but large ones are retained.
Dialysis depends upon diffusion and can be hastened
by adequate stirring. Ultrafiltration requires a pressure
difference across the membrane. The more sophisticated
procedures of gel filtration and perfusion chromato-
graphy were introduced in 1959.
6165
A column is
packed with material such as the crosslinked dextran
Sephadex, polyacrylamide gels (such as the Bio-Gel P
Series), or agarose gels (e.g., Bio-Gels A and Sepharoses).
These come in the form of soft beads, the interior net-
work of which is a three-dimensional network of poly-
mer strands (Fig. 4-10). Recently porous beads of hard
crosslinked polystyrene, glass, or various other silicate
materials have been employed.
66 68
The interstices
between strands, whose size depends upon the degree
of crosslinking introduced chemically into the gel, are
small enough to exclude large molecules but to admit
smaller ones. If a mixture of materials of different
molecular size is passed through such a column the
smaller molecules are retarded because of diffusion
into the gel, while the larger molecules pass through
unretarded (Fig. 3-3). Sephadex G-25 excludes all but
salts and compounds no larger than a simple sugar
ring. Sephadex G-200, which is much less crosslinked,
permits separation of macromolecules in the range of
5 200 kDa. As is explained in Section B, gel filtration
also provides an important way of estimating M
r
for
proteins and other macromolecules.
Centrifugation. Centrifuges of many sizes and
speeds are used in the laboratory to remove debris as
well as to collect precipitated proteins and other mate-
rials at various steps in a purification scheme. The most
remarkable are the ultracentrifuges which produce
forces greater than 4 x 10
5
times that of gravity. They
can be used both for separation of molecules and for
determination of M
r
(see Section C).
When macromolecules in a solution are subjected
to an ultracentrifugal field they are accelerated rapidly
to a constant velocity of sedimentation. This is expressed
as a sedimentation constant s, which is the rate (cm/s)
per unit of centrifugal force. The unit of s is the second
but it is customary to give it in Svedberg units, S (1S =
10
13
s). Sizes of particles are often cited by their S values.
The sedimentation constant is affected by the sizes,
shapes, and densities of the particles as is discussed
further in Section C. If carried out at constant velocity
an equilibrium will eventually be attained in which
sedimentation is just balanced by diffusion and a
smooth concentration gradient forms from the top to
the bottom of the centrifuge cell or tube. Concentra-
tion gradients can also be formed by centrifuging a
Figure 3-3 Separation of oligosaccharides by gel filtration.
The sugars dissolved in distilled water were passed through
a column of Sephadex G-25. The peaks contain (right to
left) glucose, cellobiose, cellotriose, etc. From Flodin and
Aspberg.
64
0
800 900 1000 1100 1200 1300 1400 1500
ml
I
n
t
e
r
f
e
r
o
m
e
t
e
r

r
e
a
d
i
n
g
5000
4000
3000
2000
1000
101
concentrated solution of small molecules.
13,69,70
Such
a concentration gradient is also a density gradient
which can be made very steep. For example, a gradient
with over a 10% increase in density from top to bottom
can be created using 6 M cesium chloride (CsCl) and
is widely used in DNA separations. If DNA is added
prior to centrifugation it will come to rest in a narrow
band or bands determined by the buoyant densities
of the species of DNA present (see Chapter 5). After
centrifugation, which is usually done in a plastic tube,
a hypodermic needle is inserted through the bottom
of the tube and the contents are pumped or allowed
to flow by gravity into a fraction collector.
Another type of gradient centrifugation (zone
centrifugation) utilizes a preformed gradient to sta-
bilize bands of cell fragments, organelles, or macro-
molecules as they sediment.
71 74
For example, RNA
can be separated into several fractions of differing
sedimentation constants in a centrifuge tube that con-
tains sucrose ranging in concentration from 25% at
the bottom to 5% at the top. This is prepared by a
special mixing device or gradient maker prior to
centrifugation. The solution of RNA is carefully layered
on the top, the tube is centrifuged at a high speed for
several hours, and the different RNA fractions sepa-
rate into slowly sedimenting sharp bands. A 2060%
gradient of sucrose or glycerol may be used in a similar
way to separate organelles.
58
3. Separations Based on Solubility
Some fibrous proteins are almost insoluble in
water and everything else can be dissolved away.
More often soluble proteins are precipitated from
aqueous solutions by adjustment of the pH or by
addition of large amounts of salts or of organic solvents.
The solubility of any molecule is determined both by
the forces that hold the molecules together in the solid
state and by interactions with solvent molecules and
with salts or other solutes that may be present.
58a
Proteins usually have many positively and nega-
tively charged groups on their surfaces. If either a
positive or a negative charge predominates at a given
pH the protein particles will tend to repell each other
and to remain in solution. However, near the isoelec-
tric point (pI), the pH at which the net charge is zero
(see Section 7), the solubility will usually be at a mini-
mum. The pH of a tissue extract may be adjusted
carefully to the pI of a desired protein. Any protein
that precipitates can be collected by centrifugation and
redissolved to give a solution enriched in the protein
sought. Some proteins, such as those classified (by an
old system) as globulins, are insoluble in water but
are readily salted in by addition of low concentra-
tions (e.g., up to 0.1 M) of salts. Low concentrations of
salts increase the solubility of most proteins because
the salt ions interact with the charged groups on the
protein surfaces and interfere with strong electro-
static forces that are often involved in binding protein
molecules together in the solid state. Some salts,
including CaCl
2
and NaSCN, which bind to proteins,
are especially effective in salting in.
75,76
Addition of
high concentrations of salt causes precipitation of most
proteins from aqueous solutions. The most effective
and most widely used materials for this salting out
of proteins are (NH
4
)
2
SO
4
and Na
2
SO
4
. Because the
salt ions interact so strongly with water, the protein
molecules interact less with water and more with
each other. A similar intramolecular effect may cause
the stabilization of proteins of halophilic bacteria by
1 4 M KCl.
77
Different proteins precipitate at different concen-
trations of an added salt. Hence, a fraction of proteins
precipitating between two different concentrations of
salt can be selected for further purification (Fig. 3-4).
Protein concentrations can be estimated as described
in Box 3-A.
Precipitation methods are popular first steps in
purification of proteins because they can be carried
out on a large batch scale. The amounts of ammo-
nium sulfate used are often expressed as percentage
saturation, i.e., as a percentage of the amount required
to saturate the solution (4.1 M at 25). Convenient
tables are available
12,78
that permit one to weigh out
the correct amount of solid ammonium sulfate to give
a desired percentage saturation or to go from one
B. Isolating Compounds
Figure 3-4 Hypothetical behavior of a solution containing
three proteins, A, B, and C, upon ammonium sulfate frac-
tionation. The concentration of protein remaining in the
solution is plotted against ammonium sulfate concentration
(usually expressed as % saturation). Addition of ammonium
sulfate to concentration c
1
will precipitate largely protein B,
which can be removed by centrifugation. Addition of addi-
tional salt to c
2
will precipitate largely protein C, while A
remains in solution.
c
1
c
2
Concentration of (NH
4
)
2
SO
4
P
r
o
t
e
i
n

c
o
n
c
e
n
t
r
a
t
i
o
n
C
B
A
102 Chapter 3. Determining Structures and Analyzing Cells
percentage saturation to a higher one.
Proteins are often stabilized by low concentrations
of simple alcohols or ketones
76
and by higher concen-
trations of polyhydroxy alcohols, such as glycerol
77
and sucrose,
78
and also by certain inert, synthetic
polymers such as polyethyleneglycol (PEG).
79
The
latter is a widely used precipitant. The polyhydroxy-
alcohols and PEG are all hydrated but tend not to
interact strongly with the protein molecules. On the
other hand, simple alcohols may denature proteins by
their interaction with nonpolar regions.
77
4. Separation by Partition
Many of the most important separation methods
are based on repeated equilibration of a material be-
tween two separate phases, at least one of which is
BOX 3-A QUANTITATIVE ESTIMATION OF PROTEIN CONCENTRATIONS
Biochemists often need to estimate the content
of protein in a sample. For example, in devising a
purification procedure for an enzyme it is customary
to estimate the number of units of enzyme activity
(as defined in Chapter 9) per milligram of protein
(U/ mg). As progress is made in the purification
this ratio increases. It becomes constant with respect
to additional purification attempts, when a homoge-
neous enzyme is obtained.
One of the most widely used and most sensitive
protein assays (for 0.1 1 mg/ml of protein) is the
colorimetric procedure of Lowry.
a c
It makes use of
a phosphomolybdicphosphotungstic acid reagent
(the FolinCiocalteu reagent) which is reduced by
proteins in the presence of alkaline Cu
2+
to charac-
teristic blue oxides whose color can be monitored
at 750 nm. Much of the color comes from the reduc-
ing action of tyrosine and tryptophan. The color
yield varies greatly from protein to protein and users
may have trouble with reproducibility. A related
method utilizes bicinchoninic acid which forms
a purple color (measured at 362 nm) with the Cu
+1
that is formed by reduction of alkaline Cu
2+
by the
protein.
d f
This reagent is easier to use than that of
the Lowry procedure and gives stable and repro-
ducible readings.
A third widely used procedure, introduced by
Bradford
g
and modified by others, measures the
binding of the dye Coomassie brilliant blue whose
peak absorption shifts from 465 nm to 595 nm upon
binding. The change occurs within two minutes
and is stable. However, the color yield varies from
one protein to another.
a
Lowry, O. H., Rosebrough, N. J., Farr, A. L., and Randall, R. J.
(1951) J. Biol. Chem. 193, 265275
b
Peterson, G. L. (1979) Anal. Biochem. 100, 201220
c
Larson, E., Howlett, B., and Jagendorf, A. (1986) Anal. Biochem.
155, 243248
d
Smith, P. K., Krohn, R. I., Hermanson, G. T., Mallia, A. K.,
Gartner, F. H., Provenzano, M. D., Fujimoto, E. K., Goeke, N.
M., Olson, B. J., and Kenk, D. C. (1985) Anal. Biochem. 150, 7685
e
Davis, L. C., and Rodke, G. A. (1987) Anal. Biochem. 161, 152156
f
Hill, H. D., and Straka, J. G. (1988) Anal. Biochem. 170, 203208
g
Bradford, M. M. (1976) Anal. Biochem. 72, 248254
h
Fruton, J. S., and Simmonds, S. (1958) General Biochemistry, 2nd
ed., Wiley, New York (p. 130)
i
Layne, E. (1957) Methods Enzymol. III, 450451
Less sensitive but very simple and precise is
measurement of the light absorption around 280
nm. This is discussed in the main text in Section
D,6. For a typical protein an absorbance of 1.0 at
280 nm corresponds to a protein concentration of
1 mg / ml.
h
The very old biuret method is also
useful for samples containing 1 10 mg / ml of pro-
tein. The violet color that arises upon addition of
copper sulfate to an alkaline solution of a peptide
or protein is recorded at 540 560 nm.
h
The color is
especially intense for longer polypeptides. The
name of the method arises from the fact that biuret
gives a similar color
i
(see also Eq. 6-85).
usually liquid. Small molecules may be separated by
countercurrent distribution in which a material is
repeatedly partitioned between two immiscible liquid
phases, one more polar than the other. New portions
of both liquids are moved by a machine in a counter-
current manner between the equilibration steps or are
moved continuously through coiled tubes.
80 82,82a
A similar result is accomplished by using as one
phase a solid powder or fine beads packed in a verti-
cal column or spread in a thin layer on a plate of glass.
The methods are usually referred to as chromatogra-
phy, a term proposed by Tswett to describe separation
of materials by color. In 1903 Tswett passed solutions
of plant leaf pigments (chlorophylls and carotenes) in
nonpolar solvents such as hexane through columns of
alumina and of various other adsorbents and observed
separation of colored bands which moved down the
column as more solvent was passed through. Individual
C
O
H
2
N N NH
2
Biuret
H
C
O
103
pure pigments could be eluted from the column by
continued passage of solvent. This important method
is called adsorption chromatography. It is assumed
that the pigments are absorbed on the surface rather
than being disssolved in the solid material. A related
method is hydrophobic interaction chromatography
of proteins.
83 86
The packing material is similar to that
described in Section 6 (Affinity Chromatography) but
bears long-chain alkyl groups that can interact with
the hydrophobic patches on surfaces of proteins. A
very different adsorbent that is very useful in separa-
tion of proteins is carefully prepared microcrystalline
hydroxylapatite.
87,88
It presumably functions in part
by ion exchange.
Column packing materials such as silica gel
contain a large amount of water, and separation involves
partition between an immobilized aqueous phase in
the gel and a mobile, often organic, solvent flowing
through the column. Usually materials elute sooner
when they are more soluble in the mobile phase than
in the aqueous phase. These methods are closely related
to perfusion chromatography, which is described in
Section 2.
Aromatic amino acids, lipids, and many other
materials can be separated on reversed-phase columns
in which nonpolar groups, usually long-chain alkyl
groups, are covalently attached to silica gel, alumina,
or other inert materials.
66,80
The mobile phase is a
more polar solvent, often aqueous, and gradually
made less polar by addition of an organic solvent.
In reversed-phase chromatography more polar com-
pounds migrate faster through the system than do
nonpolar materials, which experience hydrophobic
interaction with the solid matrix.
Many traditional chromatographic methods in-
cluding reversed-phase chromatography have been
adapted for use in automatic systems which employ
columns of very finely divided solid materials such as
silica, alumina, or ion exchange materials coated onto
fine glass beads.
89 91
These high-performance liquid
chromatographic (HPLC) systems often utilize pres-
sures as high as 300 atmospheres. Separations are often
sharper and faster than with other chromatographic
methods.
92 97
Reversed-phase columns in which the
solid matrix may carry long (e.g. C
18
) hydrocarbon
chains have been especially popular for separation
both of peptides and of proteins. Proteins may also
be separated by gel filtration, ion exchange, or other
procedures with HPLC equipment.
A sheet of high-quality filter paper containing
adsorbed water serves as the stationary phase in
paper chromatography. However, thin-layer
chromatography, which employs a layer of silica gel
or other material spread on a glass or plastic plate, has
often supplanted paper chromatography because of its
rapidity and sharp separations (Fig. 3-5).
16,96a,98 100
An approach that requires no stationary phase at all is
field flow fractionation.
101
Here a suitable external
field (e.g., electrical, magnetic, and centrifugal) or a
thermal gradient is imposed on the particles flowing
through a narrow channel.
For volatile materials vapor phase chromatography
(gas chromatography) permits equilibration between
the gas phase and immobilized liquids at relatively
high temperatures. The formation of volatile deriva-
tives, e.g., methyl esters or trimethylsilyl derivatives
of sugars, extends the usefulness of the method.
103,104
A method which makes use of neither a gas nor a
liquid as the mobile phase is supercritical fluid
chromatography.
105
A gas above but close to its
critical pressure and temperature serves as the solvent.
The technique has advantages of high resolution, low
temperatures, and ease of recovery of products.
Carbon dioxide, N
2
O, and xenon are suitable solvents.
5. Ion Exchange Chromatography
Separation of molecules that contain electrically
charged groups is often accomplished best by ion
exchange chromatography.
105a
The technique depends
upon interactions between the charged groups of the
molecules being separated and fixed ionic groups on
B. Isolating Compounds
Figure 3-5 Photograph of a two-dimensional thin layer
(silica gel) chromatogram of a mixture of flavins formed by
irradiation of ~ 10 g of the vitamin riboflavin. The photo-
graph was made by the fluorescence of the compounds
under ultraviolet light. Some riboflavin (RF) remains. The
arrows indicate the location of the sample spot before chro-
matography. Chromatography solvents: a mixture of acetic
acid, 2-butanone, methanol, and benzene in one direction
and n-butanol, acetic acid, and water in the other. See Tread-
well et al.
102
RF
104 Chapter 3. Determining Structures and Analyzing Cells
an immobile matrix. Separation depends upon small
differences in pK
a
values and net charges and upon
varying interactions of nonpolar parts of the molecules
being separated with the matrix. Since changes in pH
can affect both the charges on the molecules being
separated and those of the ion exchange material, the
affinities of the molecules being separated are strongly
dependent on pH. For example, proteins and most
amino acids are held tightly by cation exchangers at
low pH but not at all at high pH.
Aqueous solutions are usually employed and the
columns are packed with beads of ion exchange
resins, porous materials containing bound ionic
groups such as SO
3

, COO

, NH
3
+
, or quaternary
nitrogen atoms. Synthetic resins based on a cross-
linked polystyrene are usually employed for separation
of small molecules. For larger molecules chemical
derivatives of cellulose or of crosslinked dextrans
(Sephadex), agarose, or polyacrylamide are more
appropriate. Positively charged ions, such as amino
acids in a low pH solution, are placed on a cation
exchange resin such as Dowex 50, which contains
dissociated sulfonic acid groups as well as counter
ions such as Na
+
, K
+
, or H
+
. The adsorbed amino
acids are usually eluted with buffers of increasing pH
containing sodium or lithium ions. The procedure,
Figure 3-6 Separation of amino acids by cation-exchange chromatography on a sulfonated polystyrene resin in the Na
+
form
by the method of Moore and Stein.
110
The amino acids were detected by reaction with ninhydrin (Box 3-C); areas under the
peaks are proportional to the amounts. Two buffers of successively higher pH are used to elute the amino acids from one
column, while a still higher pH buffer is used to separate basic amino acids on a shorter column. From Robyt and White.
13
1.0
0.5
0.4
0.3
0.1
50 100 150 200 250 300 350 400 450 50 100 150
CySO
3
H
Asp
MetSO
Thr
Ser
Glu
Pro
Gly
Ala
Cys
Val
Met
Ile
Leu
Tyr
Phe
Try
Lys
His
NH
3
Arg
0.2
A
b
s
o
r
b
a
n
c
e

a
t

5
7
0

n
m
150 cm column
15 cm column
pH 3.25, 0.2N Na citrate pH 4.25, 0.2N Na citrate pH 5.28, 0.35
N Na citrate
Effluent (ml)
which was developed by Moore and Stein,
106 109
is
widely used for automatic quantitative analysis of
amino acid mixtures obtained by hydrolysis of a pro-
tein or peptide (Fig. 3-6).
110 113
Ion exchange chromatography of proteins and
peptides is often done with such ion exchange materials
as carboxymethyl-Sephadex and phosphocellulose,
which carry negatively charged side chains or diethyl-
aminoethylcellulose (DEAE-cellulose), which carries
positively charged amino groups.
114
These materials
do not denature proteins or entrap them and have a
large enough surface area to provide a reasonable
absorptive capacity. The mobile phase is usually
buffered. For anion exchangers the pH should be
above ~ 4.4 to keep most carboxylate side chains on the
proteins ionized. The pH may be increased to pH 710,
where most histidine imidazolium ions have dissociated,
increasing the mobility of many proteins. For cation
exchange the pH is usually buffered below pH 6 or 7
(Fig. 3-6).
115,116
6. Affinity Chromatography
In this technique the chromatographic absorbent
is designed to make use of specific biochemical inter-
105
actions to hook selectively a particular macromolecule
or group of macromolecules.
117 122
Affinity chromato-
graphy is used in many ways, including the purifica-
tion of enzymes, antibodies, and other proteins that
bind tightly to specific small molecules.
Because of their open gel structure (Fig. 4-10) agar-
ose derivatives in bead form provide a good solid
support matrix. The hydroxyl groups of the agarose
are often linked to amino compounds. In one widely
used procedure
119
the agarose is treated with cyanogen
bromide (Br C N) in base to activate the carbohy-
drate (Eq. 3-9, step a). Then the amino compound is
added (step b). The isourea product shown is the major
one with agarose gels but dextran-based matrices tend
to react by steps ce.
Absorbents containing a large variety of R groups
of shapes specifically designed to bind to the desired
proteins can be made in this way. If the coupling is
done with a diamine [R = (CH
2
)
n
NH
2
], the resulting
-aminoalkyl agarose can be coupled with other com-
pounds by reaction with carbodiimide (Eq. 3-10). For
reaction in nonaqueous medium dicyclohexylcarbodi-
imide (R = cyclohexyl) is often used, but for linking
groups to agarose a water-soluble reagent such as
1-ethyl-3-(3-dimethylaminopropyl) carbodiimide is
recommended.
120
Carbodiimides are widely used for
forming amide or phosphodiester linkages in the
laboratory. The formation of an amide with a side
chain of -aminoalkyl agarose can be pictured as in
Eq. 3-10.
OH
OH
Polysaccharide
matrix
CNBr Br

R-NH
2
Cyanate
esters
R-NH
2
O C
O
NH R
OH
C N
O
O
R
Major products
from dextrans
H
2
O
O C
O
NH
2
OH
C NH
O
O
OH
O C N
Carbamates minor
inert products
Imidocarbonates
a b
c
d
e
An isourea; major
product from agarose
OH
O C
N
NH R
H
Many other means of preparation of adsorbents
for affinity chromatography are also available.
121,122
For example, 1,1'-carbonyl-diimidazole can be used
to couple a diamine to the matrix (Fig. 3-11). This
reagent has the advantage that it does not depend
upon the relatively unstable isourea linkages formed
by Eq. 3-9 to hold the specific affinity ligands.
121
An example of a successful
application of affinity chromatogra-
phy is the isolation of the enzyme
cytidine deaminase from cells of E.
coli. Cytidine was linked covalently
via long spacer arms to the agarose
beads as in the following diagram:
B. Isolating Compounds
(3-9)
R C
O
O C
HN
N
R"
R'
O C
N
N R"
R'
H
H
O C
O
NH(CH
2
)
n
NH
2
Agarose
R C
O
O

Carbodiimide
O C NH
O
(CH
2
)
n
NH C
O
R Agarose
R' N C N R"
(3-10)
O C
N
N Matrix
O
O C Matrix
O
NH R
R-NH
2
Imidazole
Imidazole
Matrix OH
1,1'-carbonyl-diimidazole
N
N C
O
N
N
(3-11)
106 Chapter 3. Determining Structures and Analyzing Cells
A cell extract was subjected to ammonium sulfate
fractionation and the dialyzed protein was then poured
through the affinity column which held the cytidine
deaminase molecules because of their affinity for the
cytidine structures that were bound to the agarose.
The protein was eluted with a borate buffer; the borate
formed complexes with the adjacent hydroxyl groups
of the cytidine and thereby released the protein. After
passage of the protein through an additional column
of DEAE-Sephadex the deaminase had been purified
1700-fold compared to the crude extract.
123
Another technique is to engineer genes to place a
polyhistidine tag at the C terminus of a protein chain.
Commercial cloning vehicles and kits are available for
this purpose.
124
The protein produced when the engi-
neered gene is expressed can be captured by the affini-
ty of the polyhistidine tag for Cu
2+
, Ni
2+
, Co
2+
, or Zn
2+
held in chelated form on an affinity column.
124 127
A surprising discovery was that certain dyes, for
example Cibachron blue, when covalently coupled to a
suitable matrix, often bind quite specifically to proteins
that have a nonpolar binding pocket near a positive
charge.
128
This includes many enzymes that act on
nucleotides.
Spacer arm
N
N
NH
2
O
N
NH
HN
O
O
O
O
O
OH OH
H
Cytidine
Agarose
HOCH
2
O
O NH
2
SO
3
H
NH
SO
3
H
NH
N N
N
Cl
NH
SO
3
H
Cibacron blue F3GA
7. Electrophoresis and Isoelectric Focusing
The methods considered in this section make use
of movement of molecules in an electrical field. Sepa-
ration depends directly upon differences in the net
charge carried by molecules at a fixed pH. The net
charge for compounds containing various combina-
tions of acidic and basic groups can be estimated by
considering the pK
a
of each group and the extent to
which that group is dissociated at the selected pH
using Eqs. 3-3 to 3-5. At some pH, the isoelectric
point (pI), a molecule will carry no net charge and
will be immobile in an electric field. At any other pH
it will move toward the anode (+) or cathode (). The
pH at which the protein carries no net charge in the
complete absence of added electrolytes is called the
isoionic point.
129
Electrophoresis, the process of separating mole-
cules, and even intact cells
130
(Box 3-B), by migration in
an electrical field, is conducted in many ways.
28,131 140
In zone electrophoresis, a tiny sample of protein
solution, e.g., of blood serum, is placed in a thin line
on a piece of paper or cellulose acetate. The sheet is
moistened with a buffer and electrical current is passed
through it. An applied voltage of a few hundred volts
across a 20-cm strip suffices to separate serum proteins
in an hour. To hasten the process and to prevent diffu-
sion of low-molecular-weight materials, a higher volt-
age may be used. Two to three thousand volts may
be applied to a sample cooled by water-chilled plates.
Large-scale electrophoretic separations may be con-
ducted in beds of starch or of other gels.
One of the most popular and sensitive methods
for separation of proteins is electrophoresis in a column
filled with polyacrylamide or agarose gel or on a
thin layer of gel on a plate. The method depends
upon both electrical charge and molecular size and
has been referred to as electrophoretic molecular
sieving.
28,133,135 137,141 143
Polyacrylamide gel electro-
phoresis is often carried out in the presence of ~ 1%
of the denaturing detergent sodium docecyl sulfate
which coats the polypeptide chain rather evenly. This
method, which is often referred to as SDSPAGE,
has the advantage of breaking up complex proteins
composed of more than one subunit and sorting the
resultant monomeric polypeptide chains according to
molecular mass (see Box 3-C). A disulfide-reducing
reagent (see Eq. 3-23) such as ~ 1% 2-mercaptoethanol
is usually present but may be omitted to permit detec-
tion of crosslinked peptides.
Capillary electrophoresis is increasingly popu-
lar
144 149a
and can be used to separate attomole
amounts.
150
It can be used not only for separation of
proteins but also for rapid estimation of the net charge
on a protein.
151
The separation is conducted in tubes
with internal diameters as small as 10 15 m and as
short as 1 cm. Multiple channels cut into a glass chip
107
BOX 3-B SORTING AND ANALYZING SINGLE CELLS
B. Isolating Compounds
It is often important to examine and analyze
individual cells.
a
For example, large numbers of
single blood cells can be tested for the presence of
specific antigenic determinants that arise by muta-
tion. This permits assessment of the frequency of
these mutations.
b
The complex chemical processing
of neuropeptides can be studied on the contents of a
single neuron (Chapter 30) using mass spectrometry.
c
Several methods for separating cells have been
devised. These include electrophoresis
d
or use of
magnetic microspheres.
b
Micromanipulation can
sometimes be used to select single cells for analysis.
The most impressive technique is flow cytometry,
e,f
which is used daily on human blood samples in
clinical laboratories. A suspension of cells is passed
at a high rate of flow through a narrow capillary of
~ 0.2 mM diameter. The sample stream, which is
surrounded by a larger sheath stream, has a
a
Yeung, E. S. (1994) Acc. Chem. Res. 27, 409414
b
Jovin, T. M., and Arndt-Jovin, D. J. (1980) Trends Biochem. Sci. 5,
214219
c
Li, K. W., Hoek, R. M., Smith, F., Jimnez, C. R., van der Schors,
R. C., van Veelen, P. A., Chen, S., van der Greef, J., Parish, D. C.,
Benjamin, P. R., and Geraerts, W. P. M. (1994) J. Biol. Chem. 269,
3028830292
d
Bauer, J. (1994) Cell Electrophoresis, CRC Press, Boca Raton,
Florida
e
Shapiro, H. M. (1995) Practical Flow Cytometry, 3rd ed., Wiley-
Liss, New York
f
Darzynkiewicz, Z., Robinson, J. P., and Crissman, H. A., eds.
(1994) Flow Cytometry, 2nd ed., Academic Press, San Diego
Electropherogram of major proteins from a single erythro-
cyte. Peaks A, B, and C are carbonic anhydrase (~ 7 amol),
methemoglobin (~ 5 amol), and hemoglobin A
0
(~ 450 amol),
as identified from migration times relative to standards.
a
Courtesy of Edward S. Yeung.
Flow cytometric histogram of fluorescently labeled live
and dead E. coli bacteria. The dye kit (BacLightTM) that
was used stains membrane-compromised dead bacteria
with a red fluorescing dye and live bacteria with a green
fluorescing dye. Cells were analyzed on an EPICS XL
cytometer (Coulter Corporation). By integrating the area
under each population it was possible to discern the
percentage of dead (54), dying (16), and live (30) bacteria
within a mixed population. The use of fluorescent dyes
such as these has proven useful for studying various
mechanisms employed by the food industry for killing
microorganisms in food products and for studying a
variety of bacteria derived from seawater, soil, plant
materials, and laboratory-grown cultures. Courtesy of
Kristi Harkins.
F
l
u
o
r
e
s
c
e
n
c
e
Time (min)
9 12 15
A
B
C
smaller diameter of ~ 20 m. One or more laser
beams are used to record information about each
cell over a period of a few microseconds as the cells
pass by at a rate of as much as 10
5
cells / s.
Flow cytometers developed from simpler cell
counters, but now they are used to record cell size
(from light scattering), optical absorbance, fluores-
cence, and phosphorescence. The optical properties
are often enhanced by staining. The use of two dyes
that fluoresce at different wavelengths permits the
construction of two-dimensional plots as in the
accompanying figure.
Capillary electrophoresis is one of the tech-
niques able to separate constituents of single cells
and is illustrated in the second figure.
C
o
u
n
t
L
o
g

r
e
d

f
l
u
o
r
e
s
c
e
n
c
e
Log green fluorescence
E. coli
Dead
Dying
Live
108 Chapter 3. Determining Structures and Analyzing Cells
(3-12)
can be used.
152
Whereas in conventional zone electrophoresis
most of the electrical current is carried by the buffer, in
isotachophoresis
153,154
the ions being separated carry
most of the current. In isoelectric focusing,
28,155 157
a
pH gradient is developed electrochemically in a verti-
cal column or on a thin horizontal plate between an
anode and a cathode. The pH gradient in a column is
stabilized by the presence of a density gradient, often
formed with sucrose, and the apparatus is maintained
at a very constant temperature. Proteins within the
column migrate in one direction or the other until they
reach the pH of the isoelectric point where they carry
no net charge and are focused into a narrow band.
As little as 0.01 pH unit may separate two adjacent
protein bands which are located at positions corre-
sponding to their isoelectric points. A newer develop-
ment is the use of very narrow pH gradients that are
immobilized on a polyacrylamide matrix.
158 160
With
this technique some hemoglobin mutants differing
only in substitution of one neutral amino acid for
another have been separated.
161
Special techniques are
needed for highly basic proteins.
162
A two-dimensional method in which proteins are
separated by isoelectric focusing (preferably with an
immobilized pH gradient) in the first dimension and
by SDS-gel electrophoresis in the second has become a
popular and spectacularly successful method for
studying complex mixtures of proteins (Box 3-C).
163 166
Over 2000 proteins can be separated on a single plate.
A similar procedure but without SDS can be used to
examine undenatured proteins.
167 169
Computer-
assisted methods are being developed to catalog the
thousands of proteins being identified in this way
170
172
and also to allow rapid identification of spots by
mass spectrometry. The technique can be applied to
intact proteins in subpicomole quantities, even in
whole cell lysates,
150,173,173a
or an enzyme such as
trypsin can be used to cut the proteins into pieces on
the gel plate and the mixtures of peptides can be ana-
lyzed by mass spectrometry.
174 176
Capillary electro-
phoresis or capillary isoelectric focusing can be applied
before samples are sent to the mass spectrometer.
C. Determining the Relative Molecular Mass,
M
r
The evaluation of M
r
is often of critical importance.
Minimum values of M
r
can often be computed from
the content of a minor constituent, e.g., the trypto-
phan of a protein or the iron of hemoglobin. However,
physicochemical techniques provide the basis for most
measurements.
177
Observations of osmotic pressure or
light scattering can also be used and provide determi-
nations of M
r
that are simple in principle, but which
have pitfalls.
178
1. Ultracentrifugation
Some of the most reliable methods for determining
M
r
depend upon analytical ultracentrifuges. These
instruments, capable of generating a centrifugal field
as much as 4 x 10
5
times that of gravity, were devel-
oped in the 1920s and 1930s by T. Svedburg and asso-
ciates in Uppsala, Sweden.
179 181
Driven by oil
turbines, the instruments were expensive and difficult
to use, but by 1948 a reliable electrically driven ma-
chine, the Beckman Model E ultracentrifuge, came into
widespread use. It has had a major impact on our
understanding of proteins, on methods of purification
of proteins, and on our understanding of interactions
of protein molecules with each other and with small
molecules.
182,183
Nevertheless, it was not until 1990
that a truly user-friendly analytical untracentrifuge
became available.
180,184 186
The Beckman Model XL-A
centrifuge has a very small rotor driven by an air-
cooled induction motor and is computer controlled.
Data are recorded automatically in digital form and
computer programs are available to carry out the
necessary computations. The instrument can record
ultraviolet-visible spectra at multiple radial positions
in the sample cell (Fig. 3-7).
A straightforward determination of M
r
is obtained
by centrifuging until an equilibrium distribution of the
molecules of a protein or other macromolecular mate-
rial is obtained and by recording the variation in con-
centration from the center to the periphery of the
centrifuge cell
177,183,185,187 190
(see also Section A,2).
Using short cells, this sedimentation equilibrium
can be attained in 1 5 hours instead of the 1 2 days
needed with older instruments. For a single compo-
nent system the concentration distribution at equilibri-
um is given by Eq. 3-12.
Here c(r) is the concentration c at the radial position r
(measured from the centrifuge axis), a is the radial
distance of the meniscus, M is the molecular mass in
daltons, and v is the partial specific volume in ml/gram.
For most proteins v varies from 0.69 0.75. It is the
reciprocal of the density of the particle. Rho () is the
density (g/ml) of the solvent. A plot of log c(r) against
r
2
is a straight line of slope M (1- v) / 2RT. The com-
puter can also accommodate mixtures of proteins of
differing molecular masses, interacting mixtures,
etc.
185,191
Sedimentation velocity. The relative molecular
mass M
r
can also be measured from observation of the
velocity of movement of the boundary (or boundaries
for multicomponent systems) between solution and
solvent from which the macromolecules have sedi-
c(r) = c(a) exp [M(1- v)
2
(r
2
a
2
) / 2RT]
109
(3-17)
(3-18)
using Eq. 3-14.
By combining Eqs. 3-13 and 3-14 we obtain the Sved-
berg equation:
Here R is in the cgs (cm-gram-second) unit of 8.31 x 10
7
erg mol
1
deg
1
. Using this equation the relative molec-
ular mass M
r
, which is numerically the same as M, can
be evaluated from the sedimentation constant s. Since
s, D, and v must all be measured with care, the method
is demanding. It is often necessary to measure s and D
at several concentrations and to extrapolate to infinite
dilution. It is also customary to correct the data to give
the values s
20,w
and D
20,w
expected at 20C in pure
water at infinite dilution.
To the extent that we can regard protein molecules
as spherical we can substitute for f in Eq. 3-13 the fric-
tional coefficient of a sphere:
Here r
h
is the hydrated radius or Stokes radius of the
protein. On this assumption s will be expected to
increase with the relative molecular mass approxi-
mately as M
r
2 / 3
. A plot of log s against log M
r
should
be a straight line. Figure 3-8 shows such a plot for a
number of proteins. The plots for nucleic acids, which
can often be approximated as rods rather than spheres,
fall on a different line from those of proteins. Further-
more, the sedimentation constant falls off more rapidly
with increasing molecular mass than it should for
spheres.
From analysis of a variety of well-characterized
proteins, Squire and Himmel
192
observed that if pro-
teins are assumed to contain 0.53 g H
2
O per gram of
protein and to have a mean value for v of 0.730 g / cm
3
the value of M
r
can be predicted by Eq. 3-17 with the
standard deviation indicated. Here, S is the sedimen-
tation constant in Svedberg units (10
13
s).
M
r
= 6850 S
3 / 2
) 0.090 M
r
For proteins with various values of v, Eq. 3-18 applies.
M
r
= 922 [S / (1 v)]
3 / 2
) 0.066 M
r
Figure 3-7 The scanning absorption optical system of the
Beckman Optima XL-A ultracentrifuge. Courtesy of
Beckman Coulter.
mented. This boundary, which can be visualized by
optical methods, is quite sharp initially, but it broadens
with time, because of diffusion, as the macromolecules
sediment.
A molecule in a centrifuge is acted upon not only
by the applied centrifugal force but also by an opposing
buoyant force that depends upon the difference in
density of the sedimenting particles and the solvent
and by a frictional drag, which is proportional to a
frictional coefficient f. Setting the sum of these forces
to zero for the hydrodynamic steady state yields Eq.
3-13, which defines the sedimentation constant s.
Here f is the frictional coefficient which is difficult to
predict or to measure but is often assumed to be the
same as the frictional coefficient that affects diffusion.
It can be obtained from the diffusion coefficient D
C. Determining the Relative Molecular Mass, M
r
(3-13)
D k T f RT N f ( ) cm s
B
2 1
= = (3-14)
(3-15)
f r
sphere h
= 6 (3-16)
PM tube
Imaging system for
radial scanning
Wavelength slit
Xenon
flash
lamp
Aperture
Reference
detector
Sample/reference
cell assembly
Filter
Toroidal
grating
s
M
N f
(1 v ) v
2
r
M
(1 v )
RTs
D
110 Chapter 3. Determining Structures and Analyzing Cells
BOX 3-C ISOTOPES IN BIOCHEMICAL INVESTIGATIONS
Both stable
a
and radioactive
b e
isotopes are
widely used in chemical and biological investiga-
tions. The study of metabolism was revolutionized
by the introduction of isotopic tracers. In one of the
first biological experiments with the stable isotope
15
N (detected by mass spectrometry), Schoenheimer
and associates in 1937 established the previously
unsuspected turnover of protein in living tissues
(Chapter 24, Section B). In 1937 Ruben et al. reported
the uptake of radioactive
11
CO
2
by plants.
f
A few
years later Calvin and associates first traced the
pathway of carbon in photosynthesis using the
much longer lived
14
CO
2
(Box 17-F). Wood and
Werkman, in 1941, employed the stable isotope
13
C
in studies of bacterial and mammalian metabolism
(Box 17-C). The radioactive
32
P and
35
S have served
to elucidate the metabolism of phosphorus and
sulfur. Titrium (
3
H) has been used to label many
organic substances including thymine, which has
been used extensively in the study of nucleic acid
metabolism. Radioactive isotopes provide the basis
for sensitive analytical procedures such as radio-
immunoassays of minute quantities of hormones
(Box 31-D). Through radioautography these iso-
topes facilitate numerous analytical procedures (see
accompanying photo) and have provided the basis
for important end-group methods used in sequence
determination of polynucleotides (Eq. 5-24).
Several isotopes used in biochemistry are listed
in the following table. For each radioactive isotope,
the half-life is given, as is the type of particle emit-
ted, and the energy of the particle. Gamma rays,
such as those given off in decay of
125
I or
131
I, are
very penetrating and easy to count precisely, as is
the energetic radiation from
32
P. On the other
hand,
3
H (tritium) is relatively difficult to detect
g
but its weak particle, which can travel only a
short distance through a sample, makes it uniquely
suitable for radioautography on a microscopic scale.
Positrons (
+
) travel some distance, e.g., up to a
few millimeters in the case of
13
N. They are then
destroyed by reacting with an electron to produce a
pair of rays of energy 0.511 MeV, equal to the sum
of the rest masses of an electron plus a positron.
The half-life (Eq. 9-4) determines the isotopic abun-
dance needed to achieve a given radiation rate, a
practical matter in providing a sufficient rate of
decay to permit counting with an acceptably low
statistical error. Even very short-lived isotopes such
as
13
N, have proved useful as tracers.
h
The amount
of an isotope giving 3.7 x 10
10
disintegrations per
second (this is 1 g of pure radium, 0.3 mg of
3
H, or
0.22 g of
14
C) is known as the curie (Ci). One milli-
curie (mCi) provides 2.22 x 10
9
disintegrations / min
(dpm). Radiolabeled substances ordinarily contain
only a small fraction of the unstable isotope together
with a larger number of unlabeled molecules. Com-
pounds are usually sold in millicurie or microcurie
quantities and with a stated specific activity as mCi
mmol
1
. For example, a compound labeled at a single
position with
3
H and having a specific activity of
50 mCi mmol
1
would contain about 0.17%
3
H at
that position.
Because of the development of new NMR tech-
niques and improvements in mass spectrometry
stable isotopes, such as
2
H,
13
C,
19
F, and
31
P, are
being used more frequently to study metabolism.
j
Carbon-13 containing compounds can fulfill many
Maximum energy
of radiation

(MeV)
Isotope Half-life
2
H (deuterium) Stable
3
H (tritium) 12.26 years 0.018
11
C 20.4 min (
+
)
0.511
13
C Stable
14
C 5730 years 0.156
13
N 9.96 min 1.2 (
+
)
0.511
15
N Stable
15
O 20.4 min (
+
)
0.511
18
O Stable
18
F 110 min (
+
)
0.511
22
Na 2.6 years 0.55
1.28
32
P 14.3 days 1.71
35
S 87.2 days 0.167
36
Cl 3 x 10
5
years 0.716
40
K 1.3 x 10
9
years 1.4
1.5
45
Ca 165 days 0.26
59
Fe 45 days 0.46
1.1
65
Zn 250 days 0.32
1.14
90
Sr 29 years 0.54
125
I 60 days 0.036
131
I 8.06 days 0.61
0.36
111
BOX 3-C ISOTOPES IN BIOCHEMICAL INVESTIGATIONS (continued)
of the same tracer functions as
14
C. Even the radio-
active
3
H nucleus can be utilized for in vivo NMR.
k,l
Although radioisotope labeling is very sensitive, it
gives little information unless compounds are iso-
lated and laboriously degraded to determine the
positions of the labels. NMR spectroscopy is less
sensitive but can give direct chemical information
about the positions of
13
C in compounds within
living cells. A compound containing only
13
C in one
or in many positions can be safely administered to
human individuals as well as to other organisms
and spectra of products that arise can be observed.
High-resolution deuterium NMR spectroscopy has
been used to follow
2
H incorporated at C-1, C-2, or
C-6 positions in glucose.
m
As a result of metabolic reactions an isotope
may appear at more than one position in a product,
yielding two or more isotope isomers or isotopomers.
These are seen individually by NMR spectroscopy
and the concentration and isotope labeling patterns
of the labeled compounds can be followed over a
period of time. The use of this isotopomer analysis
in studies of the citric acid cycle is illustrated in Box
17-C and its use in studies of glucose metabolism is
considered in Chapter 17, Section L.
A change in isotopic mass, especially from
1
H to
2
H or
3
H, often produces a strong effect on reaction
rates and the study of kinetic isotope effects has
provided many insights into the mechanisms of
enzymatically catalyzed reactions. Isotopes have
permitted a detailed understanding of the stereo-
Radioautogram showing the separa-
tion of proteins of E. coli labeled with
14
C amino acids. From OFarrell.
i
Twenty-five l of sample containing
180,000 cpm and ~10 g of protein
were subjected to isoelectric focusing
in a 2.5 x 130-mm tube containing
polyacrylamide gel to separate pro-
teins according to isoelectric point.
The gel was then extruded from the
column and was placed on one edge
of a slab of polyacrylamide gel. Then
SDS electrophoresis in the second
dimension separated the proteins
according to size. Over 1000 spots
could be seen in the original radio-
autogram, which was obtained by
placing a piece of photographic film
over the gel slab and exposing it to
the radiation for 875 hours. For
details see OFarrell.
i
chemistry of enzymatic reactions, an impressive
example being the synthesis and use of chiral acetate
(Chapter 13)
n
and chiral phosphate groups (Chapter
12). Specific isotopic properties provide the basis
for NMR (Section G).
a
Matwiyoff, N. A., and Ott, D. G. (1973) Science 181, 11251132
b
Wang, C. H., Willis, D. L., and Loveland, W. D. (1975) Radio-
tracer Methodology in the Biological, Environmental, and Physical
Sciences, Prentice-Hall, Englewood Cliffs, New Jersey
c
Wang, Y., ed. (1969) Handbook of Radioactive Nuclides, CRC Press,
Cleveland, Ohio
d
Thornburn, C. C. (1972) Isotopes and Radiation in Biology,
Butterworth, London
e
Slater, R. J., ed. (1990) Radioisotopes in Biology: A Practical
Approach, IRL Press, Oxford
f
Ruben, S., Hassid, W. Z., and Kamen, M. D. (1939) J. Am. Chem.
Soc. 61, 661663
g
Bransome, J., ed. (1970) Liquid Scintillation Counting, Grune &
Stratton, New York
h
Cooper, A. J. L. (1985) Adv. Enzymol. 57, 251356
i
OFarrell, P. H. (1975) J. Biol. Chem. 250, 40074021
j
Wolfe, R. R. (1992) Radioactive and Stable Isotope Tracers in
Biomedicine, Wiley, New York
k
Newmark, R. D., Un, S., Williams, P. G., Carson, P. J., Morimo-
to, H., and Klein, M. P. (1990) Proc. Natl. Acad. Sci. U.S.A. 87,
583587
l
Bergerat, A., Guschlbauer, W., and Fazakerley, G. V. (1991) Proc.
Natl. Acad. Sci. U.S.A. 88, 63946397
m
Aguayo, J. B., Gamcsik, M. P., and Dick, J. D. (1988) J. Biol.
Chem. 263, 1955219557
n
Cornforth, J. W. (1976) Science 193, 121125
C. Determining the Relative Molecular Mass, M
r
Isoelectric focusing
S
e
p
a
r
a
t
i
o
n

a
c
c
o
r
d
i
n
g

t
o

m
o
l
e
c
u
l
a
r

w
e
i
g
h
t

(
S
D
S

e
l
e
c
t
r
o
p
h
o
r
e
s
i
s
)

112 Chapter 3. Determining Structures and Analyzing Cells


This suggests a proportionality between and the
molecular radius. In fact, Eq. 3-21, in which a and b
are constants provides a fairly good approximation for
and V
e
is correlated approximately with log M
r
as
shown in Fig. 3-9.
193,194
A series of reference proteins of known molecular
masses are used to calibrate the column and M
r
for
an unknown protein is estimated from its position on
the graph.
195,196
Another modification of the method
depends upon chromatography in a high concentration
of the denaturing salt guanidinium chloride. The
assumption is made that proteins are denatured into
random coil conformations in this solvent.
196
Probably the most widely used method for deter-
mining the molecular mass of protein subunits is gel
electrophoresis in the presence of the denaturing
detergent sodium dodecyl sulfate (SDS). The protein
molecules are not only denatured but also all appear
to become more or less evenly coated with detergent.
197
The resulting rodlike molecules usually show a uniform
dependence of electrophoretic mobility on molecular
mass (plotted as log M
r
). An example is shown in Fig.
3-10. Again, the molecular mass of the protein under
investigation is estimated by comparison of its rate of
migration with that of a series of marker proteins.
195,198
3. Mass Spectrometry
Mass spectrometry has played a role in bio-
chemistry since the early 1940s when it was introduced
for use in following isotopic labels during metabo-
lism.
199200c
However, it was not until the 1990s that
suitable commercial instruments were developed to
permit mass spectrometry using two new methods
of ionization. The techniques are called matrix-
assisted laser desorption / ionization time-of-flight
(MALDI-TOF) and electrospray ionization (ESI)
mass spectrometry.
In the MALDI technique a pulsed laser beam
strikes a solid sample and heats, vaporizes, and ionizes
compounds with little decomposition.
201 209
Proteins
or other biopolymers are mixed with a matrix that
absorbs the heat of the laser beam. The protein sample
together with the matrix is dried. Most proteins form
crystals and the laser beam is directed toward individ-
ual protein crystals or aggregates. Various materials
are used for the matrix. Compounds as simple as
glycerol, succinic acid, or urea can be used with an
infrared laser. For proteins an ultraviolet nitrogen
laser tuned to 337 nm is usually employed with an
ultraviolet light-absorbing matrix such as hydroxy-
benzoic acid, 2,5-dihydroxybenzoic acid, -hydroxy-
2. Gel Filtration and Gel Electrophoresis
Several newer methods of molecular mass deter-
mination were developed in the 1960s1980s. One is
gel filtration. A column of gel beads such as Sephadex
is prepared carefully and is calibrated by passing a
series of protein solutions through it. The volume V
e
at which a protein peak emerges from the column can
be expressed as the sum of two terms (Eq. 3-19) in
which V
o
is the void volume, i.e., the elution volume
that is observed for very large particles that are com-
pletely excluded from the gel, and V
i
is the internal
volume within the beads of gel. The value of is
inversely related to the diffusion constant D, which
for a spherical particle D is related by Eq. 3-20 to the
Stokes radius r
h
. This equation comes directly from
Eqs. 3-16 and 3-14.
V V V
e o i
= + (3-19)
(3-20)
r
k T
D
h
B
=
6
= + a b
h
log r (3-21)
Figure 3-8 Plots of the logarithm of the sedimentation
constant s against the logarithm of the molecular weight for
a series of proteins and nucleic acids: ( ) globular proteins,
( ) RNA, and ( ) DNA. Proteins include a lipase (milk),
cytochrome c, ribonuclease (pancreatic), lysozyme (egg
white), follicle-stimulating hormone, bacterial proteases,
human hemoglobin, prothrombin (bovine), malate dehydro-
genase, -globulin (horse), tryptophanase (E. coli), glutamate
dehydrogenase (chicken), and cytochrome a. Double-
stranded DNA molecules are those of bacteriophage X174
(replicative form), T
7
,
b2
, T
2
, and T
4
, and that of a papilloma
virus. The RNA molecules are tRNA, rRNA, and mRNA of
E. coli, and that of turnip yellow mosaic virus.
190a,191a
10
4
10
5
10
6
10
7
10
8
10
9
Relative molecular mass, M
r
i
n

u
n
i
t
s

o
f

1
0

1
3
s
s
02
0
,
w
100
50
10
5
1
113
cinnamic acid, or sinapinic acid (Chapter 25). The
matrix ionizes, desorbs from the surface, and transfers
energy to the crystalline protein, causing it to ionize
and desorb from the surface. Oligosaccharides and
oligonucleotides can be ionized in a similar way.
MALDI spectra are relatively simple (Fig. 3-11),
often containing a single major peak corresponding to
the singly charged molecular ion [M + H]
+
of mass m + 1
and perhaps a doubly charged molecular ion [M + 2H]
2+
of mass (m + 2)/2. For oligomeric proteins the major
peak is often that of the monomer with weaker peaks
for oligomers. The instrument can also be adjusted to
generate negative ions whose detection is useful for
study of phosphorylated peptides, many oligosaccha-
rides, and oligonucleotides. With a TOF spectrometer
there is no upper limit to the mass range and masses
of over 100 kDa can be measured to about 0.1%.
Femtomole quantities can be detected.
The MALDI method is especially useful for complex
mixtures of peptides and can be utilized in peptide
sequencing. The technique is also appropriate for
studying mixtures of glycoproteins. Negative-ion
MALDI can be applied to oligonucleotide mixtures.
Further improvements in resolution in both MALDI
Glucagon
Cytochrome c
Myoglobin
Chymotrypsinogen
Ovalbumin
Malate dehydrogenase
E. coli phosphatase
Glyceraldehyde 3-phosphate dehydrogenase
Fumarase
Catalase
Ferritin
-Galactosidase
Apoferritin
250
230
210
190
170
150
130
110
90
70
Sucrose
Ovomucoid
Bovine serum albumin
Lactoperoxidase
Fetuin
Serum albumin dimer
Yeast alcohol dehydrogenase
Ceruloplasmin
-Globulins
R-Phycoerythrin
-Conarachin
Fibrinogen
Transferrin
Lactate dehydrogenase
Aldolase
Urease
-Crystallin Blue dextran
10
4
10
5
10
6
Relative molecular mass, M
r
V
e

(
m
l
.
)
3.0
2.0
2.5
1.5
1.0
V
e
/
V
O
Figure 3-9 Elution volume of various proteins on a column of Sephadex G-200 as a function of molecular mass. The right-
hand vertical axis shows the ratio of the elution volumes to that of blue dextran, a high-molecular-mass polysaccharide that is
excluded from the internal volume. After Andrews.
193
Figure 3-10 Estimation of the molecular mass of the poly-
peptide chain of the nitrogenase Fe-protein using SDS-poly-
acrylamide electrophoresis; from a set of four standard
curves. The marker proteins are (1) catalase, (2) fumarase,
(3) aldolase, (4) glyceraldehyde-phosphate dehydrogenase,
(5) -chymotrypsinogen A, and (6) myoglobin. () indicates
position of azoferredoxin. From Nakos and Mortenson.
195
4
5
6
1
2
3
Nitrogenase
Fe-protein
7
6
5
4
3
2
1
0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8
R
e
l
a
t
i
v
e

m
o
l
e
c
u
l
a
r

m
a
s
s
,

M
r


1
0

4
Electrophoretic mobility
C. Determining the Relative Molecular Mass, M
r
114 Chapter 3. Determining Structures and Analyzing Cells
and ESI methods are anticipated as a result of develope-
ment of Fourier transform mass spectrometers.
202
In ESI mass spectrometry
201,203 205,210 213
the sample,
dissolved in an appropriate solvent (usually a 50:50
mixture of methanol and water for proteins), is infused
directly into the ionization chamber of the spectrometer
through a fused silica capillary. At the end of the capil-
lary the solution is subjected to electrical stress created
by a voltage difference of about 5 kV between the elec-
trospray needle and the sampling orifice (the counter-
electrode). The process results in the formation of
singly and / or multiply charged molecular ions which
are guided into the analyzer for mass analysis. For
proteins every arginine, lysine, and histidine may bind
a hydrogen ion to form a variety of positive ions. A
100-kDa protein may easily bind 100 protons bringing
R
e
l
a
t
i
v
e

i
n
t
e
n
s
i
t
y
10
9
8
6
5
4
3
2
1
0
7
m/z 10
3
6 8 10 12 14 16 18 20 28 32 34 22 24 26 30
8095.6
7870.3
15716
31388
Figure 3-11 Matrix-assisted laser
desorption / ionization time-of-
flight (MALDI-TOF) mass spec-
trum of bovine erythrocyte Cu-Zn
superoxide dismutase averaged
over ten shots with background
smoothing. One-half l of solution
containing 10 pmol of the enzyme
in 5 mM ammonium bicarbonate
was mixed with 0.5 l of 50 mM -
cyanohydroxycinnamic acid
dissolved in 30% (v / v) of acetoni-
trile-0.1% (v / v) of trifluoroacetic
acid. The mixture was dried at 37
C before analysis. The spectrum
shows a dimer of molecular mass
of 31,388 Da, singly charged and
doubly charged molecular ions at
15,716, and 7870 Da, respectively.
The unidentified ion at mass
8095.6 may represent an adduct of
the matrix with the doubly
charged molecular ion. Courtesy
of Louisa Tabatabai.
Figure 3-12 Positive
ion electrospray mass
spectrum of horse
apomyoglobin (M
r
16,950.4). The net
charge on each ion as
well as the mass to
charge ratio m/z is
indicated at the top of
each peak. The inset
shows a computer
deconvolution of the
spectrum with the
calculated value of
molecular mass. Cour-
tesy of Kamel Harrata.
16952.9 2.6
16.4 17.0 17.6
M 10
3
100
80
60
40
20
0
100
80
60
40
20
0
R
e
l
a
t
i
v
e

a
b
u
n
d
a
n
c
e
+25
679.4
+24
707.4
+22
771.9
+21
808.6
+20
848.7
+19
893.3
+17
997.9
+15
1130.8
+14
1211.7
+13
1304.8
+11
1542.2
+10
1695.9
+12
1413.8
+9
1884.5
600 800 1000 1200 1400 1600 1800
m/z
115
linkages must be broken even to get the protein into
solution. Oxidation with performic acid (Eq. 3-22) has
been used on ribonuclease but is not often employed
because the performic acid also oxidizes tryptophan
residues.
Reduction by dithiothreitol or dithioerythritol
(Eq. 3-23) is usually successful.
Upon oxidation these dithiols cyclize to form stable
disulfides, driving the reaction to completion. These
same compounds are also widely used to protect SH
groups in enzymes against accidental oxidation by
oxygen and to dissolve highly crosslinked insoluble
proteins. Mercaptoethanol, HS CH
2
CH
2
OH, may
be used for the same purposes but requires higher
concentrations and has a disagreeable odor.
Once cleaved, disulfide bridges may be prevented
from reforming by conversion of the resulting thiol
groups to stable derivatives, e.g., with iodoacetate and
iodacetamide (Eq. 3-24).
A better reagent is acrylonitrile (Eq. 3-25).
D. Determining Amino Acid Composition and Sequence
the m/z ratio for the fully charged protein to 1000,
well below the maximum m/z ratio of ~ 2400 for a
typical quadrupole mass spectrometer. Since not all
basic groups are protonated the spectra consist of
families of peaks of differing m/z (Fig. 3-12). For a
single pure protein the molecular mass can be calculated
from the ratio of m/z values from any pair of adjacent
peaks. It is better, especially if there is a mixture of
proteins, to use a computer to extract M
r
.
214
The accu-
racy can be quite high, typically 0.01%: one mass unit
in 10,000.
Some complexity arises from the fact that each
carbon atom in a protein contains about 1% of
13
C.
This means that for a protein of mass > 10 kDa there
will be a confusing array of peaks in the mass spectrum
and it may be difficult to pick out the relatively minor
monoisotopic peak that arises from molecules con-
taining only
12
C,
1
H,
14
N,
16
O, and
32
S. In fact, the peak
representing the most abundant mass will be a few
mass units higher than the monoisotopic peak.
215,216
(see Study Question 15). New computer programs
have been devised to assist in the analysis. Use of
13
C
and
15
N-depleted nutrients also extends the applica-
bility of mass spectrometry.
217
Electrospray mass spectrometry utilizes a soft
ionization technique at nearly atmospheric pressure.
As a result, intact molecular ions are formed in high
yield. The instrument can be interfaced readily to
HPLC or capillary electrophoresis columns and sub-
femtomole amounts of proteins can be detected. A
disadvantage is that salt concentrations must be kept
low (< mM) and that the protein tends to bind Na
+
, K
+
,
and anions that may confuse interpretation of spectra.
D. Determining Amino Acid Composition and
Sequence
When they are first isolated, proteins are usually
characterized by M
r
, isoelectric point, and other easily
measured properties. Among these is the amino acid
composition
112
which can be determined by completely
hydrolyzing the protein to the free amino acids. Later,
it is important to establish the primary structure or
amino acid sequence.
51
This has been accomplished
traditionally by cutting the peptide chain into smaller
pieces that can be characterized easily. However, most
protein sequences are now deduced initially from the
corresponding DNA sequences, but further chemical
characterization is often needed.
1. Cleavage of Disulfide Bridges
Before a polypeptide chain can be degraded it is
usually necessary to break any disulfide bridges.
218 220
For some proteins such as the keratins of hair, these
CH
2
SH
H
I
I CH
2
COO
P
S CH
2
P
( CH
2
CONH
2
)
+

CH
2
COO
(I CH
2
CONH
2
)
+
(3-24)
C
C
CH
2
S
S
CH
2
HO
H
H
HO
C
C
CH
2
CH
2
HO
H
H
HO
SH
SH
S
S
CH
2
CH
2
Protein 1
Protein 2
2 Protein CH
2
SH
Dithiothreitol
(3-23)
CH
2
S P
1
S CH
2
P
2
H C
O
O OH
P
1
CH
2
SO
3

+ P
2
CH
2
SO
3

(3-22)
116 Chapter 3. Determining Structures and Analyzing Cells
R NH
3
O
O
O
R N C
O
CH CH COO
+
H
Lysine
side chain
2H
+

+
(3-26)
BOX 3-D THE PROTEOME
The ability to separate rapidly and detect
minute amounts of proteins has spawned a new
concept: The proteome is envisioned as a record
of all proteins being actively synthesized by a cell
or of all genes being actively expressed.
a c
The concept evolved from efforts to automate the
cataloging of spots on two-dimensional gels such
as that shown in Box 3-B where each spot repre-
sents a single protein. The ability to unambigu-
ously identify the spots by mass spectroscopy has
brought new optimism to the attempt to use gels
automatically to analyze all of the proteins
formed by a cell.
a,c
The methods are potentially
very important to developing new diagnostic
procedures for human medicine. Watching
changes in the proteome, including posttransla-
tional modifications in proteins, as cells develop
and grow will provide new insights into bio-
chemical regulation.
A similar concept is that of a complete tran-
scriptional map, a record of all of the different
RNA molecules being synthesized by a cell.
d,e
These include many different mRNAs, each of
which may give rise to more than one protein, as
well as many RNAs with other functions.
a
Kahn, P. (1995) Science 270, 369 370
b
Swinbanks, D. (1995) Nature (London) 378, 653
c
Shevchenko, A., Jensen, O. N., Podtelejnikov, A. V., Sagliocco,
F., Wilm, M., Vorm, O., Mortensen, P., Shevchenko.A,
Boucherie, H., and Mann, M. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1444014445
d
Richard, G.-F., Fairhead, C., and Dujon, B. (1997) J. Mol. Biol.
268, 303321
e
Schena, M., Shalon, D., Heller, R., Chai, A., Brown, P. O., and
Davis, R. W. (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 10614
10619
In modern sequencing methods vinylpyridine,
which reacts in a similar way, is often used. It can be
detected during amino acid analysis or sequencing
after derivatization with phenylisothiocyanate (Eq. 3-30).
2. Hydrolysis and Other Chain Cleavage
Reactions
Most biopolymers are inherently unstable with
respect to cleavage to monomer units by reaction with
water. Hydrolysis can be catalyzed by protons, by
hydroxyl ions, or by the protein-hydrolyzing enzymes
that are discussed in Chapter 12. Complete hydrolysis
of proteins is usually accomplished by heating under
nitrogen with 6 M HCl at 150C for 65 min. Some
amino acids, especially tryptophan, are destroyed
and the amides in the side chains of asparagine and
glutamine are converted to the free acids. Some pep-
tide linkages such as Val-Val are very resistant and
tend to be incompletely hydrolyzed. No procedure
has been found which gives the ideal complete hydro-
lysis. Use of 4 M methanesulfonic acid containing
3-(2-aminoethyl)indole instead of 6 M HCl gives less
decomposition of tryptophan.
221
Base-catalyzed hydro-
lysis of proteins also gives good yields of tryptophan
but causes extensive racemization of amino acids.
222
Complete enzymatic digestion of proteins can be
accomplished with a mixture of enzymes including
proteases produced by fungi (Pronase). However, the
enzymes attack each other, making quantitative analy-
sis difficult. The problem can be circumvented by
immobilizing the hydrolytic enzymes in a column of
agarose gel. The protein to be hydrolyzed
is passed through the gel and the constituent amino
acids emerge from the bottom of the column.
223,224
Selective enzymatic hydrolysis. The traditional
strategy in sequence determination is to cut protein
chains into smaller pieces which can be separated by
chromatography or electrophoresis and sequenced
individually. Enzymatic cleavage is especially useful
because of its specificity. Trypsin, a so-called endo-
P CH
2
S CH
2
CH
2
CN
P CH
2
SH
H
2
C CH CN
Acrylonitrile
(3-25)
H
2
C CH N
Vinylpyridine
peptidase, cleaves peptide chains at a rapid rate only
if the carbonyl group of the amide linkage cleaved is
contributed by one of the basic amino acids lysine,
arginine, or aminoethylcysteine (see Fig. 12-10). If the
protein is treated with maleic anhydride (Eq. 3-26),
117
the lysine residues are protected and trypsin will
cleave only at the Arg-X positions. If the resultant
peptides are separated and held at pH 3.5 overnight,
the blocking groups are hydrolyzed off and a second
trypsin treatment can be used to cleave at the Lys-X
positions.
225
The number of cleavage sites for trypsin
can be increased by converting an SH group to a
positively charged one by aminoethylation (Eq. 3-27).
226
The reaction can be accomplished either with ethyl-
eneimine (caution: carcinogen) as shown in this equa-
tion or by bromoethylamine, which eliminates Br

to
form ethyleneimine.
Because of its specificity for basic residues, trypsin
converts a protein into a relatively small number of
tryptic peptides which may be separated and charac-
terized. Trypsin acts primarily on denaturated proteins,
and to obtain good results the disulfide bridges must
be broken first. Chymotrypsin is less specific than
trypsin and pepsin is even less specific (Table 3-2).
Nevertheless, they can be used to cut a peptide chain
into smaller fragments whose sequences can be deter-
mined. To establish the complete amino acid sequence
CH
2
SH CH
2
NH
2
CH
2
CH
2
S CH
2
CH
2
NH
3
+
+
+
(3-27)
for a protein, overlapping peptide fragments must
be found that contain sequences from ends of two
different tryptic fragments. In this way the tryptic
peptides can be placed in the order in which they
occurred in the native protein. This tedious procedure
is rarely used today. Peptide sequencing is still impor-
tant but is usually coordinated with gene sequencing,
X-ray structure determination, or mass spectroscopy
which minimize the need for overlapping fragments.
While trypsin cuts the peptide linkages Lys-X and
Arg-X, a fungal protease cleaves only X-Lys.
227
A pro-
tease from the submaxillary glands of mice cleaves
only Arg-X,
228
one from Staphylococcus specifically at
Glu-X,
229,230
and one from kidneys at Pro-X.
231
Several enzymes catalyze stepwise removal of
amino acids from one or the other end of a peptide
chain. Carboxypeptidases
232
remove amino acids
from the carboxyl-terminal end, while aminopeptidases
attack the opposite end. Using chromatographic
methods, the amino acids released by these enzymes
may be examined at various times and some idea of
the sequence of amino acids at the chain ends may be
obtained. A dipeptidyl aminopeptidase from bovine
spleen cuts dipeptides one at a time from the amino
terminus of a chain. These can be converted to volatile
trimethylsilyl derivatives and identified by mass spec-
trometry.
233
If the chain is shortened by one residue
using the Edman degradation (Section 3) and the
dipeptidyl aminopeptidase is again used, a different
set of dipeptides that overlaps the first will be obtained
and a sequence can be deduced. Carboxypeptidase Y
can be used with MALDI mass spectrometry to deduce
the C-terminal amino acid sequence
for a peptide. However, Ile and Leu
cannot be distinquished.
Nonenzymatic cleavages. Of
the various nonenzymatic methods
that have been proposed, one has
been outstandingly useful. Cyano-
gen bromide, N C Br, cleaves pep-
tide chains adjacent to methionine
residues. The sulfur of methionine
displaces the bromide ion (Eq. 3-28)
and because of a favorable spatial
relationship, the resulting sulfonium
compound undergoes C S bond
cleavage through participation of the
adjacent peptide group (Eq. 3-28,
step b). The C = N of the product is
then hydrolyzed with cleavage of
the peptide chain in step c.
The linkage Asp-Gly can often
be cleaved specifically by treatment
with hydroxylamine at high pH.
195
Procedures for specific cleavage of
tryptophanyl bonds have been de-
D. Determining Amino Acid Composition and Sequence
TABLE 3-2
Specificities of Commonly Used Protein-Hydrolyzing Enzymes
Trypsin Lys-X, Arg-X X not Pro
Chymotrypsin
rapidly: Phe-X, Tyr-X, Trp-X X not Pro
slowly: Y-X X not Pro
Y=Leu, Asn, Gln, His, Met, Ser, Thr
Staphylococcus aureus
protease V-8 Glu-X X not Pro
Clostripain Arg-X
Pepsin
preferentially: X-Phe-X, X-Tyr-X, X-Leu-X
less so: X-Ala-X
Thermolysin
rapidly: X-Y
Y=Ile, Leu, Val, Ala, Phe, Met
slowly: X-Y
Y=Tyr, Gly, Thr, Ser
118 Chapter 3. Determining Structures and Analyzing Cells
vised.
234
The Asp-Pro linkage is susceptible to cleavage
by trifluoroacetic acid, which is used in the automated
Edman degradation employed in peptide sequencing
(Eq. 3-30). Cleavage by trifluoroacetic acid can be
used to generate peptides for subsequent sequence
determination.
Separating the peptides. A procedure that has
been very important in the development of protein
chemistry is peptide mapping or fingerprinting.
The procedure begins with cleavage of the disulfide
linkages, denaturation, and digestion with trypsin or
some other protease. The sizes and amino acid com-
positions of the resulting series of peptides are charac-
teristic of the protein under study. The mixture of
peptides is placed on a thin layer plate and subjected
to chromatography in one direction, then to electro-
phoresis in the other direction, with the peptides
separating into a characteristic pattern or fingerprint.
Fingerprinting has been especially useful in searching
for small differences in protein structure, for example,
between genetic variants of the same protein (Fig. 7-27).
Currently, the peptides are usually separated on ion
exchange or gel filtration columns, by reversed-phase
HPLC, or by capillary electrophoresis and are then
often passed, in subpicomole amounts, into a mass
spectrometer.
176,235,236
N C
C
N
O
CH
2
CH
2
S
CH
3
H
H
H
Br C N
N C
C
N
O
CH
2
CH
2
H
H
H
S
CH
3
C N
+
S C N CH
3
O
CH
2
CH
2
C
C
N
N
H
H
H
+
O
CH
2
CH
2
C
C
O
N
H
H
Methionine residue
in protein
Cyanogen bromide
a
b
Peptidyl homoserine lactone
Br

c
H
2
N
(3-28)
3. Determining Amino Acid Sequence
The covalent structure of insulin was established
by Frederick Sanger in 1953 after a 10-year effort. This
was the first protein sequence determination.
237,238
Sanger used partial hydrolysis of peptide chains
whose amino groups had been labeled by reaction
with 2,4-dinitrofluorobenzene
239
to form shorter end-
labeled fragments. These were analyzed for their
amino acid composition and labeled and hydrolyzed
again as necessary. Many peptides had to be analyzed
to deduce the sequence of the 21-residue and 30-
residue chains that are joined by disulfide linkages in
insulin.
237,238
The Sanger method is mainly of historic interest,
although end-labeling may still be used for various
purposes. A more sensitive labeling reagent than was
used by Sanger is dansyl chloride. It reacts to form a
sulfonamide linkage that is stable to acid hydrolysis
and is brilliantly fluorescent (Eq. 3-29). The related
reagent dimethylaminoazobenzene-4'-sulfonyl chlo-
ride gives highly colored derivatives easily seen on
thin-layer chromatography plates.
240
The Edman degradation. One of the most im-
portant reagents for sequence analysis is phenyl-
isothiocyanate, whose use was developed by P.
Edman.
241 243
This reagent also reacts with the N-
terminal amino group of peptides (Eq. 3-30, step a).
The resulting adduct undergoes cyclization with
cleavage of the peptide linkage (Eq. 3-30, step b) under
acidic conditions. After rearrangement (step c) the
resulting phenylthiohydantoin of the N-terminal
amino acid can be identified. The procedure can then
be repeated on the shortened peptide chain to identify
the amino acid residue in the second position. With
careful work the Edman degradation can be carried
down the chain for several tens of residues.
Ingenious protein sequenators have been de-
vised to carry out the Edman degradation automati-
cally.
242,244 246
Each released phenylthiohydantoin is
then identified by HPLC or other techniques. Com-
mercial sequenators have often required 5 20 nmol of
peptide but new microsequenators can be used with
amounts as low as 5 10 picomoles or less.
247,248
N
CH
3
H
3
C
Protein NH
2
N
CH
3
H
3
C
S
O
O NH Protein
Dansyl chloride
(5-Dimethylaminoaphthyl-
sulfonyl chloride)
SO
2
Cl
(3-29)
119
Microsequencers permit sequence analysis on minute
amounts of protein. Microsequencing can be used in
conjunction with two-dimensional electrophoretic
separations of proteins such as that shown in Box 3-C.
The proteins in the polyacrylamide gel are electropho-
retically transferred onto a porous sheet (membrane)
of an inert material such as polyvinyl difluoride.
249 251
After staining, a selected spot is cut out and placed
into the sequencer. To avoid the problems associated
with blocked N termini, the protein may be treated
with proteases on the membrane and the resulting
peptide fragments may then be separated on a narrow-
bore HPLC column and sequenced.
240
Because many proteins are modified at the N ter-
minus, blocking application of the Edman degrada-
tion, it would be useful to have a similar method for
sequencing from the C terminus. It has been difficult
to devise a suitable strategy, but there has been some
success.
252 254
a
b
c
N C S H
2
N CH
R
1
C
O
NH
NH C
N
S
C
H H
R
1
C NH
O
H
+
NH C
N
S
C
H
R
1
C O H
3
N
N C
N
O
S
R
1
H
+
Phenylisothiocyanate
Weak base
Phenylthiocarbamyl peptide
Anhydrous
acid
+
Aqueous or
methanolic HCl
Phenylthiohydantoin (PTH)
of N-terminal amino acid
H
Anilinothiazolinone
Peptide
Protein sequences from the genes. Complete
sequences of large numbers of genes have been deter-
mined and the corresponding sequences of proteins
can be read directly from those of the corresponding
genes. One method for sequencing a gene is to isolate
a specific messenger RNA, which does not contain
intervening sequences. A DNA copy (cDNA) is made
from the mRNA and is used to ascertain the sequence
of the encoded protein. The genomic DNA is also
often sequenced. Introns are recognized by the nucle-
otide sequences at their ends and the correct amino
acid sequence for the encoded protein is deduced.
In many instances, however, a gene can be identi-
fied only after part of the protein, often an N-terminal
portion has been sequenced. This knowledge permits
synthesis of an oligonucleotide probe that can be
used to locate the gene (Chapter 5). Nucleotide se-
quences can be verified by comparison with sequences
of tryptic or other fragments of a protein. Similarly,
protein sequences are often checked by sequencing
the corresponding genes as well as by study of X-ray
structures. Substantial numbers of errors are made in
sequencing of both DNA and protein so that checking
is important.
Mass spectrometry in sequencing. Proteins can
also be sequenced by mass spectrometry or by a com-
bination of Edman degradation and mass spectrome-
try.
213,255,255a
Until recently the peptides had to be con-
verted to volatile derivatives by extensive methylation
and acetylation or by other procedures. However,
newer ionization methods including MALDI (Fig. 3-11)
and ESI (Fig. 3-12) have made it possible to obtain
mass spectra on unmodified peptides. In one proce-
dure a nonspecific protease cleaves a peptide chain
into a mixture of small oligopeptides which are sepa-
rated by HPLC into 20 40 fractions, each of which
may contain 10 15 peptides but which can be sent
directly into the ionization chamber of the mass spec-
trometer.
256
Peptides can be generated from a protein
using immobilized enzymes, separated on a chromato-
graphic column, and introduced sequentially into the
mass spectrometer. Examination of peptide mixtures
by mass spectrometry provides a way of verifying
sequences deduced from DNA sequencing.
257
Mass
spectrometry is also used widely to study covalently
modified proteins.
173,216,257a,257b
As a rule, these cannot
be recognized from gene sequences.
4. Locating Disulfide Bridges
A final step in sequencing is often the location of
SS bridges. The reduced and alkylated protein can
be cleaved enzymatically (e.g., with elastase, pepsin,
or thermolysin) to relatively small fragments, each of
which contains no more than one modified cysteine.
(3-30)
D. Determining Amino Acid Composition and Sequence
120 Chapter 3. Determining Structures and Analyzing Cells
The same enzymatic cleavage can then be applied to
the unreduced enzyme. Pairs of peptide fragments
remain linked by the SS bridges. These crosslinked
pairs can be separated, the disulfide bridges cleaved,
and the resulting peptides identified, each as one of
the already sequenced fragments. Mass spectrometry
provides a rapid method for their identification.
258
Another elegant way of locating SS bridges
employs diagonal electrophoresis. Electrophoresis
of the digest containing the crosslinked pairs is con-
ducted in one direction on a sheet of filter paper. Then
the paper is exposed to performic acid vapor to cleave
the bridges according to Eq. 3-22 and electrophoresis
is conducted in the second direction and the paper is
sprayed with ninhydrin. The spots falling off the
diagonal are those that participated in SS bridge
formation. They can be associated in pairs from their
positions on the paper and can be identified with
peptides characterized during standard sequencing
procedures.
259
Diagonal electrophoresis and its relative diagonal
chromatography are useful for other purposes as well.
After electrophoresis or chromatography is conducted
in one direction, the paper or thin-layer plate may be
sprayed with a reagent that will react with some com-
ponents or may be irradiated with light before the sepa-
ration is repeated in the second direction (Fig. 3-5).
102,260
5. Detecting Products
Important to almost all biochemical activity is the
ability to detect, and to measure quantitatively, tiny
amounts of specific compounds. Color reagents,
which develop characteristic colors with specific com-
pounds, are especially popular. For example, ninhy-
drin (Box 3-E) can be used as a spray reagent to
detect a small fraction of a micromole of an amino acid
or peptide in a spot on a chromatogram. It can also be
used for a quantitative determination, the color being
developed in a solution. More sensitive than absorp-
tion of light (color) is fluorescence. Fluorescamine
(Eq. 3-31) reacts with any primary amine to form a
highly fluorescent product. As little as 50 pmol of
amino acid can be determined quantitatively.
261
A
yet more sensitive fluorogenic reagent for detection
of amino acids, peptides, and amines of all types is
o-phthaldialdehyde.
262,263
Reaction with naphthalene 2,3-dicarboxaldehyde (Eq.
3-32) increases the limit of detection 100-fold or
more.
264
Detection of proteins on thin-layer plates, gel slabs,
or membranes is often accomplished by staining with
a dye,
265 267
the most widely used being Coomassie
brilliant blue.
268
Various silver-containing stains may
also be used. After separation of a protein mixture by
electrophoresis and transfer to an inert membrane,
O
O
O
O
COOH
OH
N
O
RNH
2
R
"Fluorescamine"
(3-31)
CHO
CHO
(fluorescent)
o-Phthaldialdehyde
CHO
CHO
N
CN
R
RNH
2
CN

(3-32)
NaO
3
S
CH
2
N
NH
N
CH
2
SO
3
O
C
2
H
5
C
2
H
5
Coomassie brilliant blue

C
2
H
5
121
BOX 3-E NINHYDRIN
Ninhydrin (1,2,3-indantrione monohydrate)
forms Schiff bases (ketimines) with amino acids.
These react in ways similar to those of Schiff bases
of pyridoxal phosphate (Chapter 14). Decarboxyla-
tion of the ketimines followed by hydrolysis of the
resulting aldimines yields an intermediate amine
that can couple with a second molecule of ninhydrin
to form a characteristic purple color.
a,b
First reported
by Ruhemann in 1910, the intermediate amine can
also be hydrolyzed to free ammonia. Therefore, to
ensure maximum color yield ninhydrin solutions for
quantitative analysis usually contain reduced
ninhydrin, which can react with free NH
3
and
ninhydrin to form Ruhemanns purple (see scheme).
The reaction has been widely used in chromatogra-
phy and in quantitative amino acid analysis and
also as a convenient spray reagent for paper and
thin-layer chromatography. While -amino acids
react most readily, primary amines and peptides
also form Ruhemanns purple. In these cases a
proton rather than CO
2
is lost from the ketimine.
When pyridoxamine (Chapter 14) on chromato-
grams reacts, a bright orange product, presumably
the aldimine, appears. Secondary amines, such as
proline, give a yellow color.
Both the ninhydrin reaction and pyridoxal
phosphate-catalyzed decarboxylation of amino acids
(Chapter 14) are examples of the Strecker degrada-
tion. Strecker reported in 1862 that alloxan causes
the decarboxylation of alanine to acetaldehyde, CO
2
,
and ammonia.
c
Many other carboxyl compounds, e.g., those of the
general structure
and p-nitrosalicylaldehyde also cause the Strecker
degradation.
d
H
2
C NH
3
OH
OH
O
O
Ninydrin
O
O
O
O
O
N
H
C R

Aldimine
O
O
Ketimine
N
H
C
R
COO

O
H
C R
O
O
N

Ruhemanns purple
O
O
O
O
NH
2

Intermediate amine
+
NH
3
H
C R COO

CO
2
OH
H
O
O
Reduced ninhydrin, hydrindantin
(2-hydroxy-1,3-indanedione)
Ninhydrin
a
Wigfield, D. C., and Croteau, S. M. (1980) Biochem. Edu. 8,
2627
b
Friedman, M., and Williams, L. D. (1974) Bioorg. Chem. 3,
267280
c
Strecker, A. (1862) Annalen 123, 363365
d
Schonberg, A., and Moubacher, R. (1952) Chem. Rev. 50, 261
277
HN
N
O
O
O O
H
Alloxan
O C (CH CH)
4
C
O
D. Determining Amino Acid Composition and Sequence
122 Chapter 3. Determining Structures and Analyzing Cells
BOX 3-F BIOSENSORS AND ELECTRONIC NOSES
the resulting protein blots can be stained with spe-
cific antibodies.
269,270
Flame ionization detectors can
measure as little as a few picomoles of almost any
substance leaving a vapor-phase chromatographic
column. The importance of developing new, more
sensitive analytical methods by which the quantity of
material investigated can be scaled down can hardly
be overemphasized. Increasingly sensitive methods
of detection, including mass spectrometry, now permit
measurement of fmol (10
15
mol) quantities in some
cases. With this ability the output of neurotransmitters
from a single neuron in the brain can be measured and
the contents of single cells can be analyzed.
6. Absorption of Light
Side chains of the three aromatic amino acids
phenylalanine, tyrosine, and tryptophan absorb ultra-
violet light in the 240- to 300-nm region, while histidine
and cystine absorb to a lesser extent. Figure 3-13 shows
the absorption spectrum of a reference compound
for tyrosine. There are three major absorption bands,
the first one at 275 nm being a contributor to the well-
known 280-nm absorption band of proteins. There
is a much stronger absorption band at about 240 nm.
Sensitive methods for estimating protein concentration
depend upon the measurement of this absorption
together with that from other side chains at around 280
or 230 nm.
13,271 274
There is an even stronger absorption
band at 192 nm. However, at these wavelengths even
air absorbs light and experimental difficulties are
extreme. At 280 nm, and even more at 230 nm, it is
easy to contaminate samples with traces of light-
absorbing material invisible to the eye. Therefore,
most estimations of protein concentration from light
absorption depend upon the 280-nm band.
Figure 3-14 shows the spectra of N-acetyl ethyl
esters of all three of the aromatic amino acids and of
cystine. To a first approximation, the absorption spec-
tra of proteins can be regarded as a summation of the
spectra of the component amino acids. However, the
absorption bands of some residues, particularly of
tyrosine and tryptophan, are shifted to longer wave-
lengths than those of the reference compounds in
water. This is presumably a result of being located
within nonpolar regions of the protein. Notice that the
spectra for tyrosine, phenylalanine, and cystine in Fig.
A new approach to detection of molecules of
biological interest is the development of biosensors.
These are small devices that detect the binding of
specific molecules to a receptor which is in intimate
contact with a specially prepared surface that serves
as a transducer. The receptor might be a layer of
enzyme, antibody, hormone receptor, lectin, or
oligonucleotide. Binding of substrate, antigen,
hormone, sugar, or complementary polynucleotide
strand, respectively, induces a response consisting
of some kind of electrical or optical signal.
a d
If the
sensor is constructed on a semiconductor chip
changes in an imposed potential difference may be
detected.
a,e
However, changes in optical properties
are more often observed. Fluorescence of dyes
incorporated into the tranducing layer may be
induced by binding of a molecule to a protein that
undergoes an allosteric modification (see Chapter
9).
f
Many biosensors measure surface plasmon
resonance, a change in the evanescent wave that
develops in a surface when a light beam at the angle
of total reflectance strikes the surface. This induces
a change in the dielectric constant which can be
measured.
f m
Biosensors are used to estimate
binding constants and also rate constants. However,
read the article by Schuck and Milton
m
for tests of
the validity of kinetic data. Biosensors can serve as
electronic noses. One possible application is in
the analysis of compounds in human breath as an
aid to medical diagnosis. Over 400 volatile organic
compounds have been identified in breath using gas
chromatography and mass spectrometry.
n
a
Briggs, J. (1987) Nature (London) 329, 565 566
b
Zurer, P. (1997) Chem. Eng. News September 15, 7
c
Kress-Rogers, E., ed. (1997) Handbook of Biosensors and Electronic
Noses: Medicine, Food, and the Environment, CRC Press, Boca
Raton, Florida
d
Cunningham, A. J. (1998) Introduction to Bioanalytical Sensors,
Wiley, New York
e
McConnell, H. M., Owicki, J. C., Parce, J. W., Miller, D. L.,
Baxter, G. T., Wada, H. G., and Pitchford, S. (1992) Science 257,
1906 1912
f
Marvin, J. S., Corcoran, E. E., Hattangadi, N. A., Zhang, J. V.,
Gere, S. A., and Hellinga, H. W. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 4366 4371
g
Raether, H. (1988) Surface Plasmons, Springer Tracts in Modern
Physics, Vol. 111, Springer-Verlag, Berlin
h
Peterlinz, K. A., Georgiadis, R. M., Herne, T. M., and Tarlov, M.
J. (1997) J. Am. Chem. Soc. 119, 3401 3402
i
Hendrix, M., Priestley, E. S., Joyce, G. F., and Wong, C.-H.
(1997) J. Am. Chem. Soc. 119, 3641 3648
j
Salamon, Z., Brown, M. F., and Tollin, G. (1999) Trends Biochem.
Sci. 24, 213 219
k
Chao, H., Houston, M. E., Jr., Grothe, S., Kay, C. M., OConnor-
McCourt, M., Irvin, R. T., and Hodges, R. S. (1996) Biochemistry
35, 12175 12185
l
McNally, A. J., Mattsson, L., and Jordan, F. (1995) J. Biol. Chem.
270, 19744 19751
m
Schuck, P., and Minton, A. P. (1996) Trends Biochem. Sci. 21, 458
460
o
Phillips, M. (1992) Sci. Am. 267(July), 74 79
123
Figure 3-13 The absorption spectrum of N-acetyltyrosine ethyl ester in an aqueous
phosphate buffer of pH 6.8. Absorbance (as molar extinction coefficient, Eq. 23-5) is
plotted against increasing energy of light quanta in units of wave number. The more
commonly used wavelength scale is also given. Spectra are most often presented with
the low wavelength side to the left. In the convention adopted here the energy of a
quantum increases to the right. There are three * electronic transitions that give rise
to absorption bands of increasing intensity. The third * transition of the aromatic
ring is at ~ 52,000 cm
1
(192 nm) and reaches a molar extinction coefficient of ~ 40,000.
The n* and * transitions of the amide group in this compound also contribute to
the high energy end of the spectrum (see Chapter 23 for further discussion).
30 34 38 42 46 50
Wave number (cm
-1
) 10
-3
300 250 220 200
0
2
4
8
6
12
Wavelength (nm)
10
E
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t


1
0
-
3
(3-33)
3-14 have been multiplied by factors of 2 to 20. It is
evident that if all of the light-absorbing side chains
were present in equal numbers tryptophan would
dominate the absorption band and that phenylalanine
would contribute little except some small wiggles.
The molar extinction coefficient can be estimated
from the numbers of residues of each type per mole-
cule as follows:
274

280
(M
1
cm
1
) = 5500 (no. Trp) + 1490 (no. Tyr) +
125 (no. cystine)
For proteins of unknown composition, a useful ap-
proximation is that a solution containing 1 mg / ml of
protein has an absorbance at 280 nm of about 1.0.
E. Quantitative Determinations and
Modification Reactions of Side Chain
Groups
The functional groups present in the side chains
of proteins include NH
2
, SH, S S, OH, COO

,
the imidazole group of histidine, the guanidine group
of arginine, the phenolic group of tyrosine, the indole
ring of tryptophan, and the S CH
3
group of methio-
nine. These are able to enter into a great variety of
chemical reactions, most of which make use of the
nucleophilic properties of these groups. The reactions
are most often those of nucleophilic addition or nu-
cleophilic displacement. The basic chemistry of these
reactions often parallels biochemical reactions that are
discussed in Chapters 12 and 13. In many instances,
the reactions are nonspecific; amino, thiol, and hydroxyl
groups may all react with the same reagent. The use-
fulness of the reactions depends to a large extent on
the discovery of conditions under which there is some
selectivity. It is also important that the reactions be
complete. Only a few reactions will be considered here;
these and others have been reviewed by Glazer et al.
225,275
1. Reactions of Amino Groups
The numerous amino groups of lysine residues
and of the N termini of peptide chains usually pro-
Figure 3-14 The spectra of the first electronic transitions of
the N-acetyl derivatives of the ethyl esters of phenylalanine,
tyrosine, and tryptophan together with that of the dimethyl
ester of cystine in methanol at 25C. The spectra for the Tyr,
Phe, and cystine derivatives have been multiplied by the
factors given on the graph.
272
32 34 36 40 42 44
Wave number (cm
-1
) 10
-3
300 270 250 230
Wavelength (nm)
E
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t


1
0
-
3
38
0
1
3
5
4
9
8
7
6
2
Trp
Tyr 2
Phe 20
Cystine 4
E. Quantitative Determinations and Modification Reactions of Side Chain Groups
124 Chapter 3. Determining Structures and Analyzing Cells
R NH
2
CH
3
OH
R N C CH
3
H
NH
2
+
H
3
C C OCH
3
Methyl acetimidate
NH
2
+
(3-37)
R NH
2
CH
3
OH
C O CH
3
O-Methylisourea
NH
2
H
2
N
+
R N C NH
2
H
NH
2
+
(3-38)
trude into the aqueous surroundings of a protein.
Chemical modification can be done in such a way as
to preserve the net positive charge which amino groups
carry at most pH values, to eliminate the positive
charge leaving a neutral side chain, or to alter the charge
to a negative value. Alterations of these charges can
greatly affect interactions of the protein molecules
with each other and with other substances.
Amino groups react reversibly with carbonyl
compounds to form Schiff bases.
275 277
Reduction of
the latter by sodium borohydride or sodium cyano-
borohydride causes an irreversible change (Eq. 3-34,
steps a and b). Cyanoborohydride is specific for Schiff
bases and does not reduce the carbonyl compound.
However, side products may cause problems.
277,278
Depending upon which carbonyl compound is used,
the net positive charge on the amino group may be
retained or it may be replaced with a different charge
by this reductive alkylation sequence. Formalde-
hyde will react according to Eq. 3-34 in two steps to
give a dimethyl amino group with no change of net
charge.
279
Pyridoxal phosphate (Chapter 14) is con-
verted by Eq. 3-34 into a fluorescent label. With a
limited amount of pyridoxal phosphate only one or
a few lysine residues may be labeled, often at active
centers of enzymes. Schiff bases formed from glycer-
aldehyde in Eq. 3-34 can undergo the Amadori rear-
rangement (Eq. 4-8) to form stable products which,
however, can be reconverted to the original amino
groups upon acid hydrolysis. The borohydride reduc-
tion product of Eq. 3-34 with glyceraldehyde can be
reconverted to the original amine by periodate oxida-
tion (Eq. 4-11).
277
a
b
c
d
R NH
2
R' CHO
R N C
H
R'
R N
H
CH
2
R'
R N
C
CH
2
H
R'
R'
+
R N
CH
2
CH
2
R'
R'
+
H
Schiff base
NaBH
4
or NaCNBH
4
R' CHO
NaBH
4
For formaldehyde
R=H
(3-34)
Another addition reaction of amino groups is
carbamoylation with sodium cyanate (Eq. 3-35). A
displacement reaction by an amino group on an acid
anhydride such as acetic anhydride (Eq. 3-36) leads to
acylation, a nonspecific reaction which is also under-
gone by thiol, hydroxyl, and other groups. When
acetic anhydride is used, the net positive charge of an
amino group is lost. However, the product obtained
with succinic anhydride or maleic anhydride (Eq. 3-26)
carries a negative charge. In the latter case, the modifi-
cation can readily be reversed by altering the pH.
Both amidination (Eq. 3-37) and guanidination
(Eq. 3-38) lead to retention of the positive charge.
R NH
2
N
C
O
R N C
O
NH
2
H
Cyanate
H
+

(3-35)
R NH
2
H
3
C C O
R N C
O
CH
3
O
C CH
3
H
O
Acetic anhydride
CH
3
COOH
(3-36)
125
Bifunctional imidoesters such as dimethylsuberimidate
may be used to establish whether or not two different
proteins or subunits are close together in a complex or
in a supramolecular structure such as a membrane or
ribosome.
Another useful reaction of amino side chains is that
with dansyl chloride (Eq. 3-29). Many lysine deriva-
tives can be determined quantitatively by amino acid
analysis.
280
2. Reactions of SH Groups
In addition to the alkylation with iodoacetate (Eq.
3-24), sulfhydryl groups can react with N-ethylmale-
imide (Eq. 3-39).
281
This reaction blocks the SH groups
irreversibly and has often been used in attempts to
establish whether or not a thiol group plays a role in
the functioning of a protein. Loss of function in the
presence of this sulfhydryl reagent may mean that
an SH group has an essential role or it could be a result
of the bulk of the group added. The N-ethylmaleimide
group is large and could prevent proper contact between
an enzyme and substrate or between two proteins. To
avoid the possible effect of excessive bulk, it is useful
to convert the SH to the small thiocyanate group SCN
(Eq. 3-40).
282
Ellmans reagent 5,5'-dithiobis(2-nitrobenzoic acid;
DTNB) reacts quantitatively with SH groups (Eq. 3-41)
to form mixed disulfides with release of a thiolate anion
that absorbs light at 412 nm with a molar extinction
H
3
C O C (CH
2
)
6
NH
2
C O CH
3
NH
2
Dimethylsuberimidate
+ +
coefficient of 14,150 M
1
cm
1
.
283
While DTNB has
been widely used to determine the content of SH
groups in proteins, there are some disadvantages.
Pyridyldisulfides such as 2-pyridyldisulfide or the
isomeric 4-pyridyldisulfide react more completely and
with greater selectivity.
281
Thiol groups have a high affinity for mercury
ions including organic mercury derivatives, which
are widely used in the determination of protein struc-
tures by X-ray crystallography (Section F). Titration
of SH groups in proteins is often accomplished with
E. Quantitative Determinations and Modification Reactions of Side Chain Groups
N
O
O
R SH
CH
2
CH
3
N
O
O
CH
2
CH
3
S R
(3-39)
R SH
SCN
NO
2
COO
R SCN
S
NO
2
COO

(3-40)
(3-41)
O
2
N S S NO
2
S NO
2
S R S NO
2

Ellman's reagent
+
COO COO
R SH
Thiol anion
H
COO COO

+

N
S S
N
126 Chapter 3. Determining Structures and Analyzing Cells
p-mercuribenzoate (Eq. 3-42). The reaction may be
followed spectrophotometrically at 250 255 nm, a region
in which the mercaptide product absorbs strongly.
3. Reactions of Other Side Chains
There are no highly selective reactions for OH,
COO

, or imidazole groups. However, some hydroxyl


groups in active sites of enzymes are unusually reactive
in nucleophilic addition or displacement and can be
modified by acylation, phosphorylation, or in other
ways. Carboxyl groups, which are exceedingly nu-
merous on protein surfaces, can be modified by treating
with a water-soluble carbodiimide (Eq. 3-10) in the
presence of a high concentration of an amine such as
the ethyl ester of glycine. The imidazole groups of
residues of histidine can often be selectively destroyed
by dye-sensitized photooxidation (Ch. 12, Section D,5)
or can be acylated with ethoxyformic anhydride.
Compounds with two adjacent carbonyl groups
such as 1,2-cyclohexanedione (Eq. 3-43) react selectively
with guanidinium groups from arginine residues in
proteins. Under certain conditions the product indi-
cated in Eq. 3-43 predominates. Related reagents are
derived from camphorquinone.
284
The phenolic group of tyrosine undergoes iodina-
tion (Eq. 3-44), acylation, coupling with diazonium
compounds, and other reactions.
The following sparingly soluble chloroamide
together with I

will also iodinate tyrosine and can


be used to incorporate radiolabeled iodine into pro-
teins.
285,286
Tetranitromethane reacts slowly with tyrosyl
groups to form 3-nitrotyrosyl groups (Eq. 3-45). The
by-product nitroform is intensely yellow with
350
=
14,400. The reagent also oxidizes SH groups and
reacts with other anionic groups.
Koshland devised the following reagent for the indole
rings of tryptophan residues (Eq. 3-46).
Imidazole, lysine amino groups, and tyrosine
hydroxyl groups react with diethylpyrocarbonate
R N C
NH
2
NH
2
O O
HN
N
NH
R
HO OH
H
+
+
(3-42)
(3-43)
OH OH
I
2
I
H
+
I
pH 89

+
(3-44)
COO Hg
R SH
COO Hg R S
H
+
+

C C
N
C
N
N
C
N
O
O
Cl Cl
Cl Cl
1,3,4,6-Tetrachloro-3,6-diphenylglycouril
O
2
N C
NO
2
NO
2
NO
2
O
O
2
N C
NO
2
H
NO
2
O
NO
2
Nitroform

(3-45)
CH
2
HO NO
2
Br
R indole
N
R
CH
2
OH
NO
2
(3-46)
127
be 98% or better for every step. Even so, it is still
impractical to synthesize very large peptides. Those
that have been made, such as the hormone insulin and
the enzyme ribonuclease, have been obtained in low
yields and have been difficult to purify.
298
It is usually
more practical to obtain large peptides from natural
sources. It is often practical to clone a suitable piece
of DNA in a bacterial plasmid and to set up biological
production of the desired peptide (Chapter 26). On
the other hand, for smaller peptides, laboratory syn-
thesis is feasible. Even for large peptides it is useful
because it permits incorporation of unnatural amino
acids as well as isotopic labels.
The general procedure for making a peptide in
the laboratory is to block the amino group of what
will become the N-terminal amino acid with a group
that can be removed later. The subsequent amino
acid units activated at their carboxyl end are then
attached one by one. The chemical activation is often
accomplished by conversion of the carboxyl group of
the amino acid to an anhydride. At the end of the
synthesis, the blocking group must be removed from
the N terminus and also from various side chain groups
such as those of cysteine and lysine residues. In many
respects, this procedure is analogous to the biological
synthesis of proteins whose basic chemistry is dis-
cussed in Chapter 29.
1. Solid-Phase Peptide Synthesis
Modern methods of peptide synthesis began with
the solid-phase method introduced by Merrifield
299
in 1962 (Fig. 3-15). To begin the synthesis a suitably
protected amino acid is covalently linked to a polysty-
rene bead. The blocking t-butoxycarbonyl (Boc) group
is removed as isobutene by an elimination reaction to
give a bound amino acid with a free amino group.
This can then be coupled to a second amino acid with
a blocked amino group using dicyclohexylcarbodiimide
(Eq. 3-10). The removal of the blocking group and
addition of a new amino acid residue can then be
repeated as often as desired. The completed peptide
is removed from the polystyrene by action of a strong
acid such as HF.
Advantages of the Merrifield procedure are that
the peptide is held tightly and can be washed thor-
oughly at each step. Problems arise from repeated use
of trifluoroacetic acid and the need to use HF or other
strong acid to cleave the peptide from the matrix and
also to remove blocking benzyl groups that must be
present on many side chain groups. Newer variations
of the procedure include a more labile linkage to a
polyamide type of polymer and use of blocked amino
acids.
297,300301a
These active esters will spontaneously
condense with the free amino group of the growing
peptide and with suitable catalysis will eliminate
(Eq. 3-47) at low enough pH (below 6) that the reaction
becomes quite selective for histidine.
70,287
Reactivity
with this reagent is often used as an indication of his-
tidine in a protein.
288 290
. The reaction may be moni-
tored by observation of NMR resonances of imidazole
rings.
288,290
The thioether side chains of methionine units in
proteins can be oxidized with hydrogen peroxide to
the corresponding sulfones (Eq. 3-48). They can also
be alkylated, e.g., by CH
3
I to form R S
+
(CH
3
)
2
.
4. Affinity Labeling
To identify groups that are part of or very near to
the active site of a protein, reagents can be designed
that carry a reactive chemical group into the active
site.
291
The related photoaffinity labeling
292,293
is
also widely used (see also Chapter 23).
F. Synthesis of Peptides
The synthesis of peptides of known sequence in
the laboratory is extremely important to biochemical
research. For example, we might want to know how
the effects of a peptide hormone are altered by replace-
ment of one amino acid in a particular position by
another. The synthetic methods must be precise
294 298
and because there are so many steps the yield should
F. Synthesis of Peptides
(3-47)
R S CH
3
R S
O
CH
3
O
+ 2H
2
O
2
2H
2
O +
(3-48)
N
N
Protein
H
5
C
2
O
C O
C
O
C
2
H
5
H
O
O
H
5
C
2
O
C O
C
N
O
O
+
Diethyl pyrocarbonate
NH
Protein
H
+
C
2
H
5
OH
128 Chapter 3. Determining Structures and Analyzing Cells
Blocked amino acid
C
H
F
F F
O
F F
C
C
N
C
O
CH
2
O
O
H
R H
pentafluorophenol. The fluorenylmethoxycarbonyl
(Fmoc) blocking group is removed under mildly basic
conditions. The whole procedure has been automated
in commercially available equipment.
Smaller peptides may be joined to form longer
ones.
298
Also useful is enzymatic synthesis. Protein-
hydrolyzing enzymes under appropriate conditions
will form peptide linkages, for example, joining to-
gether oligopeptides.
302,303
Other new methods have
been devised to join unprotected peptides.
304,305
Semisynthetic approaches can also be used to place
unnatural amino acids into biologically synthesized
proteins through the use of suppressor transfer RNAs
(Chapter 29).
306
2. Combinatorial Libraries
Many chemists devote all of their efforts to the
synthesis of new compounds, including polypeptides,
that might be useful as drugs. Traditionally, this has
involved the tedious preparation of a large number of
compounds of related structure which can be checked
individually using various biochemical or biological
tests. In recent years a new approach using combina-
torial chemistry has become very popular and is
continually being adapted for new purposes.
307310
There are several approaches to creating a combinato-
rial library.
In split synthesis procedures a solid-phase
Figure 3-15 Procedure for solid-phase peptide synthesis devised by Merrifield.
299
Cl CH
2
polymer
H
3
C C CH
2
CH
3
+ CO
2
Isobutene
Ethanol
80C
Cl
HCl/acetic acid or
trifluoroacetic acid,
then trimethylamine
Dicyclohexylcarbodimide
(Eq. 3-10)
Repeat as needed
to add additional
residues Polypeptide
HF or HBr/trifluoroacetic acid

t-Butoxycarbonyl (Boc)
amino acid
Polystyrene
H
3
C C O C N CH COO
O CH
3
CH
3

R
1
H
Boc NH CHR
1
C O CH
2
polystyrene
O
H
2
N CHR
1
C O CH
2
polystyrene
O
COO

CH NH Boc
R
2
NH CHR
1
C O CH
2
polystyrene
O
C
O
CHR
2
NH Boc
129
synthesis is conducted on beads. For example, a family
of peptides, each with the same C terminus, can be
started on a large number of beads. After the first
amino acid residue is attached the beads are divided
into up to 20 equal portions and different amino acids
are added to each portion. The beads can then be
mixed and again subdivided. The third residue will
again contain many different amino acids attached to
each of the different amino acids in the second posi-
tion. By repeating the procedure again, perhaps for
many steps, a library of random peptides with each
bead carrying a single compound will be formed.
To test whether a polypeptide or other compound
carried on a given bead has a derived biological activity,
such as the ability to inhibit a certain enzyme, various
assays that require only one bead can be devised. How-
ever, if a particular bead carries a compound of inter-
est, how can it be identified? The bead carries only a
small amount of compound but it may be possible
using microsequencing procedures to identify it. An
alternative procedure is to use an encoding method to
identify the beads.
An alternative to the one beadone peptide
approach is to incorporate random sequences of a
DNA segment into a gene that can be used to display
the corresponding peptide sequence. This is illustrated
in Fig. 3-16. A protein segment (which may be a ran-
dom sequence) can be displayed either on the major
coat proteins along the shaft or on the minor coat
proteins at the end of the bacterial virus fd (see Fig. 7-7).
In the case of random insertions, each virus particle
may display a different sequence (as many as 10
8
).
Peptides may be selected by binding to a desired
receptor or monoclonal antibody and the DNA encap-
sulated in the virus particle can be used to produce
more peptides for identification purposes.
311 316
Many
other ingenious systems for constructing and testing
libraries of peptides
317 320
and other molecules
319,321,322
are being devised. One of these involves a photoli-
thography procedure for immobilizing macromolecules
in a regular addressable array, e.g., in a 0.5-mm check-
erboard pattern, on a flat surface.
323,324
G. Microscopy
The light microscope
325
was developed around
1600 but serious studies of cell structure (histology)
did not begin until the 1820s. By 1890, microscope
lenses had reached a high state of perfection
326
but the
attainable resolution was limited by the wavelength of
light. For 450 nm blue light the limit is about 300 nm
and for ultraviolet light, viewed indirectly, about 200
nm.
325,327
By the 1940s the electron microscope with its
far superior resolving power had overshadowed the
light microscope.
For both light and electron microscopy, the prepa-
ration of thin sections of cells is a very important
technique. Only with very thin sections is the image
sufficiently focused. However, confocal scanning
optical microscopy, invented in the 1950s but not
used commercially until much later,
328,329
provided an
alternative solution to the focusing problem. A conical
beam of light focused to a point is scanned across the
sample and the transmitted light (or light emitted by
fluorescence) passes through a small pinhole aper-
ture located in the primary image plane to a photo-
multiplier tube where its intensity is recorded. The
illuminating beam is moved to scan the entire field
sequentially. A series of pinholes in a spinning disk
may accomplish the same result. The focal plane can
G. Microscopy
Figure 3-16 Model of bacteriophage fd engineered to
display peptides as inserts in the coat proteins of the virus.
The native virus structure is shown in gray; proteins not
present in the native virus are shown black or green. Inserted
near the N-termini of some major coat proteins is a 6-residue
peptide. To one of these peptides a specific Fab antibody
fragment (green) has bound from solution, and a second
Fab is shown nearby. The N-terminal region of a minor
coat protein at the end of the virion has been engineered to
display a (different) Fab fragment. Steric constraints are less
stringent for inserts in the minor proteins, but fewer copies
per virion are possible. Reprinted with permission from
Barbas, et al.
313a
130 Chapter 3. Determining Structures and Analyzing Cells
be varied so that an image of a thick object such as a
cell can be optically sectioned into layers of less than 1
m thickness. Stereoscopic pairs can also be generated
(Fig. 3-17).
330
A newer development in confocal microscopy is
the use of two-photon and three-photon excitation of
the fluorescent molecules that occur naturally within
cells using short pulses of short-wavelength high-
energy laser light.
331
Distribution of such compounds
as NADH,
332
DNA, and the neurotransmitter seroto-
nin
333
can be observed without damaging cells. Indi-
vidual storage granules, each containing ~ 5 x 10
8
molecules of serotonin in a concentration of ~ 50 mM,
can be seen.
334
Another new instrument, the near-
field scanning optical microscope (NSOM),
335,336
is
a lensless instrument in which the illuminating beam
passes through a very small (e.g., 100 nm diameter)
hole in a probe that is scanned in front of the sample.
It may extend the limit of optical microscopy to ~ 1/50
the wavelength of the light.
Since it first became commercially available in
1939, the electron microscope has become one of the
most important tools of cell biology.
337,338
The practical
resolution is about 0.4 nm, but recent developments in
scanning electron microscopy have resulted in resolu-
tion of 0.14 nm.
339
Of major importance was the devel-
opment around 1950 of microtomes and knives capable
of cutting thin (20 200 nm) sections of tissues embed-
ded in plastic.
340
A bacterium such as E. coli can be
sliced into as many as 10 thin longitudinal slices (see
Fig. 1-4) and a eukaryotic cell of 10 m diameter into
100 slices. Serial sections can be examined to deter-
mine three-dimensional structures. For some results
see Bubel.
341
If a slice of fresh (frozen) tissue is examined directly,
little is seen because most of the atoms found in cells
are of low atomic mass and scatter electrons weakly
and uniformly. Therefore, thin sections must be
stained with atoms of high atomic mass, e.g., by
treatment with potassium permanganate or osmium
tetroxide. Tissues must also be fixed to prevent
disruption of cell structures during the process of
removal of water and embedding in plastic. Fixatives
such as formaldehyde react with amino groups and
other groups of proteins and nucleic acids. Some
proteins are precipitated in place and digestive en-
zymes that otherwise would destroy much of the fine
structure of the cell are inactivated. Glutaraldehyde
(a five-carbon dialdehyde) is widely used to fix and
crosslink protein molecules in the tissue. The methods
continue to be improved.
342
Small particles, including macromolecules, may
be shadowed. Chromium or platinum can be evap-
orated in a vacuum from an angle onto the surface of
the specimen, Individual DNA molecules can be
seen in this way.
343
In fact, only the shadows
are seen and they are 2 3 times wider than the DNA
molecules. In the negative contrast method a thin
layer of a solution containing the molecules to be
examined, together with an electron-dense material
such as 1% sodium phosphotungstate, is spread on a
thin carbon support film. Upon drying, a uniform
electron-dense layer is formed. Where the protein
molecules lie, the phosphotungstate is excluded, giving
an image of the protein molecule.
Surfaces of cells, slices, or intact bacteria can be
coated with a deposit of platinum or carbon. The
coating, when removed, provides a negative replica
which can be examined in the microscope. Alterna-
tively, a thin plastic replica can be made and can be
shadowed to reveal topography. In freeze fracturing
and freeze etching, fresh tissue, which may contain
glycerol to prevent formation of large ice crystals, is
frozen rapidly. Such frozen cells can often be revived;
hence, they may be regarded as still alive until the
moment that they are sliced! The frozen tissue is
placed in a vacuum chamber within which it is sliced
or fractured with a cold knife. If desired, the sample
can be kept in the vacuum chamber at about 100C
for a short time, during which some water molecules
evaporate from the surface. The resultant etching
reveals a fine structure of cell organelles and mem-
branes in sharp relief. After etching, a suitable replica
is made and examined (Fig. 1-15A and E). Fracturing
tends to take place through lipid
portions of cell membranes.
Small viruses, bacterial flagella,
ribosomes, and even molecules can
be seen by electron microscopy.
However, to obtain a clear image
in three dimensions requires a
computer-based technique of
image reconstruction or electron
microscope tomography, which
was developed initially by Aaron
Klug and associates.
344 349
A sam-
ple is mounted on a goniometer, a
device that allows an object to be
tilted at exact angles. Electron
Figure 3-17 Confocal micrograph showing a forty-micrometer stereo slice in
a 90-m thick section of mouse cerebellum.
330
Courtesy of A. Boyde.
131
micrographs are prepared with the sample untilted
and tilted in several directions at various angles, e.g.,
up to 90 in 10 increments. The micrographs are
digitized and a computer is used to reconstruct a
three-dimensional image.
In electron crystallography
350
micrographs of
two-dimensional crystalline arrays of molecules or
larger particles are prepared. A Fourier transform of
the micrograph gives a diffraction pattern which can
be treated in a manner similar to that usual for X-ray
diffraction to give a three-dimensional image. An
important milestone in use of this technique was the
determination of the structure of bacteriorhodopsin
(Fig. 23-45) at 0.3-nm resolution. Bear in mind that
X-ray crystallography can also be viewed as a form of
microscopy.
Invention of the scanning tunneling microscope
(STM) by Binnig and Roher
351 353
initiated a new revo-
lution in microscopy. The STM and similar scanned
probe microscopes examine surfaces by moving a fine
probe mounted in a piezoelectric x,y,z-scanner
354
across
the surface to be examined. The tiny tungsten probe
of the STM is so fine that its tip may consist of a single
atom. When a small voltage is applied a minuscule
quantum mechanical tunnelling current flows across
the small gap between the probe and the surface and a
high-resolution image, sometimes at atomic resolution,
is created from the recorded variation in current.
354-359
The STM theoretically responds only to surfaces that
conduct electrons, but nonconducting samples have
been imaged at high humidity; presumably by con-
ductance of electrons or ions through the surface water
layer. The success of the STM spurred the development
of many other types of scanned probe microscopes.
Among these, the atomic force microscope (AFM;
Fig. 3-18) has been especially useful for biological
materials, including proteins and nucleic acids. The
AFM moves a fine-tipped stylus directly across the
sample surface or, alternatively, vibrates the probe
above the surface. The small up-and-down move-
ments of the stylus are recorded
359 366
and thereby
create a topographic or force-field map of the sample.
AFM images contain three-dimensional information
and can be used to view individual molecules (Fig.
3-19).
367,367a
Chemical force microscopy is sensitive
to adhesion and friction as a function of the interaction
between defined chemical groups on the tip and sam-
ple.
368
An emerging field is force spectroscopy, in which
the AFM measures interaction forces between and
within individual molecules.
369 371a
Under development
are NMR microscopes (Section I). There is continual
effort to see small objects more directly and more
clearly!
G. Microscopy
Figure 3-18 Schematic diagram of the atomic force micro-
scope.
360
Courtesy of Paul Hansma.
Figure 3-19 AMF images of cholera toxin
364
(see also Box
11-A). Courtesy of Z. Shao.
Lens
Hotodiode
Cantilever
Light
XYZ translator
Plexiglas
Sample
O ring
Liquid
132 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-20 An X-ray diffraction photograph such as was used to determine
the structure of hemoglobin. This precession photograph was obtained from a
crystal of human deoxyhemoglobin by rotating the crystal along two different
axes in a defined manner before a narrow X-ray beam. The film also was
moved synchronously. The periodicity observed in the photograph is a result
of a diffraction phenomenon arising from the periodic arrangement of atoms
in the crystal. The distances of the spots from the origin (center) are inversely
related to the distances between planes of atoms in the crystal. In this photo-
graph (which shows only two dimensions of the three-dimensional diffraction
pattern) the spots at the periphery represent a spacing of 0.28 nm. If the
intensities of the spots are measured, and if the phases of the harmonic func-
tions required for the Fourier synthesis can be assigned correctly, the structure
can be deduced to a resolution of 0.28 nm from a set of patterns of this type.
In the case of human deoxyhemoglobin, a complete set of data would consist
of about 16,000 spots. With modern equipment the resolution limit can be
extended to better than 0.15 nm (or more than 100,000 unique spots).
Photograph courtesy of Arthur Arnone.
tallography of hemoglobin. The project seemed hope-
less at times, but in 1968, 31 years later, Perutz had
determined the structure of hemoglobin.
383 386
If X-rays could be focused easily, could one build
an X-ray microscope that would permit the immediate
viewing of molecular structures? X-ray holograms at
the molecular level have been obtained.
387,388
However,
currently the only practical X-ray microscope for protein
structures involves the measurement of diffraction
patterns created by the scattering of X-rays (or of neu-
trons) from the crystalline lattice. The details of this
procedure can be found in other sources.
372375,378,389390
It is sufficient to point out here that a pattern of many
H. X-ray and Neutron Diffraction
One of the most important techniques by which
we have learned bond lengths and angles and precise
structures of small molecules is X-ray diffraction.
Today, this technique, which involves measurements
of the scattering of X-rays by crystalline arrays of
molecules, is being used with spectacular success to
study macromolecules of biochemical and medical
importance.
372 378
X-rays were described by Rntgen in 1896 but
there was uncertainty as to their wave nature.
274
It
was not until 1912 that the wavelengths of X-rays had
been measured and it was recog-
nized that they were appropriate for
the use of X-rays in structure deter-
mination. Consider the fact that
with a conventional light micro-
scope we cannot distinguish two
small objects that are much closer
together than the distance represent-
ed by the wavelength of the light
with which we observe them. This
is about 460 nm for blue light. By
comparison the wavelength of the
copper K

radiation, which is used


in protein crystallography, is 0.1542
nm entirely appropriate for seeing
the individual atoms of which mat-
ter is composed. Recognizing this,
W. L. Bragg in Cambridge, England,
in 1913 used X-ray diffraction to
establish the structures of NaCl,
KCl, and KBr in the crystalline state.
The science of X-ray crystallography
had been founded.
379
In 1926, James Summer crystal-
lized the enzyme urease (Chapter 16)
and crystallization of other enzymes
soon followed.
380
In 1934, J. B.
Bernal brought back to Cambridge
from Uppsala, Sweden, some crys-
tals of pepsin almost 2 mm long that
had been grown in T. Svedbergs
laboratory. Bernal and Crowfoot
showed that these delicate crystals,
which contained almost 50% water,
gave a sharp diffraction pattern
when they were protected by enclo-
sure in a narrow capillary tube
containing some of the mother
liquor from which the crystals had
been grown.
381,382
After this, diffrac-
tion patterns were obtained for
many protein crystals, and in 1937
Max Perutz chose for his thesis
work at Cambridge the X-ray crys-
133
spots, such as that in Fig. 3-20, is obtained. In a typical
determination of a protein structure from 10,000 to
several times that number of spots must be measured.
The needed information is contained in the coordinates
of the spots (i.e., in the angles through which the
scattering occurs) and in the intensities of the spots.
The structure is obtained by a Fourier synthesis in
which a large number of mathematical functions in
the form of three-dimensional sinusoidal waves are
summed. The wavelengths of these functions are
determined by the positions of the spots in the diffrac-
tion pattern. The amplitudes of the waves are related
to the intensities of the spots.
374
However, the waves
are not all in phase and the phases must be learned in
some other way. This presents a difficult problem.
Mathematical methods have been devised that auto-
matically determine the phases for small molecules
and usually allow the structures to be established
quickly. For proteins it is more difficult.
In 1954, Perutz introduced the isomorphous
replacement method for determining phases. In this
procedure a heavy metal, such as mercury or platinum,
is introduced at one or more locations in the protein
molecule. A favorite procedure is to use mercury
derivatives that combine with SH groups. The resulting
heavy metal-containing crystals must be isomorphous
with the native, i.e., the molecules must be packed the
same and the dimensions of the crystal lattice must be
the same. However, the presence of the heavy metal
alters the intensities of the spots in the diffraction
pattern and from these changes in intensity the phases
can be determined. Besides the solution to the phase
problem, another development that was absolutely
essential was the construction of large and fast com-
puters. It would have been impossible for Perutz to
determine the structure of hemoglobin in 1937, even
if he had already known how to use heavy metals to
determine phases.
The first protein structure to be learned was that of
myoglobin, which was established by Kendrew et al. in
1960.
391 393
That of the enzyme lysozyme was deduced
by Blake et al. in 1965.
394
Since then, new structures
have appeared at an accelerating rate so that today we
know the detailed architecture of over 6000 different
proteins
395
with about 300 distinctly different folding
patterns.
396
New structures are being determined at
the rate of about one per day. X-ray diffraction has
also been very important to the study of naturally or
artifically oriented fibrous proteins
397
and provided
the first experimental indications of the structure of
proteins.
Suppose that you have isolated a new protein.
How can you learn its three-dimensional structure?
The first step is crystallization of the protein, some-
thing that a biochemist may be able to do. Crystalliza-
tion is done in many ways, often by the slow diffusion
of one solution into another or by the slow removal
of solvent through controlled evaporation.
374,398
Am-
monium sulfate and polyethylene glycol are two
commonly used precipitants. The presence of the
neutral detergent -octyl glucoside improves some
crystals.
399
Droplets of protein solution mixed with
the precipitant are often suspended on microscope
cover glasses in small transparent wells or are placed in
depression plates within closed plastic boxes.
374,400,401
In either case, a reservoir of a solution with a higher
concentration of precipitant is present in the same
compartment. Water evaporates from the samples
into the larger reservoir, concentrating the protein
and causing its crystallization. Crystals grow slightly
better in a spacecraft than on Earth.
402,403
Some pro-
teins, notably myosin from muscle, crystallize well
only after reductive methylation of all lysine side
chains to dimethyllysine with formaldehyde and
sodium borohydride (Eq. 3-34).
278
The next step is for a protein crystallographer to
mount a small perfect crystal in a closed silica capil-
lary tube and to use an X-ray camera to record diffrac-
tion patterns such as that in Fig. 3-20. These patterns
indicate how perfectly the crystal is formed and how
well it diffracts X-rays. The patterns are also used to
calculate the dimensions of the unit cell and to assign
the crystal to one of the seven crystal systems and
one of the 65 enantiomorphic space groups. This
provides important information about the relationship
of one molecule to another within the unit cell of the
crystal. The unit cell (Fig. 3-21) is a parallelopiped
H. X-ray and Neutron Diffraction
Figure 3-21 Diagram showing an asymmetric unit as it
might appear in a unit cell of space group P2
1
2
1
2
1
. This unit
cell has three pairs of nonintersecting twofold screw axes
which are marked by the shaded rods. These are designated
by arrows and the symbol. Two asymmetric units are related
one to another by rotation around a twofold axis together
with translation by one-half the dimension of the unit cell.
134 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-22 Stereoscopic view of a section of the structure of cutinase from
the fungus Fusarium solani determined to a resolution of 0.10 nm. The three
amino acid residues shown are serine 120 (top), histidine 188, and aspartate
175 (lower left). The structure is presented as a contour map with a wire
mesh drawn at a cutoff level of density equal to 1 above the average,
where is the root mean square density of the entire map. The side chains
of these three residues constitute the catalytic triad in the active site of this
enzyme (see Chapter 12). At this resolution more than one conformation of a
group may often be seen. For example, the gamma oxygen (OG) of S120 is
seen in two positions, the major one being toward His 188. When the map is
drawn with a lower contour level the N-H proton on His 188 that is hydrogen
bonded to Asp 175 can also be seen.
410
Courtesy of Christian Cambillau.
whose sides are parallel to crystal-
lographic axes and which, by
translation in three dimensions,
gives rise to the entire crystal. The
unit cell must contain at least one
asymmetric unit, the smallest
unit of structure that lacks any
element of crystal symmetry. The
asymmetric unit may consist
of one or a small number of pro-
tein subunits.
In crystals of the triclinic
system all of the asymmetric units
are aligned in the same manner
and there are no axes of symmetry.
Monoclinic crystals have a single
twofold or dyad axis (see Fig. 7-
11). The unit cell might contain
two molecules related one to
another by the dyad or one dimer-
ic molecule with the dyad located
as in Fig. 7-11C. Orthorhombic
crystals have three mutually per-
pendicular twofold axes and the
unit cell is a rectangular solid.
Trigonal, tetragonal, and hexago-
nal crystal systems have three,
four and sixfold axes of symmetry,
respectively, while the cubic crys-
tal contains four threefold axes
along with diagonals of the cube
as well as two-fold axes passing
through the faces (see Fig. 15-14).
Within each crystal system there
are several space groups. An exam-
ple is the orthorhombic space
group P2
1
2
1
2
1
, which is often met with small organic
molecules and proteins. In this space group the unit
cell contains three mutually perpendicular but nonin-
tersecting twofold screw axes (Fig. 3-21). The position
of one molecule in the unit cell is related to the next by
both a 180 degree rotation about the twofold axes and
a translation of one-half the length of the unit cell.
There is such a screw axis for each of the three direc-
tions.
372
The third step in the structure determination is
collection of the X-ray diffraction data. This may be
done with a diffractometer in which a narrow colli-
mated pencil source of X-rays is aimed at the crystal
and the intensities and positions of the diffracted
beams are measured automatically. The computer-
controlled diffractometer is able to measure the angles
to within less than one-hundredth of a degree. If
sufficient time is allowed, very weak spots can be
counted. Today, diffractometers are more likely to be
used for preliminary measurements, while the major
data collection is done with an area detector, an
instrument that collects many reflections simulta-
neously. There are some difficulties. For example,
during the long periods of irradiation needed to mea-
sure the weak spots with a diffractometer the protein
crystals decompose and must be replaced frequently.
Data collection may last for months. The newest
methods utilize more powerful X-ray sources and
often synchrotron radiation, which delivers very
short and extremely intense pulses of X-rays and
allows data to be collected on very small well-formed
crystals.
404
The fourth step is the preparation of isomorphous
crystals of heavy metal-containing derivatives. The
heavy metal may be allowed to react with the protein
before crystallization or may be diffused into preformed
crystals. A variety of both cationic and anionic metal
complexes, even large Ta
6
Br
12
2+
tantalum clusters,
have been used.
405
Two or more different heavy metal
derivatives are often required for calculation of the
phases. The heavy metal atoms must be present at
only a very small number of locations in the unit cell.
135
An entire data set must be collected for each of these
derivatives. The evaluation of the phases from these
data is a complex mathematical process which usually
involves the calculation first of a difference Patterson
projection.
406
This is derived by Fourier transforma-
tion of the differences between the scattering intensities
from the native and heavy atom-containing crystals.
The Patterson map is used to locate the coordinates
of the heavy metal atoms which are then refined and
used to compute the phases for the native protein.
Alternative methods of solving the phase problem
are also used now. When a transition metal such as Fe,
Co, or Ni is present in the protein, anomolous scatter-
ing of X-rays at several wavelengths (from synchro-
tron radiation) can be used to obtain phases. Many
protein structures have been obtained using this
multiple wavelength anomalous diffraction (MAD
phasing) method.
404,407,408
Selenocysteine is often
incorporated into a protein that may be produced in
bacteria using recombinant DNA procedures. Crystals
are prepared both with protein enriched in Se and with-
out enrichment. In some ways it is better to incorporate
tellurium (
127
Te) in telluromethionine.
409
In the fifth step of an X-ray structure determination
the electron density map is calculated using the inten-
sities and phase information. This map can be thought
of as a true three-dimensional image of the molecule
revealed by the X-ray microscope. It is usually dis-
played as a stereoscopic view on a computer graphics
system (Fig. 3-22). It is also often prepared in the form
of a series of transparencies mounted on plastic sheets.
Each sheet represents a layer, perhaps 0.1 nm thick,
with contour lines representing different levels of
electron density.
Using the electron density map a three-dimensional
model of the protein can be built. For years, the cus-
tomary procedure was to construct a model at a scale
of 2 cm = 0.1 nm using an optical comparator, but
H. X-ray and Neutron Diffraction
Figure 3-23 (A) Stereoscopic
-carbon plot of the cystolic
aspartate aminotransferase
dimer viewed down its dyad
symmetry axis. Bold lines are
used for one subunit (subunit 1)
and dashed lines for subunit 2.
The coenzyme pyridoxal 5'-
phosphate (Fig. 3-24) is seen
most clearly in subunit 2 (center
left). (B) Thirteen sections,
spaced 0.1 nm apart, of the
2-methylaspartate difference
electron density map superim-
posed on the -carbon plot
shown in (A). The map is con-
toured in increments of 2
(the zero level omitted), where
= root mean square density of
the entire difference map. Posi-
tive difference density is shown
as solid contours and negative
difference density as dashed
contours. The alternating series
of negative and positive differ-
ence density features in the
small domain of subunit 1
(lower right) show that the
binding of L-2-methylaspartate
between the two domains of this
subunit induces a right-to-left
movement of the small domain.
(Continues)
A
B
136 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-23 (Cont.)
(C) Nine sections, spaced
0.1 nm apart, of a part of
the 2-methylaspartate
difference map superim-
posed on the atomic
model shown in (A) and
(D). The coenzyme is
shown as the internal
aldimine with Lys 258
(see Fig. 14-6, 14-10). The
positive and negative
contours on the two sides
of the coenzyme ring
indicate that the coen-
zyme tilts over to form
the external aldimine
when substrates react.
413
(D) Superimposed
structure of the active site
of the enzyme in its free
form as in (A) (bold lines)
and the refined structure
of the -methylaspartate
complex, (dashed lines).
411
This illustrates the tilting
of the coenzyme ring,
which is also shown in
Eq. 14-39 and Fig. 14-10.
Courtesy of Arthur Arnone
and Sangkee Rhee.
C
D
crystallographers now use computer graphics systems.
The three-dimensional image of the electron density
map and a computer-generated atomic model are super-
imposed on the computer screen. An example is shown
in Fig. 3-22. When the superposition has been com-
pleted, the coordinates of all atoms are present in the
memory of the computer.
The final step in the structure determination is
refinement using various mathematical methods.
From the coordinates of the atoms in the model the
expected diffraction pattern is computed and is com-
pared with that actually observed. The differences
between predicted and observed density are squared
and summed. The sum of the squares constitutes an
error function which is then minimized by moving
the various atoms in the model short distances while
keeping bond lengths, angles, and van der Waals
distances within acceptable limits and recalculating
the error function. This complex refinement pro-
cedure must be repeated literally hundreds of thou-
sands of times with every part of the structure being
varied. Structures at very high resolution (0.07 0.1
nm) may reveal multiple positions for hydrogen-
bonded side chains
410a,b
as well as hydrogen atoms
(Fig. 3-22) and even bonding electrons.
410b
Once the three-dimensional structure of the pro-
tein is known, further experiments are usually done
using the X-ray diffraction technique. Since protein
crystals contain channels of solvent between the
packed molecules, it is usually possible to diffuse
small molecules into the crystal. These may be sub-
strates, inhibitors, or allosteric effectors. Diffraction
data are collected after diffusion of the small molecules
into the crystal and a difference electron density
map may be calculated. This may show exactly where
those molecules were bound. An example is shown in
Fig. 3-23. Sometimes difference maps not only show
the binding to an enzyme of a substrate or other small
molecule but also reveal conformational changes in
proteins. Such is the case for Fig. 3-23, which shows
the binding of -methylaspartate, an inhibitor that
behaves initially like a substrate and goes part way
through the reaction sequence for aspartate amino-
transferase until further reaction is blocked by the
methyl group. This difference map also shows that
the part of the protein to the left of the binding site in
the figure has moved.
411
Using the X-ray data it was
possible subsequently to deduce the nature of the
conformational change.
Examination of the effect of temperature on the
diffraction pattern of a protein can give direct informa-
tion about the mobility of different parts of the mole-
137
cules.
412
This complements information obtainable in
solution from NMR spectra.
Because hydrogen atoms contain only one electron,
and therefore scatter X-rays very weakly, they are
usually not seen at all in X-ray structures of proteins.
However, neutrons are scattered strongly by hydrogen
atoms and neutron diffraction is a useful tool in
protein structure determination.
414,415
It has been used
to locate tightly bonded protons that do not exchange
with
2
H
2
O as well as bound water (
2
H
2
O).
The development of synchrotron radiation as an
X-ray source
404,416 418
has permitted accumulation of
data for electron density difference maps in less than
1 s and it is expected that such data can eventually be
acquired in ~ 1 ps.
419 421
If a suitable photochemical
reaction can be initiated by a picosecond laser flash, a
substrate within a crystalline enzyme can be watched
as it goes through its catalytic cycle. An example is
the release of inorganic phosphate ions from a caged
phosphate (Eq. 3-49) and study of the reaction of the
released phosphate with glycogen phosphorylase
(Chapter 12).
422,423
However, caged substrates usually must diffuse
some distance before reacting, so very rapid events
cannot be studied. An alternative approach is to diffuse
substrates into crystals at a low temperature at which
reaction is extremely slow but a substrate may become
seated in an active site ready to react. In favorable
cases such frozen Michaelis complexes may be heated
by a short laser pulse to a temperature at which the
reaction is faster and the steps in the reaction may be
observed by X-ray diffraction.
424,425
I. Nuclear Magnetic Resonance (NMR)
Organic chemists and biochemists alike have long
relied on NMR spectroscopy to assist in identification
and determination of structures of small compounds.
Most students have some familiarity with this tech-
nique and practical information is available in many
places.
426 428
Measurements can be done on solids,
liquids, or gases but are most often done on solutions
held in special narrow NMR tubes. Volumes of samples
are typically 0.5 ml (for a 15-mM solution containing
15 mol of protein) but less for small molecules and
I. Nuclear Magnetic Resonance (NMR)
higher concentrations. Newer techniques allow use of
a volume as small as 5 nl and containing < 0.1 nmol of
sample.
429
For many years progress in biochemical
application of NMR was slow, but a dramatic increase
in the power of the spectrometers, driven in great
measure by the revolution in computer technology,
has made NMR spectroscopy a major force in the
determination of structures and functions of proteins
and nucleic acids.
430,431
1. Basic Principles of NMR Spectroscopy
The basis of NMR spectroscopy lies in the absorp-
tion of electromagnetic radiation at radiofrequencies
by atomic nuclei.
426,427,432 437
All nuclei with odd mass
numbers (e.g.,
1
H,
13
C,
15
N,
17
O,
19
F, and
31
P), as well as
those with an even mass number but an odd atomic
number, have magnetic properties. Absorption of a
quantum of energy E = h occurs only when the nuclei
are in the strong magnetic field of the NMR spectrom-
eter and when the frequency of the applied electro-
magnetic radiation is
appropriate for resonance
with the nucleus being ob-
served. In the widely used
500-megahertz NMR spec-
trometers the liquid helium-
cooled superconducting
electromagnet has a field
strength of 11.75 tesla (T). In
this field a proton resonates at
~ 500 megahertz (MHz) and
nuclei of
31
P,
13
C, and
15
N at ~ 202, 125, and 50 MHz,
respectively. At 500 MHz the energy of a quantum is
only E = 3.3 x 10
33
x 10
8
J = 0.2 J mol
1
, more than four
orders of magnitude less than the average energy of
thermal motion of molecules (3.7 kJ mol
1
). Thus, the
spin transitions induced in the NMR spectrometer
have no significant effect on the chemical properties of
molecules.
The resonance frequency at which absorption
occurs in the spectrometer is given by Eq. 3-50, where
H
o
is the strength of the external magnetic field, is
the magnetic moment of the nucleus being investigated,
and h is Plancks constant. The basis for NMR spectros-
copy lies in the fact that nuclei in different positions in
a molecule resonates at slightly different frequencies.
In a protein each one of the hundreds or thousands
of protons resonate at its own frequency. With older
NMR instruments a spectrum at the constant magnetic
field H
o
can be obtained by varying the frequency and
observing the values at which absorption occurs, much
as is done for ultraviolet, visible, and infrared spectra
(Chapter 23). With newer pulsed NMR spectrometers
the measurement is done differently but the spectra
(3-50) = H
o
/h
N
O
H
C
CH
3
O
O P O
O
OH
N
O
C
CH
3
O
O P O
O
OH

+
Caged phosphate
NO
C
CH
3
O
+
h
~315 nm

HPO
4
2
H

k = 10
5
s
-1
20C, pH 7
(3-49)
138 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-24 The 60-MHz
proton magnetic resonance
spectrum of pyridoxal 5'-
phosphate at neutral pH
(apparent pH = 6.65). The
internal standard is DSS.
Chemical shifts in parts per
million are indicated beside
the peaks. Spectrum courtesy
of John Likos.
10.0 8.0 6.0 4.0 2.0 0.0
10.42
CHO
7.71
6-H
SSB
CH
2
J =6.4
SSB
4.75
1
H
2
HO
2.45
CH
3
0
DSS
(ppm)
N
O
CH
3
C
O
C
O
P
O
O
O
H
H

4'
6
5
H
H
H
2'
2
1

5.11
look the same. The higher the magnetic field, the
greater the variation in resonance frequency and the
higher the sensitivity. The most powerful commercial
NMR spectrometers currently available operate at
about 750 MHz for
1
H and a few higher frequency
instruments have been built.
The proton NMR spectrum of the coenzyme
pyridoxal phosphate in
2
H
2
O is shown in Fig. 3-24 as
obtained with a 60-MHz spectrometer. Four things can
be measured from such a spectrum: (1) the intensity
(area under the band). In a proton NMR spectrum,
areas are usually proportional to the numbers of
equivalent protons giving rise to absorption bands;
(2) the chemical shift, the difference in frequency
between the peak observed for a given proton and a
peak of some standard reference compound. In Fig.
3-24 the reference peak is at the right edge; (3) the
width at half-height (in hertz), a quantity that can
provide information about molecular motion and
about chemical exchange; and (4) coupling constants
which measure interactions between nearby magnetic
nuclei. These are extremely important to the determi-
nation of structures of both small and large molecules.
With a magnetic field of H
o
= 11.75 T and a 500-
MHz oscillator, the positions of proton resonances in
organic compounds are spread over a range of ~10,000
Hz. This is 20 parts per million (ppm) relative to 500
MHz. Positions of individual resonances are usually
given in ppm and are always measured in terms of a
shift from the resonance position of some standard
substance. For protons this is most often tetramethyl-
silane (TMS), an inert substance that can be added
directly to the sample in its glass tube. Biochemists
often use
2
H
2
O as solvent and the water-soluble sodium
3-trimethylsilyl 1-propane sulfonate (DSS or Tiers salt)
as a standard. Its position is insignificantly different
from that of TMS. For NMR spectra measured in
2
H
2
O,
the pD of the medium is sometimes indicated. It
has often been taken as the pH meter reading plus 0.4.
However, because of uncertainty about the meaning of
pD, most workers cite the apparent pH measured with
a glass electrode and standardized against aqueous
buffers.
438
It is important to describe how the mea-
surement was made when publishing results.
Band widths. The narrowness of a band in an
NMR spectrum is limited by the Heisenberg uncer-
tainty principle, which states that E x t = h/2,
where h is Plancks constant, E is the uncertainty in
the energy, and t is the lifetime of the magnetically
excited state. Since E = h for electromagnetic radia-
tion, E is directly proportional to the width of the
absorption band (customarily measured at one-half
its full height). The magnetic nucleus is well shielded
from external influences and the lifetime of its excited
state tends to be long. Hence, is small, often
amounting to less than 0.2 Hz. This fact is very favor-
able for the success of high-resolution proton magnetic
resonance. However, bands are often much broader
for large macromolecules.
The chemical shift. In a molecule such as TMS,
the electrons surrounding the nuclei shield the
nucleus so that it does not experience the full external
magnetic field. For this reason, absorption occurs at a
high frequency (high energy). Protons that are bound
139 I. Nuclear Magnetic Resonance (NMR)
to an atom deficient in electrons (because of attach-
ment to electron withdrawing atoms or groups) are
deshielded. The greater the deshielding, the further
downfield from the TMS position is the NMR peak.
The magnitude of this chemical shift may be stated
in hertz, but it is most often expressed in ppm as
(Eq. 3-51). The value of is the shift in frequency
relative to frequency of the oscillator in parts per million
and is independent of the field strength. It still depends
upon use of a particular reference standard which
must be stated when a value is given. In the spec-
trum shown in Fig. 3-24, the methyl protons appear
2.45 ppm below the DSS peak but still at a relatively
high field. Characteristic chemical shift ranges for
other protons (Table 3-3) extend to ~20 ppm.
Aromatic rings lead to strong deshielding of at-
tached protons because of a ring current induced in
the circulating electrons. Thus, in Fig. 3-24 the peaks
of the methylene protons which are adjacent to the
aromatic ring occur at 5.10 and 5.12 ppm. The 6-H,
which is bound directly to the ring, is more strongly
deshielded and appears at 7.71 ppm. The hydrogen
of the aldehyde groups is deshielded as a result of a
similar diamagnetic electronic circulation in the
carbonyl group. Its peak is even further downfield at
10.4 ppm. Ring current and other effects on chemical
shifts are important in NMR spectroscopy of proteins.
Aromatic proton resonances sometimes stand out
because they have been shifted far downfield. Ring
current effects on chemical shifts can be predicted quite
accurately if three-dimensional structures are known.
439
Additionally, computer programs are available for
predicting them.
440 443
Hydrogen bonding has a very large effect on the
chemical shift of protons. The resonance of a strongly
hydrogen-bonded proton is usually shifted downfield
from its position in non-hydrogen-bonding media.
This is especially true for hydrogen bonds to charged
groups, e.g., the NH of a histidine or tryptophan side
chain hydrogen bonded to a carboxylate group, a
situation often met in the active sites of proteins.
The chemical shift of
13
C in the carboxyl group is also
affected.
444,445
While ring current shifts can be pre-
dicted quite well, it is much more difficult to predict
the total chemical shift.
446,447
Scalar coupling (J coupling). The energy of the
spin transition of a hydrogen nucleus is strongly influ-
enced by the local presence of other magnetic nuclei,
e.g., other protons that are covalently attached to the
same or an adjacent atom. These neighboring protons
can be in either of the two spin states, a fact that results

( )
( )
( )
ppm
Hz
Hz of oscillator
=
10
6
(3-51)
in easily measured differences in the energy of the
NMR transition under consideration. This spinspin
interaction (coupling) leads to a splitting of NMR bands
of protons into two or more closely spaced bands. The
ethyl group often appears in NMR spectra as a quartet
of four evenly spaced peaks that arise from the CH
2
group and a triplet of peaks arising from the CH
3
pro-
tons. Protons attached to the same carbon (geminal
protons), and in similar environments, do not ordinarily
split each others peaks, while the protons on the
neighboring carbon do.
The coupling constant J is the difference in hertz
between the successive peaks in a multiplet. It is a
field-independent quantity and the same no matter
what the frequency of the spectrometer. In Fig. 3-24
the peak of the methylene protons is split by
1
H
31
P
coupling, with a value of J ~ 6.4 Hz. While spinspin
coupling is most pronounced when magnetic nuclei
are close together in a structure, the effect can some-
times be transmitted through up to five covalent
bonds. The technique of double irradiation or spin
decoupling can be used to detect spin coupling. The
sample is irradiated at the resonance frequency of one
of the nuclei involved in the coupling, while the spec-
trum is observed in the frequency region of the other
nucleus of the coupled pair. Under these conditions
the multiplet collapses into a singlet and the mutual
coupling of the two nuclei is established. The cou-
pling can be seen directly in appropriate two-dimen-
sional NMR spectra.
The coupling constant between two vicinal protons
which are attached to adjacent carbon atoms (or other
atoms) depends upon the torsion angle.
The Karplus equation (Eq. 3-52) relates J to the
torsion angle (so labeled to distinguish it from pep-
tide torsion angle ; Fig. 2-8).
J
H,H
A cos
2
+ B cos + C
This equation was predicted on theoretical grounds,
but the constants, A, B, and C are empirical.
450
Other
forms of the equation, some of them simplified, have
also been proposed.
451
The Karplus relationship is
often used to estimate time-averaged torsion angles
in peptides. For a C

HNH torsion angle the para-


meters A, B and C of Eq. 3-52 are ~ 6.4, 1.4, and 1.9,
respectively. For a C

H C

H they are ~ 9.5, 1.6, and


C C
H H' '
(3-52)
140 Chapter 3. Determining Structures and Analyzing Cells
1.8, respectively.
452
Coupling constants between
1
H
and
13
C or
15
N are of importance in determination of
three-dimensional structure of proteins.
453
The nuclear Overhauser effect (NOE) is the
result of transfer of magnetization from one nucleus
to a nearby nucleus directly through space rather than
via J-coupling.
427,454
This was observed first as a result
of irradiation of a resonance in a one-dimensional
spectrum resulting in an increased intensity of the
resonance of the nearby nucleus. Magnetization trans-
fer can occur from a given nucleus to one or more
nearby nuclei. Each such transfer that is detected is
usually referred to simply as an NOE. For an NOE
to be observable the two nuclei must be very close
together, < 0.5 nm (5 ). The strength of the magneti-
zation transfer falls off approximately as the sixth
power of the interatomic distance. Consider two
hydrogen atoms, both tightly hydrogen-bonded to
an intervening oxygen atom, e.g., of a carboxylate or
phosphate group. The expected H H distance would
be ~ 0.3 nm and a strong NOE between them would be
anticipated. Two nonbonded hydrogen atoms (e.g., on
methyl groups of amino acid side chains) can be as
close together as 0.24 nm at van der Waals contact and
could show a very strong NOE. However, contact is
rarely this close in proteins unless in a hydrogen bond.
2. Nuclei Other than Hydrogen
Deuterium (
2
H). The natural abundance is very
low so that use of
2
H-labeled compounds is practical
for study of metabolism, e.g., for following an
2
H label
in glucose into products of fermentation
455
or in mam-
malian blood flow.
456
Deuterium NMR has been used
extensively to study lipid bilayers (Chapter 8).
Carbon - 13. Use of
13
C in NMR developed
slowly because of the low natural abundance of this
isotope. Another complication was the occurrence of
13
C
1
H coupling involving the many protons normally
present in organic compounds. The latter problem was
solved by the development of wide-band proton
decoupling (noise decoupling). With a natural abun-
dance of only 1.1%,
13
C is rarely present in a molecule
at adjacent positions. Thus,
13
C
13
C coupling does
not introduce complexities and in a noise-decoupled
natural abundance spectrum each carbon atom gives
rise to a single peak. Even so,
13
C NMR spectroscopy
was not practical until pulsed Fourier transform (FT)
spectrometers were developed (Section 2).
Chemical shifts in
13
C spectra are often 200 ppm
or more downfield relative to TMS. The effects of
substituents attached to a carbon atom are often addi-
tive when two or more substituents are attached to the
same atom.
457,458
It is often necessary (but costly) to
prepare compounds enriched in
13
C beyond the natural
abundance. For proteins this may be done by growing
an organism on a medium containing [
13
C]glucose or a
single amino acid enriched in
13
C.
459,460
Using metabo-
lites enriched in
13
C, it is also possible to observe
metabolism of living tissues directly. For example,
glycogen synthesis from
13
C-containing glucose has
been observed in a human leg muscle using a wide-
bore magnet and surface coils for transmitting and
receiving.
461
This topic is discussed further in Box 17-C.
Nitrogen - 15. Despite difficulties associated
with low natural abundance (0.37%) and low sensitivity,
15
N NMR is practical and with isotopically enriched
samples has become very important. Proteins with a
high content of
15
N can be produced easily and inex-
pensively from cloned genes in bacterial plasmids.
For example, cells of E. coli can be grown on a minimal
medium containing [
15
N] NH
4
Cl. Since
13
C can also be
introduced in a similar way it is possible to incorporate
both isomers simultaneously. Production of uniformly
labeled protein containing
15
N and / or
13
C provides
the basis for multidimensional isotope-edited spectra
necessary for protein structure determination (next
section) and for study of tautomerization of histidine
rings (Eq. 2-6).
460,462 464 15
N chemical shifts of groups
in proteins are spread over a broad range (Table 3-3).
465
Phosphorus - 31. NMR spectroscopy using
31
P,
the ordinary isotope of phosphorus, also has many
uses.
466
Application of
31
P NMR to living tissues has
been extraordinarily informative
467
and is dealt with in
Chapter 6. The many phosphorus nuclei in nucleotides,
coenzymes, and phosphorylated metabolites and pro-
teins are all suitable objects of investigation by NMR
techniques.
Fluorine - 19. Although not abundant in nature,
19
F gives an easily detected NMR signal and can be
incorporated in place of hydrogen atoms into many
biochemical compounds including proteins.
468,469
In
one study genetic methods were used to place 3-fluoro-
tyrosine separately into eight positions in the lac repres-
sor protein (Chapter 29).
470
Measurements of
19
F NMR
spectra were used to study domain movement. Active
site groups of enzymes can be modified to incorporate
19
F.
471
Binding of fluorinated substrates can be stud-
ied.
472
Nontoxic
19
F-containing compounds are useful
as intracellular pH indicators, the NMR spectrometer
O H N
0.28 nm
~0.3 nm
H N
0.28 nm

141
serving as the pH meter (Box 6-A).
473,474
An atom of
fluorine attached to an aromatic ring is highly and
predictibly sensitive to inductive effects of substituents
in the para position to the fluorine.
475
For example,
fluorine at the 6-position in pyridoxal phosphate (Fig.
14-4) can be observed in enzymes and reports changes
in coenzyme structure.
476,477
Some other nuclei. Here are a few reported uses
of NMR on other nuclei.
3
He, binding into little cavities
in fullerenes;
478 11
B, binding of boronic acids to active
sites;
479 23
Na, measurement of intracellular [Na
+
];
480 482
35
Cl and
37
Cl, binding to serum albumin;
483 113
Cd,
reporter that can replace Zn
2+
(no magnetic moment)
in active sites of many enzymes and in nonenzymatic
systems as well;
484
Tl, replacing K
+
in enzyme binding
sites;
485 17
O, study of dynamics of protein hydration;
486
and
77
Se, observation of acetylchymotrypsin interme-
diate.
487
3. Fourier Transform Spectrometers and
Two-Dimensional NMR
Although NMR spectroscopy was widely used by
the 1950s it was revolutionized by two developments,
pioneered by Richard Ernst in the mid 1960s and
1970s.
488
The first of these was pulsed Fourier trans-
form (FT) spectroscopy, which permits rapid accumu-
lation of high-resolution spectra. In an FT NMR
spectrometer a strong pulse of radiofrequency (RF)
radiation is delivered to the sample over a period of
a few microseconds and its effects are observed at all
frequencies simultaneously. Although 1 2 or more
seconds must be allowed before the next pulse is
delivered, one complete NMR spectrum is obtained
with each pulse. Often the results of hundreds, thou-
sands, or even hundreds of thousands of pulses are
added to provide greater sensitivity. With good tem-
perature control this may be accomplished over periods
of minutes to days.
489,490
Free induction decay. The strong exciting RF
pulse is delivered with an orientation at right angles
to that of the static field H
o
of the magnet and whose
direction defines the z axis. As a result of this pulse,
the magnetization of a nucleus is tilted away from the
z axis and precesses around the z axis at its resonance
(Larmor) frequency, which is ~500 MHz for
1
H in a
500-MHz spectrometer. The frequency that is measured
is actually a difference from the carrier frequency of
the RF pulse. The precessing magnetization of the
nuclei has a component in the xy plane which induces
an electrical signal in the coil of the NMR probe which
defines the y axis. This signal, which contains the
Larmor frequencies of all of the nuclei of a given ele-
ment, is recorded as a function of time over a period of
a few seconds. The signal decays away exponentially.
However, this curve of free induction decay (FID) is
not smooth but contains within it all of the Larmor
frequencies. If enough points (perhaps 500 in a 2-s
acquisition) are recorded and stored in the computers
memory, Fourier transformation of the data will pro-
duce the frequency-dependent NMR spectrum.
489,490
Relaxation times T
1
and T
2
. When a very strong
pulse of electromagnetic radiation is applied in the
NMR spectrometer, virtually all of the nuclei are
placed in the magnetically excited state. If another
pulse were applied immediately, little energy would
be absorbed because the system is saturated. In the
I. Nuclear Magnetic Resonance (NMR)
CH
3
04.0
CH
2
1.14.4
CH 2.45
Peptide H
random coil
a
3.95.0
OH
b
~ 56
58
NH
2
31 37
NH
Peptide 712 103 142
Aromatic H 79
Imidazole
C
1
H ~7.7
C
2
H ~7.0
NH ~10 165 180
Imidazolium
C
1
H ~8.7
C
2
H ~7.4
NH 1018
CHO 9.410.4
Aldehyde
COOH 11.312.2
Indole NH ~10 130 145
Guanidinium
N

2
69 77
N

H 3137
TABLE 3-3
Approximate Chemical Shift Ranges in
1
H- and in
15
N- NMR Spectra
Group
1
H chemical shift
15
N chemical shift
(ppm from TMS) (ppm from liquid NH
3
)
a
See Wishart et al.
448
b
See Linderstrm-Lang
449
C C
H
142 Chapter 3. Determining Structures and Analyzing Cells
1 3 4
1
3
( ) k T r
B
older continuous-wave NMR spectrometers, the
energy is always kept small so that little saturation
occurs. However, in FT NMR instruments, the strong
pulses lead to a high degree of saturation. Application
of repeated pulses would produce no useful information
were it not for the fact that the excited nuclei soon
relax back to their equilibrium energy distribution.
Relaxation occurs through interactions of the nuclei
with fluctuating magnetic fields in the environment.
For organic molecules in solution the fluctuations that
are most often effective in bringing about relaxation
are the result of moving electrical dipoles in the imme-
diate vicinity. Even so, relaxation of protons in water
requires seconds.
Relaxation of nuclear magnetic states is character-
ized by two relaxation times. The longitudinal or spin-
lattice relaxation time T
1
measures the rate of relax-
ation of the net magnetic vector of the nuclei in the
direction of H
o
. The transverse or spinspin relax-
ation time T
2
measures the relaxation in the xy plane
perpendicular to the direction of H
o
. The two relax-
ation times can be measured independently. In general,
T
2
< T
1
. For solids, T
2
is quite short (~ 10
5
s) whereas
relaxation times of seconds are observed in solutions.
This lengthening of the lifetime of the excited state in
going from solid to liquid leads to a narrowing of
absorption lines and explains why NMR bands in
liquids are often narrow. However, an increase in
viscosity or a loss of fluidity in a membrane leads to
broadening.
How can T
2
and T
1
be measured? T
2
for fluids can
often be estimated from the width of the band at
half-height (Eq. 3-53). However, pulsed NMR methods
are usually employed, the measurement of T
1
being
especially easy.
An attempt is often made to relate T
1
and T
2
to
the molecular dynamics of a system. For this purpose
a relationship is sought between T
1
or T
2
and the
correlation time
c
of the nuclei under investigation.
The correlation time is the time constant for exponen-
tial decay of the fluctuations in the medium that are
responsible for relaxation of the magnetism of the
nuclei. In general, 1/
c
can be thought of as a rate
constant made up of the sum of all the rate constants
for various independent processes that lead to relax-
ation. One of the most important of these (1/
1
) is for
molecular tumbling.
This equation is closely related to that of rotational
diffusion (Eq. 9-35). Another term is the reciprocal of
the residence time
m
, the mean time that a pair of
dipoles are close enough together to lead to relaxation.
In the usual solvents at room temperature,
c
is of
the order of 10
12
to 10
10
s. Thus, relaxation rates in
solutions are considerably faster than the frequencies
of radiation absorbed in the NMR spectrometer (~ 10
8
s
1
). Relaxation is relatively ineffective and T
1
and T
2
are usually large and equal. Bands remain sharp. As
the correlation time increases (as happens, for example,
if the viscosity is increased), T
1
and T
2
decrease with T
1
reaching a minimum when
c
~ v, the frequency of the
absorbed radiation. Lines are broadened and hyper-
fine lines (from coupling between nuclei) cannot be
resolved. As
c
is increased further, T
2
reaches a con-
stant low value, while T
1
rises again. NMR measure-
ments can be made in the region where
c
exceeds ,
a circumstance that is favored by the use of high-
frequency spectrometers. On the other hand, in fluids
it is more customary to work in the range of extreme
motional narrowing at low values of
c
. Both T
1
and
T
2
rise as the mobility of the molecules increases.
A limitation of use of NMR measurements of
proteins comes from the increase in tumbling time
with increasing size of the molecules. Since 1/
r
is
often the most important term in the relaxation rate
constant, only small proteins of mass < 20 kDa give
very sharp bands. Nevertheless, usable spectra are
often obtainable on proteins ten times this size.
A practical problem in
13
C NMR arises from slow
relaxation (long T
1
). Partial saturation is attained and
signal intensities are reduced for those carbon atoms
for which relaxation is especially ineffective. Relax-
ation times can be measured separately for each carbon
atom in a molecule and can yield a wealth of informa-
tion about the segmental motion of groups within a
molecule. Although the relationships between relax-
ation times and molecular motion are complex, they
are often relatively simple for
13
C. Carbon atoms are
usually surrounded by attached hydrogen atoms, and
dipoledipole interactions with these hydrogen atoms
cause most of the nuclear relaxation. For a carbon
atom attached to N equivalent protons in a molecule
undergoing rapid tumbling, Eq. 3-55 holds. Where
h = h/2 and
C
and
H
are the magnetogyric ratios of
carbon and hydrogen nuclei.
This equation permits a calculation of an effective
correlation time
eff
for each carbon atom.
491
T
1
and T
2
can also be evaluated for individual
15
N or
13
C nuclei
in labeled proteins.
491
Two-dimensional and multidimensional NMR
spectra. Proteins have such complex NMR spectra that,
except for small regions at the upfield and downfield
(3-53) T
2
1
1
1
2
2 2
6
T
Nh
r


c H eff
(3-55)
(3-54)
143
ends (see Fig. 3-26A), it is impossible to interpret one-
dimensional spectra. A solution to this problem came
from the development by Jeener, Ernst, and Freeman
of methods of displaying NMR spectra in two dimen-
sions.
428,490,492,493
All two-dimensional and multidimen-
sional NMR methods make use of one basic procedure:
After the initial RF pulse, a second or a series of subse-
quent RF pulses are introduced. This is done before
the nuclei have had time to relax completely. The time
t
1
from the initial pulse to the second pulse is called
the evolution period and allows accumulation of
information about NOEs or J- coupling, whether homo-
nuclear (e.g.
1
H
1
H or
13
C
13
C) or heteronuclear (e.g.,
1
H
13
C or
1
H
15
N).
Two-dimensional spectra usually require hours or
days of acquisition because separate FIDs are collected
I. Nuclear Magnetic Resonance (NMR)
COSY
NOESY
9
8 7
ppm
8
7
5
4
3
p
p
m
9
Figure 3-25 (A) Alpha-carbon plot of the
structure of ribosomal protein L30 from E. coli
as deduced by NMR spectroscopy and model
building. (B) Combined COSY-NOESY dia-
gram for ribosomal protein L30 used for eluci-
dation of d
NN
connectivities (see Fig. 3-27). The
upper part of the diagram represents the finger-
print region of a COSY spectrum recorded for
the protein dissolved in H
2
O. The sequential
assignments of the crosspeaks is indicated. The
lower part of the diagram is part of a NOESY
spectrum in H
2
O. The d
NN
walks are indicat-
ed by : () S11-A12; () H19 to L26; (- - - -)
I43 to S51; (- - - -) G32 to H33; () F52 to
V54. From van de Ven and Hilbers.
494
20
25
30
15
50
35
45
40
10
5
55
58
1
A B
for a series of many different values of t
1
. This provides
a second timescale for the experiment. The second
pulse is often used to rotate the directions of magneti-
zation of the nuclei that are being observed from the
xy plane into the yz plane but with the z component at
180 to the H
o
vector. One very important type of two-
dimensional plot is the NOESY (NOE spectroscopy)
spectrum, which detects NOEs between all excited
nuclei that are close enough together (Fig. 3-25B). The
transfer of magnetization occurs during a mixing
time , which follows the second pulse. For a protein
may be 25 300 ms. A third 90 pulse returns the z
components of the magnetization to be parallel with
the y axis and the FID is collected over the period t
2
.
The amplitude of the resonances detected is modulat-
ed by the frequencies that existed during the evolution
144 Chapter 3. Determining Structures and Analyzing Cells
period. After Fourier analysis
436,490
a two-dimensional plot with two
frequency axes is generated and is
usually displayed as a contour plot.
Along the diagonal of Fig. 3-25B are
peaks representing the one-dimen-
sional spectrum. All of the peaks off
of the diagonal are NOEs which can
be related back to the peaks on the
diagonal as shown by the horizontal
and vertical lines.
A second important two-dimen-
sional method is correlation spec-
troscopy (COSY), in which the
pairs of off-diagonal peaks result
from spinspin coupling. A related
method called TOCSY provides
correlations that extend through
more than three bonds. The COSY
plot in Fig. 3-26B is for the synthetic
cyclic decapeptide cyclo-(
3
-Pro D-
p-Cl-PheD-Trp Ser Tyr D-Trp N-
Me-Leu Arg Pro -Ala). It was
obtained in six hours on a 500-MHZ
instrument. The region marked I
reveals couplings of protons on -
carbons to those on adjacent car-
bons within the same residue (J

;
Fig. 3-27) and other couplings with-
in the side chain. Each amino acid
has a characteristic pattern. From
careful study of this region it is
possible to correlate each -H reso-
nance with a particular amino acid
side chain. However, some residues
are difficult to distinguish, e.g., His,
Trp, Phe, and Tyr have similar J

values. Region II of Fig. 3-26B


reveals connectivities of and
hydrogens to N-H protons in the
7 9 ppm region. Each -H is cou-
pled to the N-H of the same residue
(J
H
, Fig. 3-27). A section of a COSY
plot is also shown in Fig. 3-25B and
indicates how resonances can be
related to those in the NOESY plot
made on the same sample.
Of great importance in the
determination of protein structures
is the use of
15
N- or
13
C-enriched
samples to obtain isotope-edited
spectra. For example in HSQC or in
15
N-multiple quantum cohenence
(HMQC) spectra we see only NH
protons in a plot of
1
H chemical
shift in one dimension versus the
15
N chemical shift of the attached
Cosy
Sulfolane
I
IV
V
II
III
10.0
8.0
6.0
4.0
2.0
0.0
10.0 8.0 6.0 4.0 2.0 0.0
PPM
PPM
(ppm)
-CH
3
N-CH
3
Solvent Solvent
CH
a
s

3
-Pro
Amides and aromatics
Trp
Indole
NHs
10 9 8 7 6 5 4 3 2 1
Figure 3-26 Proton NMR spectra of a cyclic decapeptide analog of the gona-
dotropin-releasing hormone in the solvent sulfolane at 500 MHz. (A) One-
dimensional spectrum. This figure also illustrates upfield methyl group
regions, -hydrogen, amide, aromatic, and downfield (10 20 ppm) regions.
The indole NH resonances are shifted downfield by the ring current of the
indole. (B) COSY spectrum plotted as a contour map. The outlined areas
represent five unique J-coupled regions; area I, the C

to C

to C

etc., side
chain connectivities; area II, the NH to C

connectivities; area III, the indole


NH of tryptophan; area IV, the connectivities of the
3
-Prol residue; and area
V, the aromatic resonances. Peaks that appear on the solid vertical line and the
dashed diagonal lines are artifacts. From Baniak et al.
495
B
A
145 I. Nuclear Magnetic Resonance (NMR)
nitrogen atom in the other (Fig. 3-28). Furthermore,
as shown in this figure, particular types of NH bonds
(peptide, NH
2
, imidazole, indole, amide, and guani-
dinium) appear in different regions. There is only one
peptide NH per residue and, for a small protein, each
may be separately visible.
4. Three-Dimensional Structures and
Dynamics of Proteins
The first three-dimensional structure of a small
protein was determined solely from NMR measure-
ments in 1984. To date hundreds of NMR structures
have been deduced. For small proteins of M
r
< 10,000
two-dimensional COSY and NOESY spectra can suffice.
For larger proteins use of
15
N- and/or
13
C-enriched
proteins is essential.
428,430,496
This permits generation
of a third and even a fourth frequency axis and three-
and four dimensional NMR. For example, using
various complex pulse sequences an
15
N-correlated
1
H
2
H NOESY spectrum can be generated. This
shows directly which NOEs arise from NH protons
with resonances in the amide region of an HSQC or
HMQC spectrum (Fig. 3-28). With both
15
N and
13
C
present, many additional coupling patterns and J
values can be observed.
497 502
It is also possible to
measure NOEs from atoms in a protein to those of a
relatively weakly bound ligand such as a coenzyme or
substrate analog and to determine the conformation of
the bound ligand from this transferred NOE.
503 505
Assignment of resonances. After acquisition
of the necessary data, which may require 10 15 mg of
protein, the observed resonances must be assigned to
specific amino acid residues in the peptide chain. The
connectivities of the individual CH and NH groups
that have been identified in the COSY spectrum and
information about the relationship of one atom to
another atom nearby in space are required. The closest
neighbors to either H or peptide NH protons are often
protons in a neighboring residue (d
N
, d
NN
, Fig. 3-27).
As was pointed out in the preceding section, NOE
correlations obtained from plots such as that in Fig.
3-25B provide much of the information needed to
establish which resonances belong to each residue in a
known sequence.
494
The three-dimensional structure
of the ribosomal protein L30 of E. coli. (Fig. 3-25A) was
deduced entirely by NMR spectroscopy. A downfield
part of the NOESY and COSY plots used is shown in
Fig. 3-25B. This figure also shows how cross-peaks in
the NOESY spectrum were correlated with identified
COSY peaks. It is helpful initially to hunt for unique
dipeptides that can be identified in the NOESY spec-
trum. Ambiguities that arise can be resolved by use of
various additional techniques.
NOEs and distance constraints. NOESY plots
also contain the essential information needed to deter-
mine which side chains distant in the sequence are close
together in space. A NOE observed for a pair of nuclei
falls off as the inverse sixth power of the distance
between them. For this reason, NOEs are observed
only for pairs of atoms closer than about 0.4 nm. It is
possible, in principle, to calculate distances between
nuclei from the NOE intensity, but this is not accurate.
Often, the NOE cross-peaks are grouped into three
categories that correspond to maximum possible
distances of 0.25, 0.30, and 0.40 nm. These can be
related for the most part to intraresidue (e.g., d
N
of
Fig. 3-27) sequential (e.g., d
N
and d
NN
, Fig. 3-27) and
long range backbonebackbone distances. These
values constitute a series of distance constraints
which are applied while making an automated com-
puter search for a folding pattern that will meet these
constraints and at the same time have acceptable
torsion angles and good side chain packing throughout.
This process makes use of distance geometry algorithms
and other methods.
430,506,507
An early success was the
solution of a 75-residue amylase inhibitor indepen-
dently by crystallographers
508
and NMR spectrosco-
pists.
509
The NMR structure was based on 401 NOE
distance constraints, 168 distance constraints imposed
by hydrogen bonds and 50 torsion angles deduced
from J values. Recently, refinement of NMR structures
has been done as in X-ray crystallography.
507
Some
structures have been refined using both NMR and
X-ray data.
442
The spectra in Figs. 3-25, 3-26 and 3-28 are for
relatively small proteins. Spectra of larger proteins are
more complex and lines are broader. Many techniques
are used to simplify spectra. The NMR spectrum of a
protein is simplified considerably if the protein is dena-
tured by heating, and
1
H NMR spectra of random
coil proteins can be predicted well from tables of
standard chemical shifts for the individual amino
acids.
510
Many amide NH protons exchange with
solvent rapidly, making it easier to assign the remaining
peaks. However, nearly all NH peaks will be seen in
an HMQC or HSQC (Fig. 3-28) spectrum. Partial, or
even complete, substitution of deuterium for hydrogen
will also simplify spectra.
511 513
Microorganisms that
will grow in a medium rich in D
2
O can be used as
sources of partially deuterated proteins. Because the
remaining protons usually have
2
H rather than
1
H as
a neighbor, dipolar line broadening is reduced and
sharper resonances are observed.
514
Substitution of
15
N for
14
N in the backbone amide groups can also
yield spectra with narrower lines.
515
Isotope-edited
NMR spectra allow simplification of complex two-
dimensional spectra by observation of only those
protons attached to an isotopically labeled nucleus,
e.g.,
13
C or
15
N.
516 518
Measurement of
15
NH-C
H
J
couplings facilitates structure determinations.
519
146 Chapter 3. Determining Structures and Analyzing Cells
Figure 3-27 Illustration of some distances (d
NN
, d
N
and d
N
obtained from NOESY spectra and some coupling constants (J

and J
N
obtained from
1
H COSY spectra or J-resolved spectra.
The coupling constants and proton chemical shifts provide a
fingerprint for each residue and J
N
may also provide an estimated
value for torsion angle f. The distances establish residue-to-residue
connectivities as well as distance constraints that may permit a
calculation of three-dimensional structure. Additional coupling
constants can be measured for
15
N- or
13
C- enriched proteins.
Coupling from -hydrogens to other side chain hydrogens provides
fingerprint information about individual residues.
C
C
C C
H
H
H
H
H
O
N
N

J
N
d
NN
d
N
d
N

d
N
*
*
Figure 3-28 A
15
N
1
H HSQC spectrum of partially denatured 129-residue hen lysozyme. Boxes enclose the tryptophan
indole region (upper left), the arginine side chain N

region (upper left), and a portion of the amide NH region (lower center
and enlarged in the insert). Resonances of pairs of hydrogen atoms in side chain (Asn and Gln) amide groups are indicated by
horizontal lines. From Buck et al.
524
147
Many aspects of the dynamics of the action of enzymes
of this size can be studied by NMR spectroscopy. New
approaches allow study of proteins up to ~30 kDa in
size.
519a
Computer programs can analyze data and
make automatic assignments of resonances.
520,521
Also useful are techniques for measuring NMR
spectra on solids, including microcrystalline pro-
teins
522
(see also Box 9-C). In some cases, e.g., for
heme proteins,
522,523
the crystals can be oriented in
a magnetic field permitting measurements of NMR
spectra with more than one orientation of the crystals.
This potentially affords more information than the
usual techniques. For example, growth of bacteria in
2
H
2
O media containing
1
H-containing pyruvate yields
proteins with almost complete deuteration in the C-
an C- positions but with highly protonated methyl
groups. This gives rise to good CH
3
to CH
3
NOEs
and provides other advantages in both NMR spectros-
copy and mass spectrometry.
512
Isotopically enriched
amino acids, e.g.,
13
C-enriched leucine,
525
or isoleucine
containing
15
N,
13
C, and
2
H as well
526
can be fed to
growing bacteria. The use of paramagnetic shifts by
ions such as Gd
3+
may be helpful.
527
Very large shifts
are sometimes induced in heme proteins by the Fe
3+
of the heme that is embedded within the protein.
528
5. Other Information from NMR Spectra
NMR titrations. How do pH changes affect NMR
resonances? Resonances of
1
H nuclei close enough to
a proton with a pK
a
in the pH region under study will
experience a shift, which may be either upfield or down-
field when the proton dissociates. The resonance of
13
C in a carboxyl group will shift downfield when the
proton on the carboxyl group dissociates. If the proton
that dissociates is tightly hydrogen bonded in a protein,
its rate of dissociation may be slow compared to the
NMR frequency used. If so, the original peak will
decrease in value as the pH is raised and a new peak
will appear at a position characteristic of the dissociated
form. However, protons attached to N or O are usually
in rapid exchange with the solvent. In this case, the
NMR resonance of the nucleus being observed will
move continuously from one chemical shift value at low
pH to a different one at high pH. In an intermediate
case the resonance will shift and broaden.
Both the C
2
-H and C
1
-H protons of histidine can
often be seen in proteins (Fig. 3-29A).
529 531
As is shown
in Fig. 3-29 A and B, their chemical shifts are strongly
dependent upon the state of protonation of the ring
nitrogen atoms. At low pH, the positive charge that is
shared by the two NH groups attracts electrons away
from both CH positions, causing deshielding of the CH
protons. The effect is greater for the C
1
-H than for
C
2
-H protons (Table 3-3). If the groups being titrated
do not interact strongly with other nearby basic or
I. Nuclear Magnetic Resonance (NMR)
Figure 3-29 (A)
1
H NMR spectra of human myoglobin in
D
2
O showing the C-H
1
resonances of the imidazole rings of
histidines 36, 48, 81, and 119 at three values of the apparent
pH (pH*). From Bothelho and Gurd.
529
(B)
1
H NMR titration
curves for the histidine C
1
and C-
2
ring protons of glycyl-L-
histidylglycine. Data obtained at 100 MHz using 0.1 M
tripeptide in D
2
O containing 0.3 M NaCl. The pH values
(labeled pH
*
) are uncorrected glass electrode pH meter
readings of D
2
O solutions using an electrode standardized
with normal H
2
O buffers. Chemical shifts are downfield
from TMS. Courtesy of J. L. Markley.
438
81
48
119
36 pH* 5.1
119 48
81
36
pH* 7.0
pH* 9.8
1
H chemical shift (ppm)
8.30 7.90 7.50 7.0 8.70
48
119
81
36
8.6
8.4
8.2
8.0
7.8
7.6
7.4
7.2
7.0
5 6 7 8 9
pH*
C
h
e
m
i
c
a
l

s
h
i
f
t

H3N
N
N COO
O
O
H
N
C N
C
H H
1

2
1
H
2

H
+
+
H
C -H
1
C -H
2
B
A
148 Chapter 3. Determining Structures and Analyzing Cells
acidic groups, pK
a
values can be estimated for individ-
ual histidines.
530,532
With
15
N-containing proteins the
tautomeric states (NH
2
vs NH
1
) of the imidazole
rings can also be deduced.
533
Observation of imidazole
rings shows that in proteins pK
a
values of imidazolium
groups are sometimes less than five and sometimes
greater than ten.
463,534
Observing exchangable protons.
1
H spectra of
proteins are often recorded in D
2
O because of inter-
ference from the very strong absorption of H
2
O at
approximately 4.8 ppm. However, resonances of some
rapidly exchanging protons are lost. Special pulse
sequences, as well as improvements in spectrometer
design, allow many of these resonances to be seen in
H
2
O.
535 537
At the far downfield end (>10) of
1
H NMR
spectra there are often weak peaks arising from NH
protons of imidazole or indole side chains that can be
observed in H
2
O.
463,538 540
These resonances are often
shifted 2 5 ppm downfield from the positions given
in Table 3-3. The
1
H resonance for a carboxyl ( COOH)
proton is shifted to 20 ppm or more in spectra of very
strongly hydrogen-bonded anionic complexes such as
the malonate dianion.
541 544
(see Chapter 9, Section D,4).
The strongest hydrogen bonds cause the greatest down-
field shifts because the negative charge pulls the
1
H
proton away from the electrons of the atom to which
it is attached, deshielding the proton.
545
A good example is provided by the imidazole NH
proton of the active site of trypsin and related serine
proteases (Chapter 12), which is seen at ~16 ppm.
Another example is provided by aspartate aminotrans-
ferase, whose
1
H NMR spectrum in a dilute aqueous
phosphate buffer is shown in Fig. 3-30. The peak
labeled A is the resonance of the NH proton on the
ring of the pyridoxal phosphate coenzyme (marked in
Fig. 14-6) and peak B belongs to an adjacent imidazole
group of histidine 143. Both of these protons move
with pH changes around a pK
a
of ~ 6.2 which is associ-
ated with the Schiff base proton 6 8 nm away.
534,546 551
Peak A moves upfield 2.0 ppm and peak B downfield
1.0 ppm when the pH is raised around this pK
a.
These
hydrogen-bonded protons act as sensitive reporters
of the electronic environment of the active site. Many
proteins contain carboxylate or phosphate groups in
their active sites and observation of NMR resonances
of protons hydrogen bonded to them or to groups in
substrates or inhibitors may be a useful technique for
study of many enzymes and other proteins.
Exchange rates of amide protons. The NH
protons of the peptide backbone can be observed in H
2
O
in the 6 11 ppm region. If the spectrum is recorded
for a sample in D
2
O, many of these resonances disap-
pear gradually as the protons of the peptide units
exchange with the deuterium ions of the medium. A
study of the observed exchange rates can shed light on
Figure 3-30 Spectra of the pyridoxal phosphate (PLP),
pyridoxamine phosphate (PMP) and apoenzyme forms of
pig cytosolic aspartate aminotransferase at pH 8.3, 21C.
Some excess apoenzyme is present in the sample of the PMP
form. Spectra were recorded at 500 MH
2
. Chemical shift
values are in parts per million relative to that of H
2
O taken
as 4.80 ppm at 22C. Peak A is from a proton on the ring
nitrogen of PLP or PMP, peaks B and D are from imidazole
NH groups of histidines 143 and 189 (see Fig. 14-6), and peaks
C and D are from amide NH groups hydrogen bonded to
carboxyl groups.
17 16 15 14 13 12 11 10
Chemical shift
+ PLP
+ PMP
Apoenzyme
A
B
C
D
D
E
E
Apo
B C D
F
F
G
the dynamics of protein molecules in solution. Exchange
of amide protons in proteins with
2
H or
3
H has been
studied on a relatively slow timescale by such tech-
niques as observation of infrared vibrations of the
amide group (see Chapter 23).
552,553
However, the
development of two-dimensional NMR dramatically
improved the ability to study proton exchange.
552,554
556
The measurements may involve use of quenching
by rapid solvent change,
556
special pulse sequences,
555
and study of T
1
relaxation rates (seconds timescale).
Recently, electrospray mass spectrometry has also
been exploited.
557,558
Exchange patterns for large
proteins may be followed,
559
and proteolytic fragmen-
tation into short peptides may be used after various
lengths of exchange time to investigate the exchange
in specific regions of a protein.
216,560
Unfolding of a
protein under denaturing conditions can be studied,
as can refolding of a completely denatured protein.
561
Amide hydrogen exchange is usually discussed
in terms of a model proposed by Linderstrm-Lang
in 1955.
554,556
He suggested that portions of protein
molecules unfold sporadically to allow rapid exchange
which can be catalyzed by H
+
, HO

, or other acids or
149
bases. The pH dependence of exchange for a given
amide NH can be described as follows:
k
ex
= k
H
[H
+
] + k
OH
[OH

] + k
w
where k
H
, k
OH
, and k
w
are rate constants for acid-cata-
lyzed, base-catalyzed, and the very slow water-cata-
lyzed exchange.
554,556,562
While many amide protons
exchange rapidly, hydrogen-bonded NH protons in
well-packed hydrophobic core regions exchange slow-
ly,
554,563
sometimes remaining in the protein for years
in a D
2
O solution. Some unusually stable small pro-
teins such as the seed protein crambin show little
exchange. The C
1
protons of histidine imidazoles
also exchange slowly with D
2
O from the medium.
The exchange rates
529,564
are rapid at higher tempera-
tures, with an average half-life of about 11 min at 65C.
Different residues may exchange at different rates.
Binding of substrates or inhibitors can stabilize the
protein, slowing all exchange rates of both amide and
imidazole groups.
Solid-state NMR and other topics. Little can
be said about these topics, but NMR measurement
on solid crystalline materials is now practiced and
is providing a wealth of information. It is being
applied more often to biochemically related prob-
lems.
465,542,565 567
NMR spectroscopy of nucleic acids is discussed
briefly in Chapter 5. An important medical application
of NMR is in imaging, a topic dealt with in Box 30-A.
J. The Protein Data Bank, Three-Dimensional
Structures, and Computation
The x,y,z coordinates of all atoms in published,
refined three-dimensional structures have been depos-
ited in the Protein Data Bank (Table 3-4).
568 571
Many
other related databases are available,
572
e.g., covering
molecular modeling,
573
gene sequences, proteome
data,
574
and much, much more. A good way to keep
up to date is to read the computer corner in Trends
in Biochemical Sciences (TIBS). Most databases can be
reached on the World Wide Web.
572
A selected list is
J. The Protein Data Bank, Three-Dimensional Structures, and Computations
(3-56)
TABLE 3-4
Selected World Wide Web Servers Related to Protein Structures and Sequences
a
a
From Holm and Sander,
570
Hogue et al.,
573
Laskowski et al.,
580
and Walsh et al.
572
Question / area Tool Access / URL
Database search by comparison Dali server http://www.embl-heidelberg.de/dali/dali.html
of 3D structures
Structural classification of proteins SCOP http://www.bio.cam.ac.uk/scop/
Summary and analysis of PDP structures PDBSum http://www.biochem.ucl.ac.uk/bsm/pdbsum
Retrieve 3D coordinates Protein Data Bank http://www.rcsb.org/pdb/
SWISS-PROT sequence database, ExPASy http://expasy.hcuge,ch/
Swissmodel homology modeling, etc.
Molecular graphics viewer for RasMol http://www.bernstein-plus-sons.com/software/rasmol
PCs and workstations
World Wide Web -Entrez and http://www.ncbi.nlm.nih.gov
Molecular Modeling Database access
Protein Science Kinemages MAGE and PREKIN http://www.prosci.uci.edu/kinemages/
Kinemagelndex.html
Pedros Biomolecular Research Tools http://www.fmi.ch/biology/research_tools.html
Predict secondary structure from sequence Predict Protein server http://www.sander.embl-heidelberg.de
Browse databanks in molecular biology SRS server http://www.embl-heidelberg/srs/srsc
The Human Genome Database http://gdbwww.gdb.org/
Image Library of Biological Macromolecules http://www.imb-jena.de/IMAGE.html
Bacterial Nomenclature http://www.gbf-braunschweig.de/DSMZ/
bactnom/bactnam.htm
150 Chapter 3. Determining Structures and Analyzing Cells
1. Strang, R. (1981) Trends Biochem. Sci. 6, VII VIII
2. Michaelis, L. (1922) Die Wasserstoffionenkon-
zentration, p. 48, Springer-Verlag, Berlin
3. Dixon, M., and Webb, E. C., eds. (1979)
Enzymes, 3rd ed., pp.138163, Academic
Press, New York
4. Dryer, R. L., and Lata, G. F. (1989) Experimental
Biochemistry, Oxford Univ. Press, New York
5. Kuramitsu, S., and Hamaguchi, K. (1980)
J. Biochem. 87, 1215 1219
6. Beynon, R. J., and Easterby, J. S. (1996) Buffer
Solutions: The Basics, IRL Press, Oxford
7. Bates, R. G. (1964) Determination of pH, Wiley,
New York
8. Stoll, V. S., and Blanchard, J. S. (1990) in Guide
to Protein Purification (Deutscher, M. P., ed),
pp. 2438, Academic Press, San Diego,
California
9. Ellis, K. J., and Morrison, J. F. (1982) Methods
Enzymol. 87, 405426
10. Good, N. E., Winget, G. D., Winter, W.,
Connolly, T. N., Izawa, S., and Singh, R. M. M.
(1966) Biochemistry 5, 467 477
11. Ferguson, W. J., Braunschweiger, K. I.,
Braunschweiger, W. R., Smith, J. R.,
McCormick, J. J., Wasmann, C. C., Jarvis, N.
P., Bell, D. H., and Good, N. E. (1980) Anal.
Biochem. 104, 300310
12. Dawson, R. M. C., Elliot, D. C., Elliot, W. H.,
and Jones, K. M. (1986) Data for Biochemical
Research, 3rd ed., Oxford Science Pub., London
13. Robyt, J. F., and White, B. J. (1990) Biochemical
Techniques, Theory and Practice, 2nd ed.,
Waveland Press, Inc., Prospect Height, Ill
14. Cooper, T. G. (1977) The Tools of Biochemistry,
Wiley, New York
15. Alexander, R. R., and Griffiths, J. M. (1992)
Basic Biochemical Methods, 2nd ed., Wiley, New
York
16. Wilson, K., and Walker, J., eds. (1994)
Principles and Techniques of Practical Biochemistry,
4th ed., Cambridge Univ. Press, New York
17. Boyer, R. F. (1992) Modern Experimental
Biochemistry, 2nd ed., Benjamin-Cummings
Publ., Redwood City, California
18. le Maire, M., Chabaud, R., and Herve, G.
(1991) Laboratory Guide to Biochemistry,
Enzymology, and Protein Physical Chemistry,
Plenum, New York
19. Fini, C., Floridi, A., and Finelli, V. N., eds.
(1989) Laboratory Methodology in Biochemistry,
CRC Press, Boca Raton, Florida
20. Scopes, R. K. (1993) Protein Purification:
Principles and Practice, 3rd ed., Springer, New
York
21. Harris, E. L. V., and Angal, S. (1990) Protein
Purification: A Practical Approach, IRL Press,
Oxford, 2 volumes
22. Deutscher, M. P., ed. (1990) Guide to Protein
Purification: Methods in Enzymology, Vol. 182,
Academic Press, San Diego, California
23. Celis, J. E., ed. (1994) Cell Biology: A Laboratory
Handbook, Academic Press, New York (3 volumes)
24. Fasman, G. D., ed. (1989) CRC Practical
Handbook of Biochemistry and Molecular Biology,
CRC Press, Boca Raton, Florida
25. Rosenberg, I. M. (1996) Protein Analysis and
Purification, Birkhuser, Cambridge,
Massachusetts
26. Walker, J. M., ed. (1996) The Protein Protocols
Handbook, Humana Press, Totowa, New Jersey
27. Marshak, D. R., Kadonaga, J. T., Burgess, R.
R., Knuth, M. W., Brennan, W. A., Jr., and Lin,
S.-H. (1996) Strategies for Protein Purification
and Characterization, Cold Spring Harbor Lab.
Press, Cold Spring Harbor, New York
28. Creighton, T. E., ed. (1989) Protein Structure: A
Practical Approach, IRL Press, Oxford
29. Marshak, D. R., Kadonaga, J. T., Burgess, R.
R., Knuth, M. W., Brennan, W. A., and Lin, S.-
H. (1995) Strategies for Protein Purification and
Characterization, Cold Spring Harbor Lab.
Press, Cold Spring Harbor, New York
30. Crabb, J. W., ed. (1995) Techniques in Protein
Chemistry, Vol. VI, Academic Press, San Diego,
California
31. Walker, J. M., ed. (1994) Basic Protein and
Peptide Protocols, Humana Press, Totowa, New
Jersey
32. Bollag, D. M., and Edelstein, S. J. (1990)
Protein Methods, Wiley-Liss, New York
33. Creighton, T. E., ed. (1989) Protein Structure
and Protein Function: A Practical Approach, IRL
Press, Oxford
34. Copeland, R. A. (1994) Methods for Protein
Analysis. A Practical Guide to Laboratory
Protocols, Chapman and Hall, New York
35. Matsuidaira, P. T., ed. (1995) A Practical Guide
to Protein and Peptide Purification for Micro-
sequencing, Academic Press, San Diego, CA
36. Hugli, T. E., ed. (1989) Techniques in Protein
Chemistry, Academic Press, New York
37. Means, G. E., and Feeney, R. E. (1973)
Chemical Modification of Proteins, Holden-Day,
San Francisco
38. Keleti, G., and Lederer, W. H. (1974) Handbook
of Micromethods for the Biological Sciences, Van
Nostrand-Reinhold, New York
39. Switzer, R. L., and Garrity, L. F. (1999)
Experimental Biochemistry:Theory and Exercises
in Fundamental Methods, 3rd ed., Freeman,
New York
40. Schulz, G. E., and Schirmer, R. H. (1979)
Principles of Protein Structure, Springer-Verlag,
New York
References
41. Scopes, R. K. (1994) Protein Purification. Principles
of Protein Structure, Springer-Verlag, New York
42. Villafranca, J., ed. (1990) Current Research in
Protein Chemistry: Techniques, Structure, and
Function, Academic Press, New York
43. Coligan, J., Dunn, B., Ploegh, H., Speicher, D.,
and Wingfield, P., eds. (1995) Current Protocols
in Protein Science, Wiley, New York
44. Matsudaira, P. T., ed. (1989) A Practical Guide
to Protein and Peptide Purification for Micro-
sequencing, Academic Press, San Diego,
California
45. Doonan, S., ed. (1996) Protein Purification
Protocols, Humana Press, Totowa, New Jersey
46. Little, P., and Hughes, S., eds. (1992)
Technique, Vol. 4, Academic Press, London
47. Methods: A Companion to Methods in
Enzymology, 1990 present, Academic Press,
San Diego, California
48. Wankat, P. C., Van Oss, C. J., and Henry, J. D.,
Jr., eds. (1992) Separation and Purification
Methods, Dekker, New York
49. protein expression and purification, (1990 present),
Academic Press, San Diego, California
50. Coligan, J., Dunn, B., Ploegh, H., Speicher, D.,
and Wingfield, P., eds. (1996 present) Current
Protocols in Protein Science, Wiley, New York
51. Hirs, C. H. W., and Timasheff, S. N., eds.
(1983) Methods in Enzymology, Enzyme
Structure, Vol. 91, Academic Press, New York
52. Doolittle, R. F., ed. (1990) Methods in
Enzymology, Molecular Evolution: Computer
Analysis of Protein and Nucleic Acid, Vol. 183,
Academic Press, New York
53. Bollag, D. E., Rozycki, M. D., and Edelstein, S.
J., eds. (1996) Protein Methods, 2nd ed., Wiley,
New York
54. Birnie, G. D., and Fox, S. M., eds. (1969)
Subcellular Components, Butterworth, London
55. Estabrook, R. W., and Pullman, M. E., eds.
(1967) Methods in Enzymology, Vol. 10,
Academic Press, New York
56. Kaback, H. R. (1971) in Methods in Enzymology,
Vol. 22 (Jakoby, W. B., ed), pp. 99120,
Academic Press, New York
57. Dingle, J. T. (1972) Lysosomes. A Laboratory
Handbook, North-Holland Publ., Amsterdam
58. Rasmussen, N. (1996) Trends Biochem. Sci. 21,
319321
58a. Jenkins, W. T. (1998) Protein Sci. 7, 376382
59. McPhie, P. (1971) in Methods in Enzymology,
Vol. 22 (Jakoby, W. B., ed), pp. 2332,
Academic Press, New York
60. Blatt, W. F. (1971) in Methods in Enzymology,
Vol. 22 (Jakoby, W. B., ed), pp. 3949,
Academic Press, New York
61. Porath, J., and Flodin, P. (1959) Nature
(London) 183, 16571659
given in Table 3-4. The widely used viewer called RasMol
can be used with your PC or MacIntosh computer
575576a
or UNIX workstation
569,575
. Another way to view
macromolecules is to use the Protein Science Kinemages
(kinetic images) using the program MAGE.
577,578
Although it is mentioned in a few places, this
book does not begin to describe the rapid growth of
computation in biochemistry, biophysics, and biology
in general. Very fast methods of protein structure
determination are being developed.
579,582
One of the
major goals in current computation is to predict folding
patterns of proteins from their sequences.
582,583
By
comparing sequences we can often guess an approxi-
mate structure but accurate predictions are still not
possible. Having a structure, we would like to predict
properties and reactivities and to be able to guess how
two more macromolecules interact to form macromolec-
ular complexes. We would like to understand the
complex chain of nonpolar and electrostatic interac-
tions that underlie the fundamental properties of
catalysis, movement, and responsiveness of organ-
isms. Many computers and ingeneous minds are
working to help us match theory with reality in these
areas. Read the current journals!
151
References
62. Porath, J. (1978) Trends Biochem. Sci. 3, N100
63. Lathe, G. H. (1978) Trends Biochem. Sci. 3,
N99N100
64. Flodin, P., and Aspberg, K. (1961) Biol. Struct.
Funct. Proc. IUB / IUBS Int. Sym, 1st ed., Vol. 1,
pp. 345-349 (p. 346)
65. Weir, M., and Sparks, J. (1987) Biochem. J. 245,
85
66. Regnier, F. E. (1991) Nature (London) 350, 634
635
67. Afeyan, N. B., Gordon, N. F., and Mazsaroff, I.
(1990) J. Chromatogr. 519, 129
67a. Wu, C.-s. (1999) Column Handbook for Size
Exclusion Chromatography, Academic Press,
San Diego, California
68. Liapis, A. I., and McCoy, M. A. (1992) J.
Chromatogr. 599, 87104
69. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Part II, Freeman, San
Francisco, California (pp. 549685)
70. Kyte, J. (1995) Structure in Protein Chemistry,
Garland Publ., New York
71. Brakke, M. K. (1953) Arch. Biochem. Biophys.
45, 275290
72. Britten, R. J., and Roberts, R. B. (1960) Science
131, 3233
73. Martin, R. G., and Ames, B. N. (1961) J. Biol.
Chem. 236, 13721379
74. Meselson, M., Stahl, F. W., and Vinograd, J.
(1957) Proc. Natl. Acad. Sci. U.S.A. 43, 581588
75. da Silva, J. R. R. F. (1978) in New Trends in Bio-
organic Chemistry (Williams, R. J. P., and da
Silva, J. R. R. F., eds), Academic Press, London
76. Arakawa, T., and Timasheff, S. N. (1982)
Biochemistry 21, 65456552
77. Rao, J. K. M., and Argos, P. (1981) Biochemistry
20, 65366543
78. Green, A. A., and Hughes, W. L. (1955) in
Methods in Enzymology, Vol. 1 (Colowick, S. P.,
and Kaplan, N. O., eds), pp. 6790, Academic
Press, New York
79. Dawson, R. M. C., Elliott, D. C., Elliott, W. H.,
and Jones, K. M. (1969) Data for Biochemical
Research, 2nd ed., Clarendon Press, Oxford
80. Ito, Y. (1982) Trends Biochem. Sci. 7, 4750
81. Ito, Y. (1987) Nature (London) 326, 419420
82. Mandava, N. B., and Ito, Y., eds. (1988)
Countercurrent Chromatography, Dekker, New
York
82a. Menet, J.-M., and Thibaut, D., eds. (1999)
Countercurrent Chromatography, Dekker, New
York
83. Er-el, Z., Zaidenzaig, Y., and Shaltiel, S. (1972)
Biochem. Biophys. Res. Commun. 49, 383390
84. Arakawa, T., and Timasheff, S. N. (1982)
Biochemistry 21, 65366544
85. Atha, D. H., and Ingham, K. C. (1981) J. Biol.
Chem. 256, 1210812117
86. Kenney, A., and Fowell, S., eds. (1992)
Practical Protein Chromatography, Humana
Press, Totowa, New Jersey
87. Tiselius, A., Hjerten, S., and Levin, O. (1956)
Arch. Biochem. Biophys. 65, 132
88. Wolf, W. J., and Sly, D. A. (1964) J. Chromatogr.
15, 247250
89. Regnier, F. E. (1987) Science 238, 319323
90. Hutchens, T. W., and Porath, J. (1987)
Biochemistry 26, 71997204
91. Molnr, I., Horvth, C., and Jatlow, P. (1978)
Chromatographia 11, 260265
92. Hamilton, R. J., and Sewell, P. A. (1982)
Introduction to High Performance Liquid
Chromatography, 2nd ed., Chapman and Hall,
London
93. Berridge, J. C. (1985) Techniques for the
Automated Optimization of HPLC Separations,
Wiley, New York
94. Simpson, C. F. (1982) Techniques in Liquid
Chromatography, Wiley, New York
95. Mant, C. T., and Hodges, R. S. (1991) High-
Performance Liquid Chromatography of Peptides
and Proteins: Separation, Analysis, and
Conformation, CRC Press, Boca Raton, Florida
96. Gooding, K. M., and Regnier, F. E., eds. (1990)
HPLC of Biological Macromolecules, Dekker,
New York
96a. Beesley, T. E., Buglio, B., and Scott, R. P. W.
(2000) Quantitative Chromatographic Analysis,
Dekker, New York
97. Oliver, R. W. A., ed. (1989) HPLC of
Macromolecules: A Practical Approach, IRL
Press, Oxford
98. Fried, B., and Sherma, J., (1999) Thin-Layer
Chromatography, 4th ed., Dekker, New York
99. Zlatkis, A., and Kaiser, R. E. (1977) High
Performance Thin-Layer Chromatography,
Elsevier Scientific-North Holland,
Amsterdam
100. Sherma, J., and Fried, B., eds. (1996) Practical
Thin-Layer Chromatography: A Multidisciplinary
Approach, CRC Press, Boca Raton, Florida
101. Liu, M.-K., Li, P., and Giddings, J. C. (1993)
Protein Sci. 2, 15201531
102. Treadwell, G. E., Cairns, W. L., and Metzler,
D. E. (1968) J. Chromatogr. 35, 376388
103. Novotny, M. V. (1989) Science 264, 5157
104. Schomvurg, G. (1990) Gas Chromatography: A
Practical Course, VCH Publ., Cambridge, UK
105. Lee, M. L., and Markides, K. E. (1987) Science
235, 13421347
105a. Muraviev, D., Gorshkov, V., and Warshawsky,
A. (1999) Ion Exchange, Dekker, New York
106. Moore, S., and Stein, W. H. (1949) Cold Spring
Harbor Symp. Quant. Biol. 14, 179190
107. Moore, S., and Stein, W. H. (1949) J. Biol.
Chem. 178, 5377
108. Manning, J. M. (1993) Protein Sci. 2, 11881191
109. Svasti, J. (1980) Trends Biochem. Sci. 5, 89
110. Moore, S., and Stein, W. H. (1951) J. Biol.
Chem. 192, 663681
111. Moore, S., and Stein, W. H. (1954) J. Biol.
Chem. 211, 893906
112. Moore, S., and Stein, W. H. (1963) in Methods
in Enzymology, Vol. 6 (Colowick, S. P., and
Kaplan, N. O., eds), pp. 819831, Academic
Press, New York
113. Yamamoto, S., Nakanishi, K., and Matsuno, R.
(1988) Ion Exchange Chromatography of Proteins,
Dekker, New York
114. Peterson, E. A. (1970) Cellulosic Ion Exchangers,
Elsevier-North Holland, Amsterdam
115. Nlting, B., Golbik, R., Neira, J. L., Soler-
Gonzalez, A. S., Schreiber, G., and Fersht, A.
R. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 826
830
116. Lundell, N., and Markides, K. (1992)
Chromatographia 34, 369375
117. Dean, P. D. G., Johnson, W. S., and Middle, F.
A., eds. (1985) Affinity Chromatography, A
Practical Approach, IRL Press, Oxford
118. Parikh, I., and Cuatrecasas, P. (1985) Chem.
Eng. News 63 (Aug 26), 1732
119. Porath, J. (1968) Nature (London) 218, 834838
120. Inman, J. K. (1974) in Methods in Enzymology,
Vol. 34 (Jakoby, W. B., and Wilchek, M., eds),
pp. 3058, Academic Press, New York
121. Bethell, G. S., Ayers, J. S., Hearn, M. T. W., and
Hancock, W. S. (1981) J. Chromatogr. 219, 361
372
122. Wilchek, M., and Miron, T. (1987) Biochemistry
26, 21552161
123. Ashley, G. W., and Bartlett, P. A. (1984) J. Biol.
Chem. 259, 1361513620
124. Hengen, P. N. (1995) Trends Biochem. Sci. 20,
285286
125. Loo, T. W., and Clarke, D. M. (1995) J. Biol.
Chem. 270, 2144921452
126. Janknecht, R., de Martynoff, G., Lou, J.,
Hipskind, R. A., Nordheim, A., and
Stunnenberg, H. G. (1991) Proc. Natl. Acad. Sci.
U.S.A. 88, 89728976
127. Hochuli, E., Dobeli, H., and Schacher, A.
(1987) J. Chromatogr. 411, 177184
128. Turner, A. J. (1981) Trends Biochem. Sci. 6, 171
173
129. Vesterberg, O. (1971) in Methods in Enzymology,
Vol. 22 (Jakoby, W. B., ed), pp. 389412,
Academic Press, New York
130. Bauer, J. (1994) Cell Electrophoresis, CRC Press,
Boca Raton, Florida
131. Allison, S. A., and Tran, V. T. (1995) Biophys. J.
68, 22612270
132. Shuster, L. (1971) in Methods in Enzymology,
Vol. 22 (Jakoby, W. B., ed), pp. 412433,
Academic Press, New York
133. Chrambach, A., and Rodbard, D. (1971)
Science 172, 440451
134. Andrews, A. T. (1986) Electrophoresis: Theory,
Techniques and Biochemical and Clinical
Applications, 2nd ed., Clarendon-Oxford Univ.
Press, Oxford
135. Gordon, A. H. (1980) Electrophoresis of Proteins
in Polyacrylamide and Starch Gels, Revised ed.,
Elsevier-North Holland, Amsterdam
136. Chrambach, A., Dunn, M. J., and Radola, B. J.,
eds. (1987) Advances in Electrophoresis, Vol. I,
VCH Publ., Cambridge, UK
137. Serwer, P., Easom, R. A., Hayes, S. J., and
Olson, M. S. (1989) Trends Biochem. Sci. 14, 47
138. electrophoresis, (1995 present), VCH Publ.,
New York
139. advances in electrophoresis, (1987 present),
VCH Publ., Cambridge
140. Hawcroft, D. M. (1997) Electrophoresis: The
Basics, IRL Press, Oxford
141. Hengen, P. N. (1995) Trends Biochem. Sci. 20,
202203
142. Garfin, D. E. (1990) Methods Enzymol. 182,
425441
143. Hames, B. D., and Rickwood, D., eds. (1990)
Gel Electrophoresis of Proteins: A Practical
Approach, 2nd ed., IRL Press, Oxford
144. Righetti, P. G., ed. (1996) Capillary Electro-
phoresis in Analytical Biotechnology, CRC Press,
Boca Raton, Florida
145. Camilleri, P., ed. (1993) Capillary Electrophoresis:
Theory and Practice, CRC Press, Inc., Boca Raton
146. Altria, K. D., ed. (1996) Capillary Electrophore-
sis Guidebook, Humana Press, Totowa, New
Jersey
147. Landers, J. P., ed. (1994) Handbook of Capillary
Electrophoresis, CRC Press, Boca Raton, Florida
148. Landers, J. P. (1993) Trends Biochem. Sci. 18,
409414
149. Wehr, T., Rodrguez-Daz, R., and Zhu, M.
(1998) Capillary Electrophoresis of Proteins,
Dekker, New York
149a. Yao, S., Anex, D. S., Caldwell, W. B., Arnold,
D. W., Smith, K. B., and Schultz, P. G. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 5372 5377
150. Valaskovic, G. A., Kelleher, N. L., and
McLafferty, F. W. (1996) Science 273, 11991202
151. Gao, J., Gomez, F. A., Hrter, R., and
Whitesides, G. M. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 1202712030
152. Harrison, D. J., Fluri, K., Seiler, K., Fan, Z.,
Effenhauser, C. S., and Manz, A. (1993) Science
261, 895897
153. Radola, B. J., and Graesslin, D., eds. (1977)
Electrofocusing and Isotachophoresis, de Gruyter,
Berlin
154. Bocek, P., Dem, M., Gebauer, P., and Dolnik,
V. (1987) Analytical Isotachophoresis, VCH
Publ., Cambridge, UK
152 Chapter 3. Determining Structures and Analyzing Cells
210. Smith, R. D., Loo, J. A., Edmonds, C. G.,
Barinaga, C. J., and Udseth, H. R. (1990) Anal.
Chem. 62, 882899
211. Fenn, J. B., Mann, M., Meng, C. K., Wong, S.
F., and Whitehouse, C. M. (1990) Mass
Spectrom. Rev. 9, 37
212. Dole, M., Mack, L. L., Hines, R. L., Mobley, R.
C., Ferguson, L. D., and Alice, M. B. (1968) J.
Chem. Phys. 49, 2240
213. Mann, M., and Wilm, M. (1995) Trends
Biochem. Sci. 20, 219224
214. Mann, M., Meng, C. K., and Fenn, J. B. (1989)
Anal. Chem. 61, 17021708
215. Covey, T. R., Bonner, R. F., Shushan, B. I., and
Henion, J. (1988) Rapid Commun. Mass
Spectrom. 2, 249256
216. Miranker, A., Kruppa, G. H., Robinson, C. V.,
Aplin, R. T., and Dobson, C. M. (1996) J. Am.
Chem. Soc. 118, 74027403
217. Marshall, A. G., Senko, M. W., Li, W., Li, M.,
Dillon, S., Guan, S., and Logan, T. M. (1997) J.
Am. Chem. Soc. 119, 433434
218. Torchinsky, Y. M. (1981) Sulfur in Proteins,
Pergamon, Oxford
219. Friedman, M. (1973) The Chemistry and
Biochemistry of the Sulfhydryl Group in Amino
Acids, Peptides and Proteins, Pergamon, Oxford
220. Jocelyn, P. C. (1972) Biochemistry of the SH
Group, Academic Press, New York
221. Simpson, R. J., Neuberger, M. R., and Liu, T.-Y.
(1976) J. Biol. Chem. 251, 19361940
222. Hugli, T. E., and Moore, S. (1972) J. Biol. Chem.
247, 28282834
223. Chin, C. C. Q., and Wold, F. (1974) Anal.
Biochem. 61, 379391
224. Royer, G. P., Schwartz, W. E., and Liberatore,
F. A. (1977) Methods Enzymol. 47E, 4045
225. Glazer, A. N. (1976) in The Proteins, 3rd ed.,
Vol. II (Neurath, H., and Hill, R. L., eds),
pp. 1103, Academic Press, New York
226. Plapp, B. V., Raftery, M. A., and Cole, R. D.
(1967) J. Biol. Chem. 242, 265270
227. Doonan, S., and Fahmy, H. M. A. (1975) Eur. J.
Biochem. 56, 421426
228. Mitchell, W. M. (1977) Methods Enzymol. 47E,
165169
229. Gadasi, H., Maruta, H., Collins, J. H., and
Korn, E. D. (1979) J. Biol. Chem. 254, 36313636
230. Drapeau, G. R. (1977) Methods Enzymol. 47E,
189191
231. Koida, M., and Walter, R. (1976) J. Biol. Chem.
251, 75937599
232. Hayashi, R. (1977) Methods Enzymol. 47E, 8493
233. Krutzsch, H. C., and Pisano, J. J. (1977)
Methods Enzymol. 47E, 391404
234. Mahoney, W. C., and Hermodson, M. A.
(1979) Biochemistry 18, 38103814
235. Hancock, W. S., ed. (1996) New Methods in
Peptide Mapping for the Characterization of
Proteins, CRC Press, Boca Raton, Florida
236. Lundblad, R. L. (1995) Techniques in Protein
Modification, CRC Press, Boca Raton, Florida
237. Sanger, F. (1949) Biochem. J. 45, 563574
238. Sanger, F. (1988) Ann. Rev. Biochem. 57, 128
239. Ruthen, R. (1993) Sci. Am. 269(Oct), 3031
240. Aebersold, R. H., Leavitt, J., Saavedra, R. A.,
Hood, L. E., and Kent, S. B. H. (1987) Proc.
Natl. Acad. Sci. U.S.A. 84, 69706974
241. Edman, P. (1956) Acta Chem. Scand. 10, 761768
242. Edman, P., and Begg, C. (1967) Eur. J. Biochem.
1, 8091
243. Niall, H. D. (1977) Nature (London) 268, 279280
244. Schlesinger, D. H., ed. (1988) Macromolecular
Sequencing and Synthesis, Liss, New York
245. Shively, J. E., ed. (1986) Methods of Protein
Microcharacterization, Humana Press, Clifton,
New Jersey
155. Righetti, P. G. (1983) Isoelectric Focusing:
Theory, Methodology and Applications, Elsevier -
North Holland Biomedical Press, Amsterdam
156. Catsimpoolas, N., and Drysdale, J., eds. (1977)
Biological and Biomedical Applications of
Isoelectric Focusing, Plenum, New York
157. Egen, N. B., Bliss, M., Mayersohn, M., Owens,
S. M., Arnold, L., and Bier, M. (1988) Anal.
Biochem. 172, 488494
158. Bjellqvist, B., Ek, K., Righetti, P. G., Gianazza,
E., Grg, A., Westermeier, R., and Postel, W.
(1982) J. Biochem. Biophys. Methods 6, 317339
159. Righetti, P. G., Gianazza, E., and Gelfi, C.
(1988) Trends Biochem. Sci. 13, 335338
160. Righetti, P. G. (1990) Immobilized pH Gradients:
Theory and Methodology, Elsevier, Amsterdam
161. Whitney, J. B., III, Cobb, R. R., Popp, R. A.,
and ORourke, T. W. (1985) Proc. Natl. Acad.
Sci. U.S.A. 82, 76467650
162. Madden, M. S. (1995) Anal. Biochem. 229, 203
206
163. Anderson, N. (1979) Nature (London) 278, 122
123
164. Wade, N. (1981) Science 211, 3335
165. Henslee, J. G., and Srere, P. A. (1979) J. Biol.
Chem. 254, 54885491
166. Hanash, S. M., and Strahler, J. R. (1989) Nature
(London) 337, 485486
167. Neukirchen, R. O., Schlosshauer, B., Baars, S.,
Jackle, H., and Schwarz, U. (1982) J. Biol.
Chem. 257, 1522915234
168. Racine, R. R., and Langley, C. H. (1980) Nature
(London) 283, 855857
169. Celis, J. E., and Bravo, R., eds. (1984) Two-
Dimensional Gel Electrophoresis of Proteins:
Methods and Applications, Academic Press,
New York
170. Celis, J. E., and Bravo, R. (1981) Trends
Biochem. Sci. 6, 197201
171. Garrels, J. I. (1989) J. Biol. Chem. 264, 52695282
172. Krauss, M. R., Collins, P. J., and Blose, S. H.
(1989) Nature (London) 337, 669670
173. Wood, T. D., Chen, L. H., Kelleher, N. L.,
Little, D. P., Kenyon, G. L., and McLafferty, F.
W. (1995) Biochemistry 34, 1625116254
173a. McLafferty, F. W., Fridriksson, E. K., Horn, D.
M., Lewis, M. A., and Zubarev, R. A. (1999)
Science 284, 1289 1290
174. Shevchenko, A., Jensen, O. N., Podtelejnikov,
A. V., Sagliocco, F., Wilm, M., Vorm, O.,
Mortensen, P., Shevchenko.A, Boucherie, H.,
and Mann, M. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1444014445
175. Wilm, M., Shevchenko, A., Houthaeve, T.,
Breit, S., Schweigerer, L., Fotsis, T., and Mann,
M. (1996) Nature (London) 379, 466469
176. Hess, D., Covey, T. C., Winz, R., Brownsey, R.
W., and Aebersold, R. (1993) Protein Sci. 2,
13421351
177. Van Holde, K. E. (1985) Physical Biochemistry,
2nd ed., Prentice-Hall, Engelwood Cliffs,
New Jersey
178. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Freeman, San Francisco,
California
179. Svedburg, T., and Pederson, K. O. (1940) The
Ultracentrifuge, Oxford Univ. Press, London
180. Schachman, H. K. (1989) Nature (London) 341,
259260
181. Schachman, H. K. (1992) in Analytical
Ultracentrifugation in Biochemistry and Polymer
Science (Harding, S. E., Rowe, A. J., and
Horton, J. C., eds), pp. 315, Royal Society for
Chemistry, Cambridge, UK
182. Schachman, H. K. (1959) Ultracentrifugation in
Biochemistry, Academic Press, New York
183. Rivas, G., and Minton, A. P. (1993) Trends
Biochem. Sci. 18, 284287
184. Harding, S. E., Rowe, A. J., and Horton, J. C.,
eds. (1992) Analytical Ultracentrifugation in
Biochemistry and Polymer Science, CRC Press,
Boca Raton, Florida
185. Hansen, J. C., Lebowitz, J., and Demeler, B.
(1994) Biochemistry 33, 1315513163
186. Ralston, G. (1993) Introduction to Analytical
Ultracentrifugation, Beckman Instruments,
Fullerton, California
187. Stafford, W. F., and Schuster, T. M. (1995) in
Introduction to Biophysical Methods for Protein
and Nucleic Acid Research (Glasel, J. A., and
Deutscher, M. P., eds), pp. 111145, Academic
Press, San Diego
188. Bowen, T. J. (1970) An Introduction to Ultra-
centrifugation, Wiley (Interscience), New York
189. Bothwell, M. A., Howlett, G. J., and
Schachman, H. K. (1978) J. Biol. Chem. 253,
20732076
190. Pollet, R. J., Haase, B. A., and Standaert, M. L.
(1979) J. Biol. Chem. 254, 3033
190a. CRC Handbook of Biochemistry, (1968) Chem.
Rubber Publ. Co., Cleveland, Ohio
191. Jacobsen, M. P., Wills, P. R., and Winzor, D. J.
(1996) Biochemistry 35, 1317313179
191a. Colowick, S. P., and Kaplan, N. O., eds., (1968)
Methods Enzymol. 12B, 388389
192. Squire, P. G., and Himmel, M. E. (1979) Arch.
Biochem. Biophys. 196, 165177
193. Andrews, P. (1965) Biochem. J. 96, 595606
194. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Freeman, San Fransico,
California (pp. 670684)
195. Nakos, G., and Mortenson, L. (1971)
Biochemistry 10, 455458
196. Mann, K. G., and Fish, W. W. (1972) Methods
Enzymol. 26C, 2842
197. Igou, D. K., Lo, J. T., Clark, D. S., Mattice, W.
L., and Younathan, E. S. (1974) Biochem.
Biophys. Res. Commun. 60, 140145
198. Weber, K., Pringle, J. R., and Osborn, M.
(1972) Methods Enzymol. 26C, 329
199. Wood, H. G., Werkman, C. H., Hemingway,
A., and Nier, A. O. (1941) J. Biol. Chem. 139,
377381
200. Biemann, K. (1995) Protein Sci. 4, 19201927
200a. Niessen, W. M. A. (1998) Liquid Chromatogra-
phy - Mass Spectrometry, 2nd ed., Dekker, New
York
200b. Chapman, J. R., ed. (2000) Mass Spectrometry of
Proteins and Peptides, Hamana Press, Totowa,
New Jersey
200c. Burlingame, A. L., Carr, S. A., and Baldwin,
M. A., eds. (2000) Mass Spectrometry in Biology
and Medicine, Hamana Press, Totowa, New
Jersey
201. Senko, M. W., and McLafferty, F. W. (1994)
Annu. Rev. Biophys. Biomol. Struct. 23, 763785
202. Li, Y., Hunter, R. L., and McIver, R. T., Jr.
(1994) Nature (London) 370, 393395
203. Siuzdak, G. (1994) Proc. Natl. Acad. Sci. U.S.A.
91, 1129011297
204. Caprioli, R. M., and Suter, M. J.-F. (1995) in
Introduction to Biophysical Methods for Protein
and Nucleic Acid Research (Glasel, J. A., and
Deutscher, M. P., eds), pp. 147-204, Academic
Press, San Diego
205. Burlingame, A. L., and Carr, S. A., eds. (1996)
Mass Spectrometry in the Biological Sciences,
Humana Press, Totowa, New Jersey
206. Karas, M., and Hillenkamp, F. (1988) Anal.
Chem. 60, 22992303
207. Tanaka, K., Waki, H., Ido, Y., Akita, S.,
Yoshida, Y., and Yoshida, T. (1988) Rapid
Commun. Mass Spectrom. 2, 151156
208. Beavis, R. C., and Chait, B. T. (1989) Rapid
Commun. Mass Spectrom. 3, 233237
209. Hillenkamp, F., Karas, M., Beavis, R. C., and
Chait, B. T. (1991) Anal. Chem. 63, 1193A1198A
References
153
246. Bhown, A. S. (1988) Protein/Peptide Sequence
Analysis: Current Methodoligies, CRC Press,
Boca Raton, Florida
247. Hunkapiller, M. W., and Hood, L. E. (1983)
Science 219, 650659
248. Totty, N. F., Waterfield, M. D., and Hsuan, J. J.
(1992) Protein Sci. 1, 12151224
249. Gershoni, J. M. (1985) Trends Biochem. Sci. 10,
103106
250. Moos, M., Jr., Nguyen, N. Y., and Liu, T.-Y.
(1988) J. Biol. Chem. 263, 60056008
251. Baldo, B. A., and Tovey, E. R., eds. (1989)
Protein Blotting, S. Karger, Farmington, CT
252. Wellner, D., Panneerselvam, C., and Horecker,
B. L. (1990) Proc. Natl. Acad. Sci. U.S.A. 87,
19471949
253. Bailey, J. M., Nikfarjam, F., Shenoy, N. R., and
Shively, J. E. (1992) Protein Sci. 1, 16221633
254. Bailey, J. M., Shenoy, N. R., Ronk, M., and
Shively, J. E. (1992) Protein Sci. 1, 6880
255. Johnson, R. S., and Walsh, K. A. (1992) Protein
Sci. 1, 10831091
255a. Keough, T., Youngquist, R. S., and Lacey, M. P.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 7131
7136
256. Hunt, D. F., Shabanowitz, J., Yates, J. R., III,
Zhu, N.-Z., Russell, D. H., and Castro, M. E.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 620623
257. Takao, T., Hitouji, T., Shimonishi, Y., Tanabe,
T., Inouye, S., and Inouye, M. (1984) J. Biol.
Chem. 259, 61056109
257a. Jaffe, H., Veeranna, and Pant, H. C. (1998)
Biochemistry 37, 1621116224
257b. Whitelegge, J. P., Gundersen, C. B., and Faull,
K. F. (1998) Protein Sci. 7, 1423 1430
258. Yazdanparast, R., Andrews, P. C., Smith, D.
L., and Dixon, J. E. (1987) J. Biol. Chem. 262,
25072513
259. Brown, J. R., and Hartley, B. S. (1966) Biochem.
J. 101, 214228
260. Svobodova, S., Hais, I. M., and Kostir, J. V.
(1953) Chem. Listy 47, 205
261. Chen, R. F., Smith, P. D., and Maly, M. (1978)
Arch. Biochem. Biophys. 189, 241250
262. Lee, K. S., and Drescher, D. G. (1979) J. Biol.
Chem. 254, 62486251
263. Chen, R. F., Scott, C., and Trepman, E. (1979)
Biochim. Biophys. Acta. 576, 440455
264. Lunte, S. M. et al. (1989) Anal. Biochem. 178,
202207
265. Sharma, Y., Rao, C. M., Rao, S. C., Krishna, A.
G., Somasundaram, T., and Balasubramanian,
D. (1989) J. Biol. Chem. 264, 2092320927
266. Wilson, C. M. (1983) Methods Enzymol. 91,
236247
267. Merril, C. R. (1990) Methods Enzymol. 182,
477488
268. Finn, F. M., and Hofmann, K. (1976) in The
Proteins, 3rd ed., Vol. II (Neurath, H., and Hill,
R. L., eds), pp. 105253, Academic Press,
New York
269. Burnette, W. N. (1981) Anal. Biochem. 112,
195203
270. Karey, K. P., and Sirbasku, D. A. (1989) Anal.
Biochem. 178, 255259
271. Strickland, E. H., Billups, C., and Kay, E.
(1972) Biochemistry 11, 36573662
272. Metzler, D. E., Harris, C., Yang, I.-Y., Siano, D.,
and Thomson, J. A. (1972) Biochem. Biophys.
Res. Commun. 46, 15881597
273. Morton, R. A. (1975) Biochemical Spectroscopy,
Wiley, New York
274. Pace, C. N., Vajdos, F., Fee, L., Grimsley, G.,
and Gray, T. (1995) Protein Sci. 4, 24112423
275. Glazer, A. N., Delange, R. J., and Sigman, D.
S. (1976) Chemical Modification of Proteins:
Selected Methods and Analytical Procedures,
Elsevier, New York
References
276. Feeney, R. E., Blankenhorn, G., and Dixon, H.
B. F. (1975) Advances in Protein Chemistry, Vol.
29, Academic Press, New York (pp. 135203)
277. Acharya, A. S., and Manjula, B. N. (1987)
Biochemistry 26, 35243530
278. Rypniewski, W. R., Holden, H. M., and
Rayment, I. (1993) Biochemistry 32, 98519858
279. Jentoft, N., and Dearborn, D. G. (1979) J. Biol.
Chem. 254, 43594365
280. Hennecke, M., and Plapp, B. N. (1984) Anal.
Biochem. 136, 110118
281. Brocklehurst, K. (1979) Int. J. Biochem 10, 259
274
282. Degani, Y., and Patchornik, A. (1974)
Biochemistry 13, 111
283. Riddles, P. W., Blakeley, R. L., and Zerner, B.
(1983) Methods Enzymol. 91, 4960
284. Pande, C. S., Pelzig, M., and Glass, J. D. (1980)
Proc. Natl. Acad. Sci. U.S.A. 77, 895899
285. Fraker, P. J., and Speck, J. C., Jr. (1978)
Biochem. Biophys. Res. Commun. 80, 849857
286. Tolan, D. R., Lambert, J. M., Boileau, G.,
Fanning, T. G., Kenny, J. W., Vassos, A., and
Traut, R. R. (1980) Anal. Biochem. 103, 101109
287. Miles, E. W. (1977) Methods Enzymol. 47, 431
442
288. Kawata, Y., Sakiyama, F., Hayashi, F., and
Kyogoku, Y. (1990) Eur. J. Biochem. 187, 255262
289. Secundo, F., Carrea, G., DArrigo, P., and
Servi, S. (1996) Biochemistry 35, 96319636
290. Miura, S., Tomita, S., and Ichikawa, Y. (1991)
J. Biol. Chem. 266, 1921219216
291. Colman, R. F. (1983) Ann. Rev. Biochem. 52,
6791
292. Chowdhry, V., and Westheimer, F. H. (1979)
Ann. Rev. Biochem. 48, 293 325
293. Jelenc, P. C., Cantor, C. R., and Simon, S. R.
(1978) Proc. Natl. Acad. Sci. U.S.A. 75, 3564
3568
294. Simmonds, R. J. (1992) Chemistry of
Biomolecules, CRC Press, Boca Raton, Florida
295. Bodanszky, M., Bodanszky, A., and Trost, B.
M., eds. (1994) The Practice of Peptide Synthesis,
2nd ed., Springer-Verlag, New York
296. Lloyd-Williams, P., Albericio, F., and Giralt, E.
(1997) Chemical Approaches to the Synthesis of
Peptides and Proteins, CRC Press, Boca Raton,
Florida
297. Kent, S. B. H. (1988) Ann. Rev. Biochem. 57,
957989
298. Kaiser, E. T., Mihara, H., Laforet, G. A., Kelly,
J. W., Walters, L., Findeis, M. A., and Sasaki, T.
(1989) Science 243, 187192
299. Merrifield, B. (1986) Science 232, 341347
300. Atherton, E., Logan, C. J., and Sheppard, R. C.
(1981) J. Chem. Soc., Perkin, I., 538546
301. Atherton, E., and Sheppard, R. C. (1985) J.
Chem. Soc. Chem. Commun., 165166
301a. Miller, S. C., and Scanlan, T. S. (1998) J. Am.
Chem. Soc. 120, 26902691
302. You-shang, Z. (1983) Trends Biochem. Sci. 8,
1617
303. Kullman, W. (1987) Enzymatic Peptide
Synthesis, CRC Press, Boca Raon, Florida
304. Liu, C.-F., and Tam, J. P. (1994) J. Am. Chem.
Soc. 116, 41494153
305. Tam, J. P., Lu, Y.-A., Liu, C.-F., and Shao, J.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 12485
12489
306. Noren, C. J., Anthony-Cahill, S. J., Griffith, M.
C., and Schultz, P. G. (1989) Science 244,
182188
307. Borman, S. (1996) Chem. Eng. News 74 (Feb 12),
2935
308. Youngquist, R. S., Fuentes, G. R., Lacey, M. P.,
and Keough, T. (1995) J. Am. Chem. Soc. 117,
39003906
309. Cortese, R., ed. (1996) Combinatorial Libraries:
Synthesis, Screening and Application Potential,
Walter de Gruyter & Co., New York
310. Ellman, J., Stoddard, B., and Wells, J. (1997)
Proc. Natl. Acad. Sci. U.S.A. 94, 27792782
311. Kishchenko, G., Batliwala, H., and Makowski,
L. (1994) J. Mol. Biol. 241, 208213
312. Scott, J. K. (1992) Trends Biochem. Sci. 17, 241
245
312a. Barbas, C. F., Burton, D. B., Scott, J. K., and
Silverman, G. J. (2000) Phage Display: A
Laboratory Manual Cold Spring Harbor Press,
Cold Spring Harbor, New York
313. Marvin, D. A., (1998) Current Opinions in
Structural Biology 8, 150158
313a. Barbas, C. F., Burton, D. B., Scott, J. K., and
Silverman, G. J. (2000) Phage Display: A
Laboratory Manual Cold Spring Harbor Lab.
Press, Cold Spring Harbor, New York
314. McConnell, S. J., and Hoess, R. H. (1995) J. Mol.
Biol. 250, 460470
315. Katz, B. A. (1995) Biochemistry 34, 1542115429
316. Allen, J. B., Walberg, M. W., Edwards, M. C.,
and Elledge, S. J. (1995) Trends Biochem. Sci. 20,
511516
317. Gates, C. M., Stemmer, W. P. C., Kaptein, R.,
and Schatz, P. J. (1996) J. Mol. Biol. 255, 373386
318. Bastos, M., Maeji, N. J., and Abeles, R. H. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 67386742
319. Han, H., Wolfe, M. M., Brenner, S., and Janda,
K. D. (1995) Proc. Natl. Acad. Sci. U.S.A. 92,
64196423
320. Chu, Y.-H., Dunayevskiy, Y. M., Kirby, D. P.,
Vouros, P., and Karger, B. L. (1996) J. Am.
Chem. Soc. 118, 78277835
321. Dprez, B., Williard, X., Bourel, L., Coste, H.,
Hyafil, F., and Tartar, A. (1995) J. Am. Chem.
Soc. 117, 54055406
322. Kim, R. M., Manna, M., Hutchins, S. M.,
Griffin, P. R., Yates, N. A., Bernick, A. M., and
Chapman, K. T. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1001210017
323. Sundberg, S. A., Barrett, R. W., Pirrung, M.,
Lu, A. L., Kiangsoontra, B., and Holmes, C. P.
(1995) J. Am. Chem. Soc. 117, 1205012057
324. Baum, R. M. (1991) Chem. Eng. News 69, 2122
325. Slayter, E. M., and Slayter, H. S. (1992) Light
and Electron Microscopy, Cambridge Univ.
Press, New York
326. Bracegirdle, B. (1989) Trends Biochem. Sci. 14,
464468
327. Matthews, C. K., and van Holde, K. E. (1996)
Biochemistry, 2nd ed., Benjamin, Menlo Park,
California
328. Lichtman, J. W. (1994) Sci. Am. 271(Aug), 4045
329. Shotton, D., and White, N. (1989) Trends
Biochem. Sci. 14, 435439
330. Boyde, A. (1985) Science 230, 12701276
331. Gura, T. (1997) Science 276, 19881990
332. Bennett, B. D., Jetton, T. L., Ying, G.,
Magnuson, M. A., and Piston, D. W. (1996)
J. Biol. Chem. 271, 36473651
333. Pennisi, E. (1997) Science 275, 480481
334. Maiti, S., Shear, J. B., Williams, R. M., Zipfel,
W. R., and Webb, W. W. (1997) Science 275,
530532
335. Pool, R. (1988) Science 241, 2526
336. Betzig, E., and Chichester, R. J. (1993) Science
262, 14221425
337. Dykstra, M. J. (1992) Biological Electron
Microscopy, Plenum, New York
338. Hand, A. R. (1995) in Introduction to
Biophysical Methods for Protein and Nucleic Acid
Research (Glasel, J. A., and Deutscher, M. P.,
eds), pp. 205-260, Academic Press, San Diego
339. Nellist, P. D., McCallum, B. C., and
Rodenburg, J. M. (1995) Nature (London) 374,
630632
154 Chapter 3. Determining Structures and Analyzing Cells
402. Day, J., and McPherson, A. (1992) Protein Sci.
1, 12541268
403. Koszelak, S., Day, J., Leja, C., Cudney, R., and
McPherson, A. (1995) Biophys. J. 69, 1319
404. Service, R. F. (1999) Science 285, 13421346
405. Knblein, J., Neuefeind, T., Schneider, F.,
Bergner, A., Messerschmidt, A., Lwe, J.,
Steipe, B., and Huber, R. (1997) J. Mol. Biol.
270, 17
406. Glusker, J. P. (1984) Trends Biochem. Sci. 9,
328330
407. Guss, J. M., Merritt, E. A., Phizackerley, R. P.,
Hedman, B., Murata, M., Hodgson, K. O., and
Freeman, H. C. (1988) Science 241, 806811
408. Walter, R. L., Ealick, S. E., Friedman, A. M.,
Blake, R. C., II, Proctor, P., and Shoham, M.
(1996) J. Mol. Biol. 263, 730751
409. Budisa, N., Karnbrock, W., Steinbacher, S.,
Humm, A., Prade, L., Neuefeind, T., Moroder,
L., and Huber, R. (1997) J. Mol. Biol. 270, 616
623
410. Longhi, S., Czjzek, M., Lamzin, V., Nicolas, A.,
and Cambillau, C. (1997) J. Mol. Biol. 268,
779799
410a. Yamano, A., Heo, N.-H., and Teeter, M. M.
(1997) J. Biol. Chem. 272, 95979600
410b. Lamzin, V. S., Morris, R. J., Dauter, Z., Wilson,
K. S., and Teeter, M. M. (1999) J. Biol. Chem.
274, 2075320755
411. Rhee, S., Silva, M. M., Hyde, C. C., Rogers, P.
H., Metzler, C. M., Metzler, D. E., and Arnone,
A. (1997) J. Biol. Chem. 272, 1729317302
412. Frauenfelder, H., Petsko, G. A., and Tsernoglou,
D. (1979) Nature (London) 280, 558563
413. Arnone, A., Rogers, P. H., Hyde, C. C., Briley,
P. D., Metzler, C. M., and Metzler, D. E. (1985)
in Transaminases (Christen, P., and Metzler, D.
E., eds), pp. 138155, Wiley, New York
414. Wlodawer, A. (1982) Prog. Biophys. and Mol.
Biol. 40, 115159
415. Kossiakoff, A. A. (1985) Ann. Rev. Biochem. 54,
11951227
416. Butler, D. (1994) Nature (London) 371, 469
417. Hellemans, A. (1997) Science 277, 12141215
418. Glanz, J. (1995) Science 267, 19041906
419. Pool, R. (1988) Science 241, 295
420. Eisenberger, P., and Suckewer, S. (1996)
Science 274, 201202
421. Neutze, R., and Hajdu, J. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 56515655
422. Hajdu, J., and Johnson, L. N. (1990)
Biochemistry 29, 16691678
423. Duke, E. M. H., Wakatsuki, S., Hadfield, A.,
and Johnson, L. N. (1994) Protein Sci. 3, 1178
1196
424. Jacoby, M. (1997) Chem. Eng. News Dec 8, 5
425. Rischel, C., Rousse, A., Uschmann, I., Albouy, P.-
A., Geindre, J.-P., Audebert, P., Gauthier, J.-C.,
Frster, E., Martin, J.-L., and Antonetti, A.
(1997) Nature (London) 390, 490492
426. Rattle, H. (1995) An NMR Primer for Life
Scientists, Partnership Press, Tuckerton, NJ
427. Roberts, G. C. K., ed. (1993) NMR of
Macromolecules. A Practical Approach, IRL
Press, Oxford
428. Evans, J. N. S. (1995) Biomolecular NMR
Spectroscopy, Oxford University Press, New York
429. Olson, D. L., Peck, T. L., Webb, A. G., Magin,
R. L., and Sweedler, J. V. (1995) Science 270,
19671969
430. Wuthrich, K. (1986) NMR of Proteins and
Nucleic Acids, Wiley, New York
431. Nageswara Rao, B. D., and Kemple, M. D.,
eds. (1996) NMR as a structural tool for
macromolecules: Current status and future
directions, Plenum, New York
432. Bovey, F. A. (1969) Nuclear Magnetic Resonance
Spectroscopy, Academic Press, New York
340. Schliwa, M. (1997) Nature (London) 387, 764
341. Bubel, A., ed. (1989) Microstructure and
Function of Cells, 1st ed., Ellis Harwood, New
York
342. Penman, S. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 52515257
343. Morel, G., ed. (1995) Visualization of Nucleic
Acids, CRC Press, Boca Raton, Florida
344. Caspar, D. L. D., and DeRosier, D. J. (1982)
Science 218, 653655
345. Frank, J., ed. (1992) Electron Tomography:
Three-Dimensional Imaging with the Transmis-
sion Electron Microscope, Plenum, New York
346. Skoglund, U., and Daneholt, B. (1986) Trends
Biochem. Sci. 11, 499503
347. Lambert, O., Boisset, N., Taveau, J.-C., and
Lamy, J. N. (1994) J. Mol. Biol. 244, 640647
348. Taveau, J.-C., Boisset, N., Lamy, J., Lambert,
O., and Lamy, J. N. (1997) J. Mol. Biol. 266,
10021015
349. De Rosier, D. J. (1997) Nature (London) 386,
2627
350. Dorset, D. L. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 17911794
351. Binnig, G., and Rohrer, H. (1987) Rev. Mod.
Phys. 59, 615625
352. Binnig, G., and Rohrer, H. (1985) Sci. Am. 253,
5056
353. Wiesendanger, R. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 1274912750
354. Wickramasinghe, H. K. (1989) Sci. Am.
261(Oct), 98105
355. Hansma, P. K., Elings, V. B., Marti, O., and
Bracker, C. E. (1988) Science 242, 209216
356. Pool, R. (1990) Science 247, 634636
357. Edstrom, R. D., Yang, X., Lee, G., and Evans, D.
F. (1990) FASEB J. 4, 31443151
358. Engel, A. (1991) Ann. Rev. Biophys. Biophys.
Chem. 20, 79108
359. Amato, I. (1997) Science 276, 19821985
360. Prater, C. B., Butt, H. J., and Hansma, P. K.
(1990) Nature (London) 345, 839840
361. Hansma, H. G., and Hoh, J. H. (1994) Annu.
Rev. Biophys. Biomol. Struct. 23, 115139
362. Radmacher, M., Tillmann, R. W., Fritz, M.,
and Gaub, H. E. (1992) Science 257, 19001905
363. Karrasch, S., Hegerl, R., Hoh, J. H., Baumeister,
W., and Engel, A. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 836838
364. Mou, J., Yang, J., and Shao, Z. (1995) J. Mol.
Biol. 248, 507512
365. Yip, C. M., and Ward, M. D. (1996) Biophys. J.
71, 10711078
366. Mou, J., Sheng, S., Ho, R., and Shao, Z. (1996)
Biophys. J. 71, 22132221
367. Kuznetsov, YuG., Malkin, A. J., Land, T. A.,
DeYoreo, J. J., Barba, A. P., Konnert, J., and
McPherson, A. (1997) Biophys. J. 72, 23572364
367a. Czajkowsky, D. M., and Shao, Z. (1998) FEBS
Letters 430, 51 54
368. Noy, A., Frisbie, C. D., Rozsnyai, L. F.,
Wrighton, M. S., and Lieber, C. M. (1995) J.
Am. Chem. Soc. 117, 79437951
369. Boland, T., and Ratner, B. D. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 52975301
370. Miles, M. (1997) Science 277, 18451847
371. Hansma, H. G., Laney, D. E., Bezanilla, M.,
Sinsheimer, R. L., and Hansma, P. K. (1995)
Biophys. J. 68, 16721677
371a. Fisher, T. E., Oberhauser, A. F., Carrion-
Vazquez, M., Marszalek, P. E., and Fernandez,
J. M. (1999) Trends Biochem. Sci. 24, 379 384
372. Blundell, T. L., and Johnson, L. N. (1976)
Protein Crystallography, Academic Press, New
York
373. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Freeman, San Francisco,
California (pp. 687811)
374. McPherson, A. (1982) Preparation and Analysis
of Protein Crystals, Wiley, New York
375. Wyckoff, H. W., Hirs, C. H. W., and Timasheff,
S. N., eds. (1985) Methods in Enzymology, Vol.
114,115, Academic Press, Orlando, Florida
376. Allewell, N. M., and Trikha, J. (1995) in
Introduction to Biophysical Methods for Protein
and Nucleic Acid Research (Glasel, J. A., and
Deutscher, M. P., eds), pp. 381431, Academic
Press, San Diego
377. Verlinde, C. L. M. J., Merritt, E. A., Van Den
Akker, F., Kim, H., Feil, I., Delboni, L. F.,
Mande, S. C., Sarfaty, S., Petra, P. H., and Hol,
W. G. J. (1994) Protein Sci. 3, 16701686
378. Woolfson, N. M. (1997) An Introduction to X-ray
Crystallography, 2nd ed., Cambridge Univ.
Press, New York
379. Thomas, J. M. (1993) Nature (London) 364,
478482
380. Dounce, A. L., and Allen, P. Z. (1988) Trends
Biochem. Sci. 13, 317320
381. Bernal, J. D., and Crowfoot, D. (1934) Nature
(London) 133, 794795
382. Glusker, J. P. (1994) Protein Sci. 3, 2465-2469
383. Perutz, M. F., Muirhead, H., Cox, J. M.,
Goaman, L. G. G., Mathews, F. S., McGandy,
E. L., and Webb, L. E. (1968) Nature (London)
219, 2932
384. Perutz, M. (1992) Protein Structure: New
Approaches to Disease and Therapy, W.H.
Freeman, San Fransico and New York
385. Eisenberg, D. (1994) Protein Sci. 3, 16251628
386. Perutz, M. F. (1978) Sci. Am. 239(Dec), 92125
387. Tegze, M., and Faigel, G. (1996) Nature (London)
380, 4951
388. Fadley, C. S., and Len, P. M. (1996) Nature
(London) 380, 2728
389. Rhodes, G. (1993) Crystallography Made Crystal
Clear, Academic Press, New York
389a. Glusker, J. P., and Trueblood, K. N. (1985)
Crystal Structure Analysis A Primer, 2nd ed.,
Oxford Univ. Press, Oxford
389b. Drenth, J. (1994) Principles of Protein X-ray
Crystallography, Springler-Verlag, New York
389c. McRee, D. E. (1999) Practical Protein
Crystallography, Academic Press, San Diego,
California
390. Sweet, R. M. (1985) Methods Enzymol. 114, 1946
391. Kendrew, J. C. (1963) Science 139, 12591266
392. Kendrew, J. C., Dickerson, R. E., Strandberg,
B. E., Hart, R. G., Davies, D. R., Phillips, D. C.,
and Shore, V. C. (1960) Nature (London) 185,
422427
393. Rossman, M. G. (1994) Protein Sci. 3, 17311733
394. Blake, C. C. F., Koenig, D. F., Mair, G. A.,
North, A. C. T., Phillips, D. C., and Sarma, V.
R. (1965) Nature (London) 206, 757761
395. Laskowski, R. A., Hutchinson, E. G., Michie,
A. D., Wallace, A. C., Jones, M. L., and
Thornton, J. M. (1997) Trends Biochem. Sci. 22,
488490
396. Gerstein, M., and Levitt, M. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 1191111916
397. Marvin, D. A., and Nave, C. (1982) in
Structural Molecular Biology (Davies, D. B.,
Saenger, W., and Danyluk, S. S., eds),
pp. 344, Plenum, New York
398. Ducruix, A., and Gieg, R., eds. (1999)
Crystallization of Nucleic Acids and Proteins,
2nd ed., IRL Press, Oxford
399. McPherson, A., Koszelak, S., Axelrod, H., Day,
J., Williams, R., Robinson, L., McGrath, M.,
and Cascio, D. (1986) J. Biol. Chem. 261, 1969
1975
400. McPherson, A., Jr. (1976) J. Biol. Chem. 251,
63006303
401. Ray, W. J., Jr., and Puvathingal, J. M. (1986)
J. Biol. Chem. 261, 1154411549
References
155
433. Sanders, J. K. M., and Hunter, B. K. (1993)
Modern NMR Spectroscopy, 2nd ed., Oxford
Univ. Press, London
434. Dybowski, C., and Lichter, R. L., eds. (1987)
NMR Spectroscopy Techniques, Vol. 5, Dekker,
New York
435. Bangerter, B. W. (1995) in Introduction to
Biophysical Methods for Protein and Nucleic Acid
Research (Glasel, J. A., and Deutscher, M. P.,
eds), pp. 317-379, Academic Press, San Diego
436. Freeman, R. (1997) A Handbook of Nuclear
Magnetic Resonance, 2nd ed., Addison Wesley
Longman Limited, Essex, England
437. Emsley, J. W., ed. (1996) Encyclopedia of NMR,
John Wiley & Sons, Ltd., Sussex, England
438. Markley, J. L. (1975) Acc. Chem. Res. 8, 7080
439. Perkins, S. J. (1982) in Biological Magnetic
Resonance, Vol. 4 (Berliner, L. J., and Reuben,
J., eds), pp. 193-334, Plenum Press, New York
and London
440. Osapay, K., and Case, D. A. (1991) J. Am.
Chem. Soc. 113, 94369444
441. Szilgyi, L. (1995) Prog. Nucl. Magn. Reson.
Spectrosc. 27, Part 4, 326443
442. Williamson, M. P., Kikuchi, J., and Asakura, T.
(1995) J. Mol. Biol. 247, 541546
443. Sitkoff, D., and Case, D. A. (1997) J. Am. Chem.
Soc. 119, 1226212273
444. Gu, Z., Zambrano, R., and McDermott, A.
(1994) J. Am. Chem. Soc. 116, 63686372
445. Gu, Z., Ridenour, C. F., Bronnimann, C. E.,
Iwashita, T., and McDermott, A. (1996) J. Am.
Chem. Soc. 118, 822829
446. de Dios, A. C., Pearson, J. G., and Oldfield, E.
(1993) Science 260, 14911496
447. Sulzbach, H. M., Schleyer, P. V. R., and
Schaefer, H. F., III (1994) J. Am. Chem. Soc. 116,
39673972
448. Wishart, D. S., Sykes, B. D., and Richards, F.
M. (1992) Biochemistry 31, 16471651
449. Linderstrm-Lang, K. (1955) Chemical Society
Special Publication, No. 2, 120
450. Barfield, M., and Karplus, M. (1969) J. Am.
Chem. Soc. 91, 116
451. Karplus, M. (1963) J. Am. Chem. Soc. 85, 2870
2871
452. Bax, A. (1989) Ann. Rev. Biochem. 58, 223256
453. Wang, A. C., and Bax, A. (1996) J. Am. Chem.
Soc. 118, 24832494
454. Stonehouse, J., Adell, P., Keeler, J., and Shaka,
A. J. (1994) J. Am. Chem. Soc. 116, 60376038
455. Aguayo, J. B., Gamcsik, M. P., and Dick, J. D.
(1988) J. Biol. Chem. 263, 1955219557
456. Ackerman, J. J. H., Ewy, C. S., Kim, S.-G., and
Shalwitz, R. A. (1987) Ann. N.Y. Acad. Sci. 508,
8998
457. Breitmaier, E., and Voelter, W. (1987) Carbon-13
NMR Spectroscopy, 3rd ed., VCH Publishers,
New York
458. Beckmann, N. (1995) Carbon-13 NMR
Spectroscopy of Biological Systems, Academic
Press, New York
459. Gronenborn, A. M., Clore, G. M., Schmeissner,
U., and Wingfield, P. T. (1986) Eur. J. Biochem.
161, 3743
460. Clore, G. M., Bax, A., Driscoll, P. C.,
Wingfield, P. T., and Gronenborn, A. M. (1990)
Biochemistry 29, 81728184
461. Jue, T., Rothman, D. L., Shulman, G. I.,
Tavitian, B. A., DeFronzo, R. A., and Shulman,
R. G. (1989) Proc. Natl. Acad. Sci. U.S.A. 86,
44894491
462. Markley, J. L. (1989) Methods Enzymol. 176,
1264
463. Pelton, J. G., Torchia, D. A., Meadow, N. D.,
and Roseman, S. (1993) Protein Sci. 2, 543558
464. Rajagopal, P., Waygood, E. B., and Klevit, R.
E. (1994) Biochemistry 33, 1527115282
References
465. Solum, M. S., Altmann, K. L., Strohmeier, M.,
Berges, D. A., Zhang, Y., Facelli, J. C.,
Pugmire, R. J., and Grant, D. M. (1997)
J. Am. Chem. Soc. 119, 98049809
466. Verkade, J. G., and Quin, L. D., eds. (1987)
Phosphorus-31 NMR Spectroscopy in
Stereochemical Analysis, VCH Publishers,
Deerfield Beach, Florida
467. Ugurbil, K., Kingsley-Hickman, P. B., Sako, E.
Y., Zimmer, S., Mohanakrishnan, P., Robitaille,
P. M. L., Thoma, W. J., Johnson, A., Foker, J. E.,
From, A. H. L. (1987) Ann. N.Y. Acad. Sci. 508,
265286
468. Feeney, J., McCormick, J. E., Bauer, C. J.,
Birdsall, B., Moody, C. M., Starkmann, B. A.,
Young, D. W., Francis, P., Havlin, R. H.,
Arnold, W. D., and Oldfield, E. (1996) J. Am.
Chem. Soc. 118, 87008706
469. Lau, E. Y., and Gerig, J. T. (1997) Biophys. J. 73,
15791592
470. Jarema, M. A. C., Lu, P., and Miller, J. H. (1981)
Proc. Natl. Acad. Sci. U.S.A. 78, 27072711
471. Slebe, J. C., and Martinez-Carrion, M. (1990)
J. Biol. Chem. 253, 20932097
472. Briley, P. A., Eisenthal, R., Harrison, R., and
Smith, G. D. (1990) Biochem. J. 163, 325331
473. Roberts, J. K. M., Wade-Jardetzky, N., and
Jardetzky, O. (1981) Biochemistry 20, 5389
5394
474. Deutsch, C. J., and Taylor, J. S. (1987) Ann.
N.Y. Acad. Sci. 508, 33
475. Gutowsky, H. S., McCall, D. W., McGarvey, B.
R., and Meyer, L. H. (1952) J. Am. Chem. Soc.
74, 48094817
476. Chang, Y. C., and Graves, D. J. (1985) J. Biol.
Chem. 260, 27092714
477. Scott, R. D., Chang, Y.-C., Graves, D. J., and
Metzler, D. E. (1985) Biochemistry 24, 76687681
478. Saunders, M., Jimnez-Vzquez, H. A., Cross,
R. J., Mroczkowski, S., Freedberg, D. I., and
Anet, F. A. L. (1994) Nature (London) 367, 256
257
479. Adebodun, F., and Jordan, F. (1988) J. Am.
Chem. Soc. 110, 309310
480. Springer, C. S., Jr. (1987) Ann. Rev. Biophys.
Biophys. Chem. 16, 375399
481. Wittenberg, B. A., and Gupta, R. K. (1985)
J. Biol. Chem. 260, 20312034
482. Jelicks, L. A., and Gupta, R. K. (1989) J. Biol.
Chem. 264, 1523015235
483. Price, W. S., Ge, N.-H., Hong, L.-Z., and
Hwang, L.-P. (1993) J. Am. Chem. Soc. 115,
10951105
484. Wang, S. M., and Gilpin, R. K. (1983) Anal.
Chem. 55, 493497
485. Markham, G. D. (1986) J. Biol. Chem. 261,
15071509
486. Denisov, V. P., and Halle, B. (1995) J. Mol. Biol.
245, 682687
487. Mullen, G. P., Dunlap, R. B., and Odom, J. D.
(1986) Biochemistry 25, 56255632
488. Aldhous, P. (1991) Nature (London) 353, 689
489. Becker, E. D., and Farrar, T. C. (1972) Science
178, 361368
490. Bax, A., and Lerner, L. (1986) Science 232,
960967
491. Yamazaki, T., Muhandiram, R., and Kay, L. E.
(1994) J. Am. Chem. Soc. 116, 82668278
492. Farrar, T. C., and Becker, E. D. (1971) Pulse and
Fourier Transform NMR, Academic Press, New
York
493. Wthrich, K. (1989) Science 243, 4550
494. van de Ven, F. J. M., and Hilbers, C. W. (1986)
J. Mol. Biol. 192, p. 419-441
495. Baniak, E. L., Rivier, J. E., Struthers, R. S.,
Hagler, A. T., and Gierasch, L. M. (1987)
Biochemistry 26, 26422656
496. Wthrich, K. (1990) J. Biol. Chem. 265, 22059
22062
497. Bax, A., and Grzesiek, S. (1993) in NMR of
Proteins (Clore, G. M., and Gronenborn, A. M.,
eds), pp. 3352, CRC Press, Boca Raton,
Florida
498. Clore, G. M., and Gronenborn, A. M. (1994)
Protein Sci. 3, 372390
499. Hoitnik, C. W. G., Driscoll, P. C., Hill, H. A. O.,
and Canters, G. W. (1994) Biochemistry 33,
35603571
500. Yamazaki, T., Tochio, H., Furui, J., Aimoto, S.,
and Kyogoku, Y. (1997) J. Am. Chem. Soc. 119,
872880
501. Yamazaki, T., Lee, W., Arrowsmith, C. H.,
Muhandiram, D. R., and Kay, L. E. (1994)
J. Am. Chem. Soc. 116, 1165511666
502. Hu, J.-S., and Bax, A. (1997) J. Am. Chem. Soc.
119, 63606368
503. Song, S., Velde, D. V., Gunn, C. W., and
Himes, R. H. (1994) Biochemistry 33, 693698
504. Barsukov, I. L., Lian, L.-Y., Ellis, J., Sze, K.-H.,
Shaw, W. V., and Roberts, G. C. K. (1996) J.
Mol. Biol. 262, 543558
505. Wu, W.-J., Anderson, V. E., Raleigh, D. P., and
Tonge, P. J. (1997) Biochemistry 36, 22112220
506. Metzler, W. J., Hare, D. R., and Pardi, A.
(1989) Biochemistry 28, 70457052
507. McDowell, L. M., Lee, M., McKay, R. A.,
Anderson, K. S., and Schaefer, J. (1996)
Biochemistry 35, 33283334
508. Pflugrath, J. W., Wiegand, G., Huber, R., and
Vrtesy, L. (1986) J. Mol. Biol. 189, 383386
509. Kline, A. D., Braun, W., and Wthrich, K.
(1986) J. Mol. Biol. 189, 377382
510. McDonald, C. C., and Phillips, W. D. (1969)
J. Am. Chem. Soc. 91, 15131521
511. Nietlispach, D., Clowes, R. T., Broadhurst, R.
W., Ito, Y., Keeler, J., Kelly, M., Ashurst, J.,
Oschkinat, H., Domaille, P. J., and Laue, E. D.
(1996) J. Am. Chem. Soc. 118, 407415
512. Rosen, M. K., Gardner, K. H., Willis, R. C.,
Parris, W. E., Pawson, T., and Kay, L. E. (1996)
J. Mol. Biol. 263, 627636
513. Venters, R. A., Farmer, B. T., II, Fierke, C. A.,
and Spicer, L. D. (1996) J. Mol. Biol. 264, 1101
1116
514. LeMaster, D. M., and Richards, F. M. (1988)
Biochemistry 27, 142150
515. Bax, A., Kay, L. E., Sparks, S. W., and Torchia,
D. A. (1989) J. Am. Chem. Soc. 111, 408409
516. Torchia, D. A., Sparks, S. W., and Bax, A.
(1989) Biochemistry 28, 55095524
517. Lowry, D. F., Redfield, A. G., McIntosh, L. P.,
and Dahlquist, F. W. (1988) J. Am. Chem. Soc.
110, 68856886
518. Stockman, B. J., Reily, M. D., Westler, W. M.,
Ulrich, E. L., and Markley, J. L. (1989)
Biochemistry 28, 230236
519. Qian, H., Mayo, S. L., and Morton, A. (1994)
Biochemistry 33, 81678171
519a. Riek, R., Pervushin, K., and Wthrich, K.
(2000) Trends Biochem. Sci. 25, 462468
520. Gnter, P., Braun, W., Billeter, M., and Wthrich,
K. (1989) J. Am. Chem. Soc. 111, 39974004
521. Oh, B.-H., Westler, W. M., and Markley, J. L.
(1989) J. Am. Chem. Soc. 111, 30833085
522. Cross, T. A., and Opella, S. J. (1983) J. Am. Chem.
Soc. 105, 306308
523. Rothgeb, T. M., and Oldfield, E. (1981) J. Biol.
Chem. 256, 14321446
524. Buck, M., Schwalbe, H., and Dobson, C. M.
(1995) Biochemistry 34, 1321913232
525. Nicholson, L. K., Kay, L. E., Baldisseri, D. M.,
Arango, J., Young, P. E., Bax, A., and Torchia, D.
A. (1992) Biochemistry 31, 52535263
526. Gardner, K. H., and Kay, L. E. (1997) J. Am.
Chem. Soc. 119, 75997600
527. Lenkinski, R. E., Dallas, J. L., and Glickson, J.
D. (1979) J. Am. Chem. Soc. 101, 30713077
156 Chapter 3. Determining Structures and Analyzing Cells
528. Carver, J. A., and Bradbury, J. H. (1984)
Biochemistry 23, 48904905
529. Bothelho, L. H., and Gurd, F. R. N. (1978)
Biochemistry 17, 51885196
530. Bothelho, L. H., Friend, S. H., Matthew, J. B.,
Lehman, L. D., Hanania, G. I. H., and Gurd, F.
R. N. (1978) Biochemistry 17, 51975205
531. Morino, Y., Yamasaki, M., Tanase, S.,
Nagashima, F., Akasaka, K., Imoto, T., and
Miyazawa, T. (1984) J. Biol. Chem. 259, 3877
3882
532. Cocco, M. J., Kao, Y.-H., Phillips, A. T., and
Lecomte, J. T. J. (1992) Biochemistry 31, 6481
6491
533. Bachovin, W. W., Wong, W. Y. L., Farr-Jones,
S., Shenvi, A. B., and Kettner, C. A. (1988)
Biochemistry 27, 76897697
534. Mollova, E. T., Metzler, D. E., Kintanar, A.,
Kagamiyama, H., Hayashi, H., Hirotsu, K., and
Miyahara, I. (1997) Biochemistry 36, 615625
535. Guron, M., Plateau, P., and Decorps, M.
(1991) Progr. in NMR Spectr. 23, 135209
536. Plateau, P., and Guron, M. (1982) J. Am.
Chem. Soc. 104, 73107311
537. Sklenar, V., and Bax, A. (1987) J. of Magnetic
Resonance 74, 469479
538. Bullitt, E., and Makowski, L. (1995) Nature
(London) 373, 164167
539. Griffin, J. H., Cohen, J. S., and Schechter, A. N.
(1973) Biochemistry 12, 20962099
540. Tong, H., and Davis, L. (1995) Biochemistry 34,
33623367
541. Gunnarsson, G., Wennerstrm, H., Egan, W.,
and Forsn, S. (1976) Chem. Phys. Lett. 38,
9699
542. McDermott, A., and Ridenour, C. F. (1996) in
Encyclopedia of NMR (Emsley, J. W., ed), pp.
38203824, J. Wiley & Sons, Ltd., Sussex, UK
543. Zhao, Q., Abeygunawardana, C., Gittis, A. G.,
and Mildvan, A. S. (1997) Biochemistry 36,
1461614626
544. Perrin, C. L., and Nielson, J. B. (1997) J. Am.
Chem. Soc. 119, 1273412741
545. Tjandra, N., and Bax, A. (1997) J. Am. Chem.
Soc. 119, 80768082
546. Kintanar, A., Metzler, C. M., Metzler, D. E.,
and Scott, R. D. (1991) J. Biol. Chem. 266,
1722217229
547. Metzler, C. M., Metzler, D. E., Kintanar, A.,
Scott, R. D., and Marceau, M. (1991) Biochem.
Biophys. Res. Commun. 178, 385392
548. Firsov, L. M., Neustroev, K. N., Aleshin, A. E.,
Metzler, C. M., Metzler, D. E., Scott, R. D.,
Stoffer, B., Christensen, T., and Svensson, B.
(1994) Eur. J. Biochem. 223, 293302
549. Metzler, D. E., Metzler, C. M., Mollova, E. T.,
Scott, R. D., Tanase, S., Kogo, K., Higaki, T.,
and Morino, Y. (1994) J. Biol. Chem. 269,
2801728026
550. Metzler, D. E., Metzler, C. M., Scott, R. D.,
Mollova, E. T., Kagamiyama, H., Yano, T.,
Kuramitsu, S., Hayashi, H., Hirotsu, K., and
Miyahara, I. (1994) J. Biol. Chem. 269, 28027
28033
551. Metzler, D. E. (1997) Methods Enzymol. 280,
3040
552. Englander, S. W., Mayne, L., Bai, Y., and
Sosnick, T. R. (1997) Protein Sci. 6, 11011109
553. Kossiakoff, A. A. (1982) Nature (London) 296,
713721
554. Rohl, C. A., and Baldwin, R. L. (1994)
Biochemistry 33, 77607767
555. Gemmecker, G., Jahnke, W., and Kessler, H.
(1993) J. Am. Chem. Soc. 115, 1162011621
556. Zhang, Y.-Z., Paterson, Y., and Roder, H.
(1995) Protein Sci. 4, 804814
557. Zhang, Z., and Smith, D. L. (1993) Protein Sci.
2, 522531
558. Wagner, D. S., Melton, L. G., Yan, Y., Erickson,
B. W., and Anderegg, R. J. (1994) Protein Sci. 3,
13051314
559. Zhang, Z., Post, C. B., and Smith, D. L. (1996)
Biochemistry 35, 779791
560. Zappacosta, F., Pessi, A., Bianchi, E.,
Venturini, S., Sollazzo, M., Tramontano, A.,
Marino, G., and Pucci, P. (1996) Protein Sci. 5,
802813
561. Kim, K.-S., Fuchs, J. A., and Woodward, C. K.
(1993) Biochemistry 32, 96009608
562. Eriksson, M. A. L., Hrd, T., and Nilsson, L.
(1995) Biophys. J. 69, 329339
563. Spyracopoulos, L., and ONeil, J. D. J. (1994) J.
Am. Chem. Soc. 116, 13951402
564. Markley, J. L. (1975) Biochemistry 14, 35463554
565. Schmidt-Rohr, K., and Spiess, H. W. (1994)
Multidimensional Solid-State NMR and
Polymers, Academic Press, San Diego,
California
566. Schmidt-Rohr, K. (1996) J. Am. Chem. Soc. 118,
76017603
567. Wu, C. H., Ramamoorthy, A., Gierasch, L. M.,
and Opella, S. J. (1995) J. Am. Chem. Soc. 117,
61486149
568. Meyer, E. F. (1997) Protein Sci. 6, 15911597
569. Stampf, D. R., Felder, C. E., and Sussman, J. L.
(1995) Nature (London) 374, 572573
570. Holm, L., and Sander, C. (1995) Trends
Biochem. Sci. 20, 478480
571. Murzin, A. G., Brenner, S. E., Hubbard, T., and
Chothia, C. (1995) J. Mol. Biol. 247, 536540
572. Walsh, L., Newell, M., Ruczaj, H., Reid, T.,
and Desmond, P., eds. (1997) Trends Guide to
the Internet, Elsevier, Cambridge, UK
573. Hogue, C. W. V., Ohkawa, H., and Bryant, S.
H. (1996) Trends Biochem. Sci. 21, 226229
574. Wilkins, M. R., Hochstrasser, D. F., Sanchez,
J.-C., Bairoch, A., and Appel, R. D. (1996)
Trends Biochem. Sci. 21, 496497
575. Peitsch, M. C., Wells, T. N. C., Stampf, D. R.,
and Sussman, J. L. (1995) Trends Biochem. Sci.
20, 8284
576. Sayle, R. A., and Milner-White, E. J. (1995)
Trends Biochem. Sci. 20, 374376
576a. Bernstein, H. J. (2000) Trends Biochem. Sci. 25,
453454
577. Sokolik, C. W. (1995) Trends Biochem. Sci. 20,
122124
578. Hunt, T. (1996) Trends Biochem. Sci. 21, 74
579. Service, R. F. (2000) Science 287, 1954 1956
580. Laskowski, R. A., Hutchinson E. G., Michie,
A. D., Wallace, A. C., Jones, M. L., and
Thornton, J. M. (1997) Trends Biochem. Sci 22,
488490
581. Cohn, E. J., and Edsall, J. T. (1943) Proteins,
Amino Acids and Peptides as Ions and Dipolar
Ions, Van Nostrand-Reinhold, Princeton, New
Jersey (pp. 9093)
582. Abbott, A. (2000) Nature (London) 408, 130132
583. Simmerling, C., Lee, M. R., Ortiz, A. R.,
Kolinski, A., Skolnick, J., and Kollman, P. A.
(2000) J. Am. Chem. Soc. 122, 83928402
References
157
1. a. From the expression for the dissociation constant
of an acid, HA (Eq. 3-1), derive the logarithmic
form pH pK
a
= log[A

] / [HA] = log
10
/(1)
(Eq. 3-11), where is the fraction of the acid
in the ionized form.
b. The apparent dissociation constant K
a
for the
H
2
PO
4

ion at 25C and 0.5 M total phosphate


concentration is 1.380 x 10
7
M. What will be the
pH of a solution 0.025 M in KH
2
PO
4
and 0.25 M
in Na
2
HPO
4
? This is a National Bureau of
Standards buffer (see Bates
7
).
c. Suppose that you wanted to prepare a buffer
of pH = 7.00 at 25C from anhydrous KH
2
PO
4
(M
r
= 136.09) and Na
2
HPO
4
(M
r
= 141.98). If you
placed 3.40 g of KH
2
PO
4
in a 1 liter volumetric
flask, how much anhydrous Na
2
HPO
4
would
you have to weigh and add before making to
volume to obtain the desired pH? If you wanted
to have the correct pH to 0.01 unit, how
accurately would you have to weigh your salts?
NOTE: It is quicker to prepare a buffer of precise
pH this way than it is to titrate a portion of
buffer acid to the desired pH with sodium
hydroxide.
2. The apparent pK
a
for 0.1 M formic acid is 3.70 at
25C.
a. Concentrated HCl was added to a liter of 0.1 M
sodium formate until a pH of 1.9 was attained.
Calculate the concentration of formate ion and
that of unionized formic acid.
b. Calculate the hydrogen ion concentration.
c. How many equivalents of HCl had to be added
in part a to bring the pH to 1.9?
3. Exactly 0.01-mol portions of glycine were placed in
several 100-ml volumetric flasks. The following
exact amounts of HCl or NaOH were added to the
flasks, the solutions were made to volume with
water, mixed, and the pH measured. Calculate the
pK
a
values for the carboxyl and amino groups from
the following, making as many independent calcu-
lations of each pKa as the data permit. At low pH
values you must correct for the free hydrogen ion con-
centration (see question 2c).
581
Study Questions
Flask No. Mol HCl Mol NaOH pH
1 0.010 1.71
2 0.009 1.85
3 0.006 2.25
4 0.002 2.94
5 0.002 9.00
6 0.004 9.37
7 0.005 9.60
4. Using the pK
a
values from problem 3, construct the
theoretical titration curve showing the equivalents
of H
+
or OH

reacting with 1 mol of glycine as a


function of pH. Note that the shape of this curve is
independent of the pK
a
. Sketch similar curves for
glutamic acid (pK
a
s equal 2.19, 4.25, and 9.67),
histidine (pK
a
s equal 1.82, 6.00, and 9.17) and
lysine (pK
a
s equal 2.18, 8.95, and 10.53).
Compare your plot for glycine with a plot of 1 M
acid or base added to 0.01 mol of glycine in 100 ml
of water. You may also compare your curves with
those for glycine published in other textbooks.
5. Make a table of characteristic pK
a
values for acidic
and basic groups in proteins. Which of these groups
contribute most significantly to the titration curves
of proteins?
6. If placed in water and adjusted to a pH of 7, will
the following migrate toward the anode or the
cathode if placed in an electrical field? (a) Aspartic
acid, (b) alanine, (c) tyrosine, (d) lysine, (e) argin-
ine, and (f) glutamine
7. The tripeptide L-Ala L-His L-Gln had the follow-
ing pK
a
values: 3.0 (-COOH), 9.1 (-NH
3

), and
6.7 (imidazolium).
a. What is the isoelectric pH (pI) of the peptide,
i.e., the pH at which it will carry no net charge?
Hint, the pI for amino acids is usually given
approximately as the arithmetic mean of two pK
a
values.
581
b. Draw the structures of the ionic forms of the
peptide that occur at pH 5 and at pH 9. At each
pH compute the fraction of the peptide in each
ionic form.
158 Chapter 3. Determining Structures and Analyzing Cells
8. a. Write the structure for glycyl-L-tryptophanyl-L-
prolyl-L-seryl-L-lysine.
b. What amino acids could be isolated from it
following acid hydrolysis?
c. Following alkaline hydrolysis?
d. After nitrous acid treatment followed by acid
hydrolysis
e. In an electrolytic cell at pH 7.0 would the peptide
migrate toward the cathode or toward the anode?
What is the approximate isoelectric point of the
peptide?
f. If a solution of this peptide were adjusted to pH
7, and then titrated with sodium hydroxide in
the presence of 10% formaldehyde, how many
equivalents of base would be required per mole
of peptide to raise the pH to 10?
9. A peptide was shown to contain only L-lysine and
L-methionine. Titration of the peptide showed 3
free amino groups for each free carboxyl group
present, and each amino group liberated 1 mole of
N
2
when the peptide was treated with HNO
2
in the
Van Slyke apparatus. When the deaminated pep-
tide was hydrolyzed completely in acid and the
hydrolyzate again treated with HNO
2
, the same
amount of N
2
is liberated as that derived from the
intact peptide. A sample of the original peptide
was treated with excess dinitrofluorobenzene to
give a dinitrophenyl (DNP) peptide, which was
shown spectrophotometrically to contain three
DNP groups per free carboxyl group. When this
DNP-peptide was completely hydrolyzed, the
following products were found: a colorless com-
pound containing S (A
1
) and a yellow compound
containing S (A
3
). Partial hydrolysis of the DNP-
peptide yields A
1
, A
2
, A
3
, plus 4 additional yellow
compounds, B
1
, B
2
, B
3
, and B
4
. On complete hydro-
lysis, B
1
yields A
1
, A
2
, and A
3
; B
2
yields A
1
and A
2
;
B
3
yields A
1
and A
3
; and B
4
yields A
3
only.
What is the most probable structure of the original
peptide?
10. (a) Explain two advantages of the isotope dilution
method of analysis. (b) From the following data,
calculate the amount of cyclic AMP (cAMP) present
per ml of human gluteus maximus muscle cells.
Cells were treated with
32
P-enriched cAMP, S.A. =
50 Ci / mol for 0.2 h (all cAMP was taken up by
cells), cells were homogenized, and the soluble
cAMP was isolated and purified. The specific
activity of the isolated cAMP = 10 Ci / mol. The
total amount of cAMP added was 1.0 x 10
7
mol per
ml of cells.
11. The figure in Box 3-C shows a high-resolution
separation of the soluble proteins of E. coli. The
investigator labeled the proteins with
14
C-contain-
ing amino acids.
a. How would you carry out the labeling
experiment?
b. What other isotope(s) could be used to label
proteins? What chemical form(s) would you
use? What limitations might there be?
c. What soluble components of an E. coli cell soni-
cate might interfere with the two-dimensional
separation, and how could they be removed?
d. What technique(s) other than radioactive
labeling could be used for locating proteins?
e. If all the soluble proteins of E. coli were detected,
about how many separate proteins would you
expect to see?
f. Indicate two or more properties of the resolution
technique which are most significant in making
it applicable to a system containing a very large
number of proteins.
12.
35
S is a beta emitter, with no gamma or other type
of radiation. It has the following properties:
t
1/2
= 86.7 days,
max
= 0.168 MeV.
a. Write the equation for the radiochemical decom-
position of
35
S.
b. Discuss the advantages and limitation of the use
of
35
S as an isotopic tracer.
Study Questions
160 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
A. Structures and Properties of Simple Sugars
1. The Variety of Monosaccharides
Ways of indicating configuration
Natural derivatives of sugars
2. Conformations of Five- and Six-Membered Ring Forms
3. Characteristic Reactions of Monosaccharides
B. Glycosides, Oligosaccharides, Glycosylamines, and
Glycation
C. Polysaccharides (Glycans)
1. Conformations of Polysaccharide Chains
2. The Glucans
3. Other Homopolysaccharides
4. Heteropolysaccharides of the Animal Body
5. Plant Heteropolysaccharides
6. Polysaccharides of Bacterial Surfaces
D. Glycoproteins and Proteoglycans
1. O-Linked Glycosyl Groups
2. Asparagine-Linked Oligosaccharides
3. Glycoproteins in Biological Recognition
Lectins and other carbohydrate-binding proteins
Carbohydrate-binding sites
Binding of viruses and bacteria to cells
Aggregation and adherence of cells
Growth and differentiation
Recognition and adhesion by leukocytes
E. Some Special Methods
1. Release of Oligosaccharides from Glycoproteins
2. Hydrolysis
3. Methylation
4. Periodate Oxidation (Smith Degradation)
5. Nuclear Magnetic Resonance
References
Study Questions
161
162
163
164
166
167
167
169
170
172
175
175
177
179
180
181
182
186
186
186
186
187
187
188
188
188
189
189
190
190
192
196
Boxes
Box 4-A Cyclodextrins
Box 4-B Silicon: An Essential Trace Element
Box 4-C The Blood Group Determinants
Box 4-D Antifreeze and Ice-Nucleation Proteins
Tables
Table 4-1 Some of the Many Polysaccharides
Found in Nature
170
171
178
184
191
Contents
Each cotton fiber is a single cell seed hair, ~ 30 mm in length. The dry fiber is
~ 95% cellulose, which constitutes the secondary cell wall (See Fig. 20-4,D) and
is also present in the primary cell wall. The fibers, which are ~ 30 nm in length,
consist of many parallel chains (Fig. 4-5) ~ 5 m in length, each containing ~ 10,000
glucose residues. Van der Waals forces, together with one hydrogen bond per
glucose, contribute to the stability of the tightly packed fiber. Cotton is one of
the major agricultural crops, ~ 87 million bales, each ~220 kg, being produced
annually world-wide. Photo and information from A. D. French and M. A.
Godshall, Southern Regional Research Center, USDA, New Orleans, LA.
161
We are all familiar with sugars, important compo-
nents of our diet which are present in fruits, honey,
table sugar, and syrups. Within our bodies the simple
sugar D-glucose is an essential source of energy. It is
present in blood at a relatively constant concentration
of 5.5 mM and is carried to all tissues. A polysaccha-
ride called glycogen, a polymeric form of glucose,
provides a reserve of readily available energy within
our cells. Starches and other polysaccharides store
energy within plants. Polysaccharides also have major
structural functions in nature. Cellulose, another
polymer of glucose, forms the fibers of cotton, plant
cell walls, and wood. Both the tough exoskeletons of
anthropods and the cell walls of fungi depend for their
strength on the nitrogen-containing polysaccharide
chitin. Polysaccharides that carry many negative
charges, such as hyaluronan, form a protective layer
between animal cells, while pectins play a similar role
in plants.
A third function of sugar residues (glycosyl groups)
is in biological recognition and communication. The
outer surfaces of cells are nearly covered by covalently
attached oligosaccharides, small polymeric arrays of
sugar rings. Some are attached to side chain groups of
proteins and others to lipids to form glycoproteins
and glycolipids, respectively. Because of the variety
of different sugars, the various ways in which they can
be linked, and their ability to form oriented hydrogen
bonds, these oligosaccharides provide much of the
chemical code for identifying cells. This coding enables
cells to attach to each other in correct ways during
development of a multicelled organism. It helps to
activate our immune system to attack parasites and
also helps bacteria to attack us! We are only beginning
to understand the numerous critical functions of the
glycosyl groups of cell surfaces.
A. Structures and Properties of Simple Sugars
The simple sugars, or monosaccharides, are poly-
hydroxyaldehydes (aldoses) or polyhydroxyketones
(ketoses).
15
All have the composition (CH
2
O)
n
, hence
the family name carbohydrate. A typical sugar, and
the one with the widest distribution in nature, is glucose.
The carbonyl group of this and other sugars is highly
reactive and a characteristic reaction is addition of
electron-rich groups such as OH. If a sugar chain is
long enough (46 carbon atoms) one of the hydroxyl
groups of the same molecule can add to the carbonyl
group to form a cyclic hemiacetal or ring form, which
reaches an equilibrium with the free aldehyde or ketone
form (Eq. 4-1). The six-membered rings formed in this
way (pyranose rings) are especially stable, but five-
membered furanose rings also exist in many carbo-
hydrates.
Sugars, Polysaccharides, and Glycoproteins
4
C
C H
C
OH
HO H
C OH H
C H
CH
2
OH
OH
H O
C
C HO
C
H
H OH
C H HO
C HO
CH
2
OH
Sugars are named as D or L
according to the configuration
about this carbon atom, one
removed from the terminal
position
D-Glucose (Glc)
showing
numbering
of atoms
1
2
3
4
5
6
L-Glucose
H
H O
D-Mannose (Man)
has the opposite
configuration at C-2
D-Galactose (Gal)
has the opposite
configuration at C-4
162 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
(4-1)
C
C
C
C
C
CH
2
OH
H O
H OH
H OH
H OH
H HO
O
OH
OH
HO
CH
2
OH
H HO
O
OH
H
HO
CH
2
OH
OH HO

DGlucose
(free aldehyde form)

OH
C
R
Covalent hydrate
of RCHO
H OH
CH
2
OH
C O
C
C
C
CH
2
OH
O H
H O H
H
H O H
D-Fructose (Fru), a ketose that is a close
structural and metabolic relative of D-glucose.
It occurs in honey and fruit juices in free form,
in the disaccharide sucrose (table sugar) as a
5-membered furanose ring, and in other
oligosaccharides and polysaccharides.
Figure 4-1 Natural abundance
13
C-NMR spectrum of honey
showing content of and pyranose ring forms of glucose
and both pyranose and furanose ring forms of fructose. Open
chain fructose, with a peak at 214 ppm, was present in a trace
amount. From Prince et al.
6
The ring forms of the sugars are the monomers
used by cells to form polysaccharides. Indeed, it is the
natural tendency of 5- and 6-carbon sugars (pentoses
and hexoses) to cyclize that permits formations of
stable sugar polymers from the reactive and unstable
monomers. When a sugar cyclizes a new chiral center
is formed at the anomeric carbon atom, the atom
that was present in the original carbonyl group. The
two configurations about this carbon atom are desig-
nated and as indicated in Eq. 4-1. In an equilibrium
mixture, ring forms of most sugars predominate over open
chains.
613
Thus, at 25C in water glucose reaches an
Because of their many polar hydroxyl groups,
most sugars are very soluble in water. However,
hydrogen bonds between molecules stabilize sugar
crystals making them insoluble in nonpolar solvents.
Intermolecular hydrogen bonds between chains of
sugar rings in cellulose account for much of the
strength and insolubility of these polysaccharides.
1. The Variety of Monosaccharides
Sugars contain several chiral centers and the
various diastereomers are given different names. The
commonly occurring sugars D-glucose, D-mannose,
and D-galactose are just three of the 16 diastereomeric
aldohexoses. The Fischer projection formulas for the
entire family of eight D-aldoses with 36 carbon atoms
are given in Fig. 4-2. Several of these occur only rarely
in nature.
Monosaccharides are classified as D or L according
to the configuration at the chiral center farthest from
the carbonyl group. If the OH group attached to this
carbon atom lies to the right when the sugar is oriented
according to the Fischer convention, the sugar belongs
to the D family. The simplest of all the chiral sugars is
glyceraldehyde. The family of aldoses in Fig. 4-2 can
be thought of as derived from D-glyceraldehyde by
upward extension of the chain. Besides D-glucose,
D-mannose, and D-galactose, the most abundant natu-
rally occurring sugar shown in Fig. 4-2 is D-ribose, a
major component of RNA. Another abundant aldose
is D-xylose, a constituent of the polysaccharides of
wood.
105 100 95 90 85 ppm 75 70 65 60
2 4 5 3 1 6
4 5 3 16 2
2 4 5 3 1 6
6 4 5 2 3 1
1 6 4 5 2 3
-D-Glucopyranose
-D-Glucopyranose
-D-Fructofuranose
-D-Fructopyranose
-D-Fructofuranose
Pyranose
ring forms
(hemiacetal
forms)
equilibriumwith ~0.001% free aldehyde, 0.004% cova-
lent hydrate of the aldehyde (see also Eq. 13-1), 39%
-pyranose form, 61% -pyranose form, and 0.15%
each of the much less stable and furanose forms.
9,12
The ketose sugar fructose in solution exists as ~73%
-pyranose, 22% -furanose, 5% -furanose, and 0.5%
open chain form (Fig. 4-1).
6,10,13
Although polysaccha-
rides are composed almost exclusively of sugar residues
in ring forms, the open chain forms are sometimes
metabolic intermediates.
163
Figure 4-2 Formulas for the D-aldoses. Prefixes derived from the names of these aldoses are used in describing various other
sugars including ketoses. The prefixes include erythro, threo, arabino, ribo, galacto, manno, etc., D-Fructose can be described in this
manner as D-arabino-hexulose. The prefixes refer to the configurations of a series of consecutive but not necessarily contiguous
chiral centers.
2
Thus, 3-arabino-hexulose is an isomer of fructose with the carbonyl group at the 3 position. The prefix deoxy,
which means lacking oxygen, is often used to designate a modified sugar in which an OH has been replaced by H, e.g.,
2-deoxyribose.
CHO
C H OH
CH
2
OH
CHO
C HO H
C
CHO
C H OH
C H OH
CH
2
OH
H OH
CH
2
OH
CHO
C HO H
C HO H
C
CHO
C H OH
C HO H
C OH H
CH
2
OH
H OH
CH
2
OH
CHO
C HO H
C H OH
C
CHO
C H OH
C H OH
C OH H
CH
2
OH
H OH
CH
2
OH
CHO
C H OH
C H OH
C OH H
C H OH
CH
2
OH
CHO
C HO H
C H OH
C OH H
C H OH
CH
2
OH
CHO
C H OH
C HO H
C OH H
C H OH
CH
2
OH
CHO
C HO H
C HO H
C OH H
C H OH
CH
2
OH
CHO
C H OH
C H OH
C H HO
C H OH
CH
2
OH
CHO
C HO H
C H OH
C H HO
C H OH
CH
2
OH
CHO
C H OH
C HO H
C H HO
C H OH
CH
2
OH
CHO
C HO H
C HO H
C H HO
C H OH
CH
2
OH
Galactose (Gal)
Arabinose (Ara)
Erythrose
Glyceraldehyde
Lyxose (Lyx)
Talose (Tal)
Threose
Mannose (Man) Glucose (Glc)
Ribose (Rib)
Gulose (Gul) Idose (Ido) Allose (All) Altrose (Alt)
Xylose (Xyl)
A. Structures and Properties of Simple Sugars
For each D sugar there is an L sugar which is the
complete mirror image or enantiomer of the D form.
Although L-arabinose occurs widely in plants, and
some derivatives of L sugars are present in glycopro-
teins, most naturally occurring sugars have the D con-
figuration. A pair of sugars, such as glucose and galac-
tose, differing in configuration at only one of the chiral
centers are known as epimers. The D-ketose sugars
with the carbonyl group in the 2 position (Fig. 4-3) are
also abundant in nature, often occurring in the form of
phosphate esters as intermediates in metabolism.
Ways of indicating configuration. The Fischer
projection formulas used in Figs. 4-2 and 4-3 are con-
venient in relating the sugar structures by their indi-
vidual carbon configurations to each other, but they
give an unrealistic three-dimensional picture. Accord-
ing to the Fischer convention each carbon atom must
be viewed with both vertical bonds projecting behind
the atom viewed. In fact, the molecule cannot assume
such a conformation because the chain folds back on
itself, bringing many atoms into collision.
Ring forms of sugars are also often drawn according
to the Fischer convention; making use of elongated
bent lines to represent ordinary simple bonds:
In this representation the hydroxyl at the anomeric
C OH H
C OH H
C OH H
C OH H
CH
2

O
C OH H
C OH H
C OH HO
C OH H

O
C
C H
C OH H
C H

O
C
CH
2
OH
H
CH
2
OH HO
HO
CH
2
OH
-D-Ribopyranose -D-Glucopyranose -D-Fructofuranose
164 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
carbon atom is always on the right side of the molecules
for forms in the D series of sugars and at the left side
for the forms. For the L sugars the opposite is true.
For example, since -D-glucopyranose and -L-gluco-
pyranose are enantiomers, their Fischer formulas must
be mirror images.
Simplified sugar rings are often drawn with
Haworth structural formulas. The lower edge of
the ring, which may be shown as a heavy line, is thought
of as projecting out toward the reader and the other
edge as projecting behind the plane of the paper.
HO
OH
OH
OH
O
CH OH
2

HO
OH
OH
OH
O
CH OH
2
-D-Glucopyranose
(Haworth structural
formula)
CH
2
OH
C O
CH
2
OH
CH
2
OH
C O
C H OH
CH
2
OH
CH
2
OH
C O
C HO H
C OH
CH
2
OH
CH
2
OH
C O
C HO H
C H HO
C H OH
CH
2
OH
CH
2
OH
C O
C H OH
C OH
CH
2
OH
H
CH
2
OH
C O
C H OH
C H HO
C H OH
CH
2
OH
CH
2
OH
C O
C HO H
C OH H
C H OH
CH
2
OH
CH
2
OH
C O
C H OH
C OH H
C H OH
CH
2
OH
CH
2
OH
C O
C HO H
C OH H
C H OH
C H OH
CH
2
OH
Xylulose
Sorbose
Ribulose
Sedoheptulose
Fructose
Erythrulose
Dihydroxyacetone (not chiral)
Tagatose Psicose
Haworth structures are easy to draw and unambiguous
in depicting configurations,
14
but they also do not show
the spatial relationships of groups attached to other
rings correctly. For this reason conformational formulas
of the type described in Section 2 and shown in Fig. 4-4
are used most often in this book.
Natural derivatives of sugars. The aldehyde
group of an aldose can be oxidized readily to a carboxyl
group to form an aldonic acid. Among the several
aldonic acids that occur naturally is 6-phosphogluconic
acid, which is pictured here as the 6-phosphogluconate
ion:
The enzymatically formed uronic acids have COOH
in the terminal position. This is position 6 in glucuronic
acid, whose structure is given here and as a ring in
Fig. 4-4. Sugar chains with COOH at both ends are
called aldaric acids, e.g., glucaric acid. The OH
group in the 2 position of glucose may be replaced by
NH
2
to form 2-amino-2- deoxyglucose, commonly
called glucosamine (GlcN), or by NHCOCH
3
to form N-acetylglucosamine (GlcNAc). Similar
derivatives of other sugars exist in nature. In many
poly-saccharides, sulfate groups are attached in ester
linkage to the sugar units. The sulfo (SO
3

) sugar
6-sulfo--D-quinovose (Fig. 4-4) is found in lipids
of photosynthetic membranes.
15,16
The sugar alcohols, in which the carbonyl group
has been reduced to OH, also occur in nature. For
example, D-glucitol (D-sorbitol), the sugar alcohol
obtained by reducing either D-glucose or L-sorbose
(Eq. 4-2), is a major product of photosynthesis and
widely distributed in bacteria and throughout the
eukaryotic kingdom. It is present in large amounts in
berries of the mountain ash and in many other fruits.
It exists in a high concentration in human semen and
COO
C
C
C
C
CH
2
OPO
3
2
OH
OH
OH
H
H
HO
H
H
CHO
C
C
C
C
COOH
OH
OH
OH
H
H
HO
H
H
6-Phosphogluconate ion Glucuronic acid (GlcA)

Figure 4-3 Formulas for the open forms of the D-ketoses.


165
Figure 4-4 Some simple sugars and sugar derivatives in ring forms. Most of these are present in polysaccharides.
accumulates in lenses of diabetics. D-Glucitol and
other sugar alcohols arise in some fungi during meta-
bolism of the corresponding sugars.
17
Mannitol, another
product of photosynthesis, is also present in many
organisms.
18
Another polyol that is found in human
blood in significant concentrations (0.060.25 mM) is
1,5-anhydro-D-glucitol (Fig. 4-4). It is largely of dietary
origin.
19
Two common 6-deoxy sugars which lack the
hydroxyl group at C-6 are rhamnose and fucose.
Both are of the unnatural L configuration but are
derived metabolically from D-glucose and D-mannose,
respectively.
C
C H OH
C H OH
C HO H
C HO H
CH
3
C
C HO H
C H OH
C H OH
C HO H
CH
3
O H H O
L-Rhamnose (Rha) L-Fucose (Fuc)
A. Structures and Properties of Simple Sugars
Vitamin C (ascorbic acid, Box 18-D) is another impor-
tant sugar derivative. Neuraminic acid is a 9-carbon
sugar made by transferring a 3-carbon piece onto a
hexosamine. Its N-acetyl and N-glycolyl derivatives
are called sialic acids. Their names may be abbreviated
NeuAc and NeuGl, respectively, or simply as Sia (see
also Fig. 4-4).
C
R
O H
C
R
OH H

+ BH
4
+ 3H
2
O

H
+ H
2
BO
3
Glucose or other aldose Glucitol or other alditol
(4-2)
O
OH
OH
HO
CH
2
H HO
HO
O
OH
H
HO
CH
2
OH
OH HO
O
OH
H
HO
H
OH HO
H
O
NH
H
HO
CH
2
OH
OH O
C
CH
3

OOC
H
C O
CH
3
O
OH
OH
HN H
H
H
COO

C
C
OH
H HOH
2
C
HO
H
C H
3
C
O
O
OH
H
HO
CH
2
OH
H HO
O
OH
OH
HO
HO
SO
3

4
1
6
Anion of muramic acid, O-Lactyl-GlcNAc (Mur),
2
1
-D-Glucose (Glc)
of starch, glycogen
-D-Glucose
of cellulose
3
9
1,5-Anhydro-D-glucitol
6-Sulfo--D-quinovose
Quinovose = 6-deoxy-D-glucose
Amylose (linear starch)
contains only -1,4-glycosidic
linkages involving the groups
indicated by the arrows
Branched starches
(amylopectin and glycogen)
have branches added at
this position
found in bacterial cell wall polysaccharides.
Circled group is replaced by COO

in
the anion of -d-glucuronic acid (GlcA), a
component of many mucopolysaccharides
This OH is
replaced by
in 2-acetamido-
2-deoxy-d-glucose (N-acetylglucosamine
or GlcNAc), the monomer unit of chitin
N C
O
CH
3
H
Anion of N-Acetylneuraminic acid (NeuAc),
one of the Sialic acids (Sia), nine carbon
sugars found in many glycoproteins.
-D-Xylose of plant xylans
DFructofuranose (Fru)
CH
2
OH
OH HOH
2
C
HO
HO
O
1
2
166 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
The sialic acids are prominent constituents of the
glycoproteins of cell surfaces. More than 30 modified
forms, for example, with added methyl or acetyl groups,
are known.
20 24
2. Conformations of Five- and Six-Membered
Ring Forms
Single-bonded six-membered molecular rings,
such as those in cyclohexane and in sugars, most often
assume a chair (C) conformation. For sugars in pyra-
nose ring forms, there are two possible chair confor-
mations,
4,25,26
which are designated
4
C
1
(or C1) and
1
C
4
(or 1C). The superscript and subscript numbers on the
designations indicate which atoms are above and
below the plane of the other four ring atoms. These
conformers are not easily interconvertible unless the ring is
opened by a chemical reaction, e.g., that of Eq. 4-1. How-
ever, manipulation with an atomic force microscope
has shown that stretching of single polysaccharide
chains can cause interconversion of the two chair con-
formations of pyranose rings, which are separated by
an energy barrier of ~ 46 kJ/ mol.
26a
Most sugars occur in the chair conformation that
places the largest number of substituents in equatorial
positions and is therefore most stable thermodynami-
cally. For D-aldoses this is usually the
4
C
1
conformation:
For L-aldoses it is the
1
C
4
form. There are exceptions
to this rule. For example, -D-idopyranose as well as
iduronate rings assume the
1
C
4
conformation because
this conformation places the maximum number of bulky
groups in equatorial positions.
27,28
It is noteworthy
that electronegative substituents on the anomeric
carbon atom of a sugar ring often prefer an axial
orientation. This anomeric effect is also reflected
in a shortening by about 0.01 nm of hydrogen bonds
involving the hydrogen atom of the anomeric axial
OH group. These effects can be explained partially as
a result of coulombic repulsion of the two CO dipoles
and of resonance of the following type:
2931
In addition to the chair conformations of six-mem-
bered rings the less stable boat (B) conformations are
also possible. The six boat forms are smoothly inter-
convertible through intermediate twist (T) forms,
which are also called skew (S) forms.
3,5
Since the
internal angle in a pentagon is 108 (close to the tetra-
hedral angle) we might anticipate a nearly planar five-
membered ring. However, eclipsing of hydrogen
atoms on adjacent carbons prevents formation of such
a flat structure. One of the atoms may be buckled out
of the plane of the other four about 0.05 nm, into an
envelope conformation (e.g.,
1
E), or only three atoms
may be in a plane, as in a twist conformation.
2
1
3
4
5
O
2
1
3
4
5
O
(or C1)
4
C
1
(or 1C)
1
C
4
HO
OH
O
H
H
H
H
OH
HO
OH
H All bulky substituents,
OH and CH
2
OH,
are in equatorial positions
projecting out from the ring.
All of the hydrogens
attached to ring carbons
project downward or
upward in axial positions.
-D-Glucose, a
pyranose ring
form in a chair
conformation.
CH
2
O
O
H
x
O
O
H
shortened
H-bond
~0.18 nm

+
O
1
The
1
E envelope
conformation
Boat
O
O
Skew or twist
COO
C
CH
2
C OH H
C
C
C
C
CH
2
OH
H
2
N H
HO H
OH
OH
H
H
O
CH
3
C
O
N
H
HOCH
2
C
O
N
H
Neuraminic acid anion (Neu)
Substitution here
of H2Nby
or
gives sialic acids

167
Any one of the five atoms of the ring can be either
above or below the plane defined by the other four in
the envelope conformation. The energy barriers sepa-
rating them are very low, and in cyclopentane or in
proline all of the envelope conformations are freely
interconvertible through intermediate skew forms.
32
Furanose sugar rings are very flexible but the presence
of the bulky substituents reduces the number of possi-
ble conformations.
3336a
See Chapter 5 for further
discussion.
3. Characteristic Reactions of
Monosaccharides
The aldehyde group of aldoses can either be oxi-
dized or reduced and ketoses can be reduced. The
best laboratory reagent for reduction is sodium boro-
hydride which acts rapidly in neutral aqueous solu-
tions (see Eq. 4-2). Since both NaB
3
H
4
and NaB
2
H
4
are
available, radioactive or heavy isotope labels can be
introduced in this way. The aldehyde groups can be
oxidized by a variety of agents to the corresponding
aldonic acids, a fact that accounts for the reducing
properties of these sugars. In alkaline solution aldoses
reduce Cu
2+
ions to cuprous oxide (Eq. 4-3), silver ions
to the free metal, or hexacyanoferrate (III) to hexacyano-
ferrate (II). These reactions provide the basis for sensi-
tive analytical procedures. Even though the aldoses
tend to exist largely as hemiacetals (Eq. 4-1) the reduc-
ing property is strongly evident. Oxidation by metal-
containing reagents is usually via the free aldehyde,
but oxidation by hypobromite BrO

(Br
2
in alkaline
solution) yields the lactone, as does enzymatic oxida-
tion (Eq. 15-10).
Sugars are unstable in acid. Boiling with concen-
trated HCl or H
2
SO
4
converts pentoses to furfural (Eq.
4-4a) and hexoses to hydroxymethylfurfural (Eq. 4-4b).
The aldehydes produced in these reactions can be
condensed with various phenols or quinones to give
colored products useful in quantitative estimation of
sugar content or in visualizing sugars on thin-layer
chromatographic plates. Phenol and sulfuric acid
yield a product whose absorbance at 470 nm can be
used as a measure of the total carbohydrate content of
most samples. Resorcinol (1,3-dihydroxybenzene) in
3 M HCl (Seliwanoffs reagent) gives a red precipitate
with ketoses. Orcinol (5-methylresorcinol) reacts
rapidly with pentoses and with ribonucleosides and
ribonucleotides. Since 2-deoxy sugars react slowly
this can be used as a test for RNA. Diphenylamine
with H
2
SO
4
gives a blue-green color specifically with
2-deoxy sugars and can be used to test for DNA if the
sample is first hydrolyzed.
37
B. Glycosides, Oligosaccharides,
Glycosylamines, and Glycation
The hydroxyl group on the anomeric carbon atom
of the ring forms of sugars is reactive and can be replaced
by another nucleophilic group such as OR from an
alcohol. The product is a glycoside (Eq. 4-5). The reac-
tion with methanol occurs readily with acid catalysis
under dehydrating conditions, e.g., in 100% methanol.
The alcohol in this equation can be a simple one such
as methanol or it can be any of the OH groups of
another sugar molecule. For example, two molecules
of -D-glucopyranose can be joined, in an indirect
synthesis, to form maltose (Eq. 4-6). Maltose is
formed by the hydrolysis of starch and is otherwise
not found in nature. There are only three abundant
naturally occurring disaccharides important to the
metabolism of plants and animals.
38
They are lactose
(milk), sucrose (green plants), and trehalose (fungi
and insects).
C
CHO
OH)
3
CH
2
OH
O
CHO
3H
2
O +
Furfural
(H
(4-4a)
O
CHO HOH
2
C
Hydroxymethylfurfural
Hexose
(4-4b)
(4-5)
C
R
O H
C
R
O O
+ 2Cu
2
+ 2OH + Cu
2
O + H
2
O
+

B. Glycosides, Oligosaccharides, Glycosylamines, and Glycation


(4-3)
O
OH
OH
HO
CH
2
OH
H HO
H
O R
O
OH
O
HO
H HO
R
The carbon atom that is
attached directly to two
oxygen atoms in the
hemiacetal group is the
anomeric carbon atom.
The attached OH group
can be replaced to form
a glycoside.
A glycoside (an -d-
glucopyranoside
in this instance).
The molecule HOR
is called the aglycone
of the glycoside.
168 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
Disaccharides are linked by glycosidic (acetal)
linkages. The symbol -1,4, used in Eq. 4-6, refers to
the fact that in maltose the glycosidic linkage connects
carbon atom 1 (the anomeric carbon atom) of one ring
with C-4 of the other and that the configuration about
the anomeric carbon atom is . While the and ring
forms of free sugars can usually undergo ready inter-
conversion, the configuration at the anomeric carbon
atom is frozen when a glycosidic linkage is formed.
To describe such a linkage, we must state this configu-
ration together with the positions joined in the two
rings (see Eq. 4-6). Lactose, whose structure follows,
can be described as a disaccharide containing one
galactose unit in a -pyranose ring form and whose
anomeric carbon atom (C-1) is joined to the 4 position
of glucose, giving a -1,4 linkage:
The systematic name for -lactose, O--D-galacto-
pyranosyl-(14)--D-glucopyranose, provides a com-
plete description of the stereochemistry, ring sizes, and
mode of linkages. This name may be abbreviated -D-
Galp-(14)--D-Glcp. Since pyranose rings are so
common and since most natural sugars belong to the
D family, the designations D and p are often omitted.
It may be assumed that in this book sugars are always
D unless they are specifically designated as L. When
linkages remain uncertain abbreviated formulas are
given. Because the glucose ring in lactose is free to
open to an aldehyde and to equilibrate (in solution)
with other ring forms, the name lactose does not imply
a fixed ring structure for the glucose half. Thus, the
name lactose can be abbreviated as Gal-(14)-Glc or,
more succinctly, as Gal14Glc. However, in crystal-
line form the sugar exists either as -lactose or -lactose.
The latter is more soluble and sweeter than -lactose,
which sometimes crystallizes in ice cream upon pro-
longed storage and produces a sandy texture. An
isomer of lactose, Gal16Glc or allolactose, is an
important inducer of transcription in cells of E. coli
(Chapter 28).
Notice that in the drawing of the lactose structure
the glucose ring has been flipped over with respect
to the orientation of the galactose ring, a consequence
of the presence of the -1,4 linkage. For maltose,
where the linkage is -1,4, the two rings are usually
drawn with the same orientation (Eq. 4-6). Maltose
can be described as -D-Glcp-(14)-D-Glcp or more
simply as Glc14Glc.
In sucrose and in ,-trehalose the reducing groups
of two rings are joined. Each of these sugars exists in
a single form. Sucrose serves as the major transport
sugar in green plants, while trehalose plays a similar
role in insects, as does D-glucose in our blood.
Trehalose, or mushroom sugar, is found not only in
fungi but also in many other organisms.
3941
. It serves
as the primary transport sugar in the hemolymph of
insects and also acts as an antifreeze in many species.
It accounts for up to 20% of the dry weight of anhy-
drobiotic organisms, which can survive complete
dehydration. These include spores of some fungi,
yeast cells, macrocysts of Dictyostelium, brine shrimp
cysts (dried gastrulas of Artemia salina), some nema-
todes, and the resurrection plant. These organisms
can remain for years in a dehydrated state. Hydrogen
bonding between the trehalose and phosphatidylcholine
may stabilize the dry cell membranes.
18,40,41
Although
they can be desiccated, fungal spores remain dormant
even when considerable water is present. One of the
first detectable changes when the spores germinate is a
rapid increase in the activity of the enzyme trehalase
O
OH
OH
CH
2
OH
HO
O
OH
OH
CH
2
OH
HO
O
-Lactose
Note
configuration
at this "reducing
end" of the
molecule of
-lactose
1 4
D-Glucose D-Galactose

O
O
O HOH
2
C
HO
CH
2
OH
OH
OH
HOCH
2
HO
HO
O
O
OH
HOCH
2
HO
HO
O
HO
OH
OH
CH
2
OH
Sucrose: Glcp12Fruf
, -Trehalose: Glcp11Glcp
1
2
(4-6)
O
OH
OH
HO
HOH
2
C
H
HO
O
OH
OH
O
HOH
2
C
H
HO
O
OH
HO
HOH
2
C
H
HO
O
OH
HOH
2
C
H
HO
H
2
O
-D-Glucopyranose (2 molecules)
1
OH
4
-1,4 Glycosidic linkage
O
Maltose, a disaccharide
H

169
which hydrolyzes trehalose to glucose.
42,43
Disaccharides, as well as higher oligosaccharides
and polysaccharides, are thermodynamically unstable
with respect to hydrolysis, for example, for lactose in
aqueous solution:
Lactose + H
2
O D-glucose + D-galactose
G 8.7 0.2 kJ mol
1
at 25C
The corresponding equilibrium constant K = [D-glucose]
[D-galactose]/[lactose] 34 M. For other oligosaccha-
rides K varies from 17 to 500 M.
44,45
However, sucrose
is far less stable with K = 4.4 10
4
M and G = 26.5
0.3 kJ mol
1
for its hydrolysis.
46
Sucrose is also less
stable kinetically and undergoes rapid acid-catalyzed
hydrolysis. This fact is exploited when vinegar is used
to convert sucrose to the less crystallizable mixture of
glucose and fructose during candy-making. In marked
contrast, trehalose is extremely resistant to acid-cata-
lyzed hydrolysis.
The joining of additional sugar rings through
glycosidic linkages to a disaccharide leads to the for-
mation of oligosaccharides, which contain a few
residues, and to polysaccharides, which contain
many residues. Among the well-known oligosaccha-
rides are the substituted sucroses raffinose, Galp(16)
Glcp(12) Fruf, and stachyose, Galp(16) Galp(16)
Glcp(12) Fruf. Both sugars are found in many legumes
and other green plants in which they are formed by
attachment of the galactose rings to sucrose. Oligo-
saccharides have many functions. For example, gram-
negative bacteria often synthesize oligosaccharides
of 612 glucose units in -1,2 linkage joined to sn-1-
phosphoglyceryl groups. They are found in the peri-
plasmic space between the inner and outer cell mem-
branes and may serve to control osmotic pressure.
47
Oligosaccharides of 1014 -1,4-linked D-galacturonic
acid residues serve as signals of cell wall damage to
plants and trigger defensive reactions against bacteria
in plants.
48
Just as alcohols can be linked to sugars by glyco-
side formation (Eq. 4-5), amines can react similarly to
give glycosylamines (N-glycosides). In this instance
it is usually the free aldehyde that reacts via formation
of a Schiff base (Eq. 4-8a). The latter can cyclize (Eq. 4-8b)
to the glycosylamine with either the or configuration.
Another important reaction of Schiff bases of sugars
is the Amadori rearrangement (Eq. 4-8c,d), which
produces a secondary ketoamine with a 1-oxo-2-deoxy
structure.
This can cyclize as in step e of Eq. 4-8. In addition,
epimerization at position 2 occurs through the reversal
of step d. The overall reaction of Eq. 4-8 is often called
glycation to distinguish it from glycosylation, the
transfer of a glycosyl group. The Amadori rearrange-
ment is important in nitrogen metabolism and in non-
enzymatic reactions of sugars. For example, small
(4-8)
amounts of glycosylated hemoglobin and other proteins
modified by glycation of the protein amino groups are
normally present in the blood.
4951
People with diabe-
tes, who have a high concentration of blood glucose,
have increased amounts of glycated protein. High
concentration of either glucose or fructose
41,52
may
cause serious problems. For example, modification of
the protein crystallins of the lens of the eye may lead
to cataracts. Similar problems with galactose may
accompany galactosemia (Chapter 20).
53
Ketoamines
formed by glycation of proteins may undergo crosslink-
ing reactions with side chains of other protein molecules
and this may be one cause of aging.
42,43
Other reactions
of the ketoamines lead to formation of fluorescent and
colored products.
41,43,50,5456
Oxidation products of sugars
also participate in these reactions,
57
and nucleic acid
bases also react.
56
C. Polysaccharides (Glycans)
Polymers of sugars are present in all cells and
serve a variety of functions.
5860
The simple sugars
commonly used in the assembly of polysaccharides
include D-glucose, D-mannose, D-galactose, D-fructose,
D-xylose, L-arabinose, related uronic acids, and amino
sugars (Fig. 4-4). These monomer units can be put
together in many ways, either as homopolysaccha-
rides containing a single kind of monomer or as hetero-
polysaccharides containing two or more different
monomers. Because there are many sugars and many
ways in which they can be linked, there is a bewildering
C. Polysaccharides (Glycans)
O
HO
H
HO
CH
2
N HO R
CH
2
CH
2
OH
HO
OH
N R
C
CH
2
OH
HO
OH
OH
C
CH
2
OH
HO
OH
OH
O
N R
H
CH
2
H
OH
HO
OH
HO
+ H
2
NR
HO
H
OH
1
N
HO
R
H
H
Glycosylamine
H
2
O
HO
Ketoamine
HO
HO
CH
2
OH
N R
HO
Amadori
rearrangement
H
O
CH
2
HO
+
a
b
c
d
e
(4-7)
170 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
variety of different polysaccharides. Their chains can
be linear or helical. The most numerous functional
groups present are the free hydroxyl groups, some
of which may form additional glycosidic linkages to
produce branched chains. Polysaccharides may also
contain COOH, NH
2
, NHCOCH
3
, and other groups.
After polymerization, hydroxyl groups are sometimes
methylated or converted to sulfate esters or to ketals
formed with pyruvic acid. Structural characteristics of
some of the major polysaccharides are listed in Table 4-1.
1. Conformations of Polysaccharide Chains
Despite the variety of different monomer units
Starch Green plants
Amylose D-Glucose 1,4
Amylopectin D-Glucose 1,4 1,6
Glycogen Animals, bacteria D-Glucose 1,4 1,6
Cellulose Green plants, some bacteria D-Glucose 1,4
Dextrans Some bacteria D-Glucose 1,6 1,3
Pullulan Yeast D-Glucose 1,6 + 1,4
Callose Green plants D-Glucose 1,3
Yeast glucan Yeast D-Glucose 1,3
Schizophyllan, curdlan, paramylon D-Glucose 1,3 1,6 on every third residue
Mannans Algae D-Mannose 1,4
Yeast D-Mannose 1,6
Xylans Green plants D-Xylose 1,3
Brown seaweed
Inulin Some plant tubers D-Fructose 2,6
Chitin Fungi, anthropods N-acetyl-D-Glucosamine 1,4
Alternating polysaccharides
Hyaluronan Animal connective tissue Glucuronic acid +
N-acetylglucosamine 1,4
Chondroitin sulfate D-Glucosamine
N-acetyl-D-Galactosamine 1,3 + 1,4
Dermatan sulfate -L-Iduronate +
N-acetyl-D-Galactosamine 1,3 + 1,4
Pectin Higher plants D-Galactunonate + others 1,4 + others
Alginate Seaweed D-Mannuronate +
L-Guluronate 1,4 + 1,4
Agar-agar Red seaweed Galactose 1,4 and 1,3
Carageenan Red seaweed Galactose-4-sulfate + 1,4 + 1,3
3,6-anhydro-
D-Galactose-2-sulfate
Murein Bacterial cell wall N-acetyl-D- 1,4
Glucosamine + N-
acetyl-D-Muramic acid
TABLE 4-1
Some of the Many Polysaccharides Found in Nature
Name Source Monomer Main linkage Branch linkages
and kinds of linkage present, the conformational
possibilities for carbohydrate chains are limited.
The sugar ring is a rigid unit and the connection of
one unit to the next can be specified by means of two
torsion angles and just as with peptides.
6163
To
specify a torsion angle four atoms must be selected
the two at the ends of the bond about which rotation
is being considered and two others. There is more
than one way to define the zero angle for and . As
illustrated in the drawing on p. 172, may be taken
as the H1C1OC4' dihedral angle and as the H4'
C4' OC1 angle. The zero angle is when H1 and
H4' are eclipsed. A related alternative is to take and
as 0 when the two midplanes of the sugar rings are
coplanar.
171
O
O
CH
2
O HO
O
O
O
O H O
O
CH
2
O
O H
O H
H
O
CH
2
O
O H
H O
H
HO
H
H
O CH
2
H O
H

BOX 4-A CYCLODEXTRINS


An enzyme produced by some bacteria of the
genus Bacillus cuts chains of amylose and converts
them into tiny rings consisting of six (-cyclodextrin),
seven (-cyclodextrin), eight (-cyclodextrin), or
more glucose units. The cyclodextrins, which were
first isolated by F. Schardinger in 1903, have in-
trigued carbohydrate chemists for many years by
their unusual properties.
a
They are surprisingly
resistant to acid hydrolysis and to attack by amy-
lases and cannot be fermented by yeast.
The torus-like cyclodextrin molecules have an
outer polar surface and an inner nonpolar surface.
The small hydrophobic
cavities within the
cyclodextrins have
diameters of 0.50, 0.62,
and 0.79 nm, respec-
tively, for the , , and
dextrins.
b
Cavities
are potential binding
sites for a great variety
of both inorganic and organic molecules.
ag
Com-
plexes of simple alcohols, polyiodides,
c
ferrocene,
d
and many other compounds have been observed.
Cyclodextrins can be used to encapsulate food
additives
b
and their complexes may be useful in
1.5 nm
0.62 nm
0.79 nm
a
French, D. (1957) Adv. Carbohydr. Chem. Biochem. 12, 189260
b
Korpela, T., Mattsson, P., Hellman, J., Paavilainen, S., and
Mkel, M. (198889) Food Biotechnology 2, 199210
c
Noltemeyer, M., and Saenger, W. (1980) J. Am. Chem. Soc. 102,
27102722
d
Menger, F. M., and Sherrod, M. J. (1988) J. Am. Chem. Soc. 110,
86068611
e
Hamilton, J. A., and Chen, L. (1988) J. Am. Chem. Soc. 110,
43794391
f
Armstrong, D. W., Ward, T. J., Armstrong, R. D., and Beesley, T.
E. (1986) Science 232, 11321135
g
Lipkowitz, K. B., Raghothama, S., and Yang, J. (1992) J. Am.
Chem. Soc. 114, 11541162
h
Anslyn, E., and Breslow, R. (1989) J. Am. Chem. Soc. 111,
89318932
i
Granados, A., and de Rossi, R. H. (1995) J. Am. Chem. Soc. 117,
36903696
j
Ueno, A., Kuwabara, T., Nakamura, A., and Toda, F. (1992)
Nature 356, 136137
k
Harada, A., Kamachi, J. L., and Kamachi, M. (1993) Nature 364,
516518
separation of enantiomers of drugs.
f,g
They can be
chemically modified by adding catalytic groups to
serve as enzyme models
h,i
or as color-change
indicators sensitive to binding of organic molecules.
j
They can be linked together to form molecular
nanotubes. Polymer chains can even be threaded
through the tubes.
k
What practical applications
may yet come from this?
Hydrogen
Oxygen
Carbon
Figure 4-5 (A) Linkages of the D-glucose
residues in cellulose. (B) Hydrogen-bonding
pattern in cellulose I. From Ross et al.
66
A
B
C. Polysaccharides (Glycans)
172 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
The IUB has proposed another convention.
64
2. The Glucans
From glucose alone various organisms synthesize
a whole series of polymeric glucans with quite different
properties. Of these, cellulose, an unbranched -1,4-
linked polyglucose (Fig. 4-5A), is probably the most
abundant. It is the primary structural polysaccharide
of the cell walls of most green plants.
65
For the whole
Earth, plants produce ~10
14
kg of cellulose per year.
A systematic examination of the possible values
for and shows that for cellulose these angles are
constrained to an extremely narrow range which
places the monomer units in an almost completely
extended conformation.
62
Each glucose unit is flipped
over 180 from the previous one. The polymer has a
twofold screw axis and there is a slight zigzag in the
plane of the rings.
6668
Remember that in the chair
form of glucose all of the OH groups lie in equatorial
positions and are able to form hydrogen bonds with
neighboring chains. This feature, together with the
rigidity of conformation imposed by the configura-
tion of the monomer units, doubtless accounts for the
ability of cellulose to form strong fibers.
It has been impossible to obtain large single crys-
tals of cellulose. However, from 60 to 90% of native
cellulose is thought to be clustered to form the needle-
like crystalline microfibrils. These microcrystals of
cellulose I can be separated from other plant materials
by prolonged boiling with dilute NaOH and HCl.
Their structure has been established by electron dif-
fraction
67
and by comparison with high-resolution
X-ray diffraction structure of the tetrasaccharide -D-
cellotetraose.
66,69
Two closely similar parallel-chained,
hydrogen-bonded structures appear to be present.
One is shown in Fig. 4-5B. The hydrogen bonds and
van der Waals forces bind the chains into sheets which
are stacked to form fibers. A typical fiber of plant
cellulose has a diameter of 3.54 nm and contains 30
40 parallel chains, each made up of 200010,000 glucose
units. The chain ends probably overlap to form essen-
tially endless fibers that can extend for great distances
through the cell wall. They interact with other polysac-
charides as is illustrated in Fig. 4-14. A single cotton
Figure 4-6 Scanning electron micrographs of starch granules.
Magnification 2500 and 3000. From Jane et al.
73
fiber may be 23 cm in length.
Cotton thread treated with concentrated NaOH
shrinks and has an increased luster. The resulting
Mercerized cellulose has changed into other crystal-
line forms. The major one is cellulose II, in which
the chains in the sheets are antiparallel.
69a
Cellulose II
may also occur to some extent in nature. Many other
modified celluloses, e.g., methylcellulose, in which
some OH groups have been converted to methyl
ethers
70
are important commercial products.
Starch, another of the most abundant polymers
of glucose, is stored by most green plants in a semi-
crystalline form in numerous small granules. These
granules, which are usually formed within colorless
membrane-bounded plastids, have characteristic shapes
and appearances (Fig. 4-6) that vary from plant to plant.
One component of starch, amylose, is a linear poly-
mer of many -D-glucopyranose units in 1,4 linkage
(Fig. 4-7) as in maltose. Starch granules always contain
a second kind of molecule known as amylopectin.
58
Potato Rice
Shoti Immature sweet corn
Wheat
10 m
10 m
Maize
10 m
15 m
10 m
10 m
l =
0
.4
2
n
m
-1,4-Linked
D-Glucose units
H
4
1
H
O
O
OH
HO
OH
O
O
OH
HO
OH
O

173
from
reducing
end
7 nm
1
0
-
1
2

n
m
~3 units
~6-7 units

O
OH
CH
2
HO
OH
O

O
H
2
C
O
O
HO
OH
OH
O
H
2
C
HO
O
HO
O
OH
OH
CH
2
HO
O
O
The linear chains of amylase grow
from the reducing end toward
the non-reducing end using only
1,4 linkages as in maltose.
For each residue the chain
bends by ~60 leading to
a helical structure with 6
residues per turn.
Branches in
amylopectin or
glycogen are
attached by
1,6 linkages.
~
6
0

~
6
0

Figure 4-7 (A) Linkages of the glucose residues in starches


and in glycogen. (B) Schematic diagram of the glycogen
molecule as proposed originally by K. H. Meyer.
74
The
circles represent glucose residues which are connected by -
1,4 linkages and, at the branch points, by -1,6 linkages. The
symbol designates the reducing group. From D. French.
75
(C) Proposed broomlike clusters in amylopectin. After D.
French.
71
Both amylopectin and glycogen (animal starch) con-
sist of highly branched bushlike molecules. Branches
are attached to -1,4-linked chains through -1,6
linkages (Fig. 4-7). When glucose is being stored as
amylopectin or glycogen the many nonreducing
ends appear to grow like branches on a bush, but
when energy is needed the tips are eaten back by
enzymatic action (Fig. 11-2). There is only one reducing
end of a starch molecule where one might expect to
find a hemiacetal ring in equilibrium with the free
aldehyde. However, this end may be attached to a
protein.
Starch granules show a characteristic pattern of
growth rings of 0.3- to 0.4-m thickness with thinner
dense layers about 7 nm apart. Study of this layered
structure suggests that the individual amylopectin
molecules have branches close together in broomlike
clusters (Fig. 4-7C).
61,71,72
The amylopectin chains are
120400 nm long and their relative molecular masses
may reach 1530 million. In addition, starch granules
usually contain molecules of the straight-chain amylose,
each containing several hundred glucose units and
having molecular masses of ~100 kDa. Most starches
contain 2021% amylose but there are special varieties
of plants that produce starch with 5070% amylose.
On the other hand, the waxy varieties of maize form
only amylopectin and lack amylose.
In both starch and glycogen the glucose units of
the main chains are linked with -1,4 linkages. An
extended conformation is not possible and the chains
tend to undergo helical coiling. One of the first helical
structures of a biopolymer to be discovered (in 1943)
76,77
was the left-handed helix of amylose wound around
molecules of pentaiodide (I
5

) in the well-known blue


starchiodine complex
78
(Fig. 4-8). The helix contains
six residues per turn, with a pitch of 0.8 nm and a
diameter of nearly 14 nm. Amylose forms complexes
of similar structure with many other small molecules.
79
Another more tightly coiled double-helical form
of amylose has been proposed.
80
Each chain would
contain six glucose units per turn and the two chains
could be arranged in either parallel or antiparallel
directions. The average amylose molecule contains
2000 glucose units and could be stretched to a slender
chain over 1 m long, longer than the crystalline regions
observed in starch granules. Thus, the chains within
the granules would have to fold back on themselves,
possibly in hairpin fashion:
X-ray diffraction studies support the double-helical
structure but suggest a parallel orientation of the amy-
lose chains.
81
Since amylose has not been obtained as
single crystals the diffraction data do not give a definitive
answer. However, if double helices are formed by
adjacent branches in amylopectin and glycogen the
two strands would be parallel. Starch granules also
contain amorphous starch which appears to contain
single helices, possibly wrapped around lipid materials.
82
A
B
C
C. Polysaccharides (Glycans)
Crystalline array of hairpin folds
proposed for linear starch molecules
174 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
Glycogen is stored in the cytoplasm of animal cells
and to some extent in the lysosomes as enormous 100-
to 200-MDa particles. These appear in the electron
microscope as aggregates of smaller particles of mole-
cular masses up to 2040 MDa. A laminated internal
structure with surface bumps is suggested by STM
microscopy (Fig. 4-9). Biosynthesis of glycogen may
be initiated by a 37-kDa protein called glycogenin,
which remains covalently attached to the reducing
end of the glycogen (Chapter 20).
83
Despite the huge
molecular masses of glycogen particles, both
1
H and
13
C NMR resonances are sharp, indicating a high
degree of mobility of the glycosyl units.
84
Beta-1,3-linked glucans occur widely in nature.
When a new green plant cell is formed the first poly-
saccharide to be synthesized is not cellu-
lose but the -1,3-linked glucose polymer
callose. Cellulose appears later. Callose
is also produced in some specialized
plant tissues, such as pollen tubes,
85
and
is formed in massive amounts at the site
of wounds or of attack by pathogens.
The major structural component of the
yeast cell wall is a -1,3-linked glucan
with some -1,6 branches.
86
Schizophyl-
lan is a -1,3-linked glucan with a -1,6-
linked glucosyl group attached to every
third residue. A glucan from the coleop-
tiles of oats contains 30% -1,3 linkages
in a linear chain that otherwise has the
structure of cellulose.
87
Other -1,3-
linked glucans serve as energy storage
molecules in lower plants and in fungi.
Among these are -1,3-linked glucans
such as paramylon,
88,89
which is stored
by the euglena. A similar polysaccharide,
curdlan, is formed by certain bacteria.
90
Some other bacteria, e.g., Leuconostoc
mesenteroides, make 1,6-linked poly-D-
glucose or dextrans.
91,92
These always
contain some -1,3-linked branches and
may also have -1,4 and -1,2 linkages,
the structures varying from species to
species. Dextrans formed by bacteria
such as Streptococcus mutans growing on
the surfaces of teeth are an important
component of dental plaque. Bacterial
Figure 4-8 (A) Structure of the helical complex of amylose with I
3

or I
5

.
The iodide complex is located in the interior of the helix having six glucose
residues per turn. (B) Model of a parallel-stranded double helix. There
are six glucose units per turn of each strand. The repeat period measured
from the model is 0.35 nm per glucose unit. Courtesy of Alfred French.
A
B
Figure 4-9 A scanning tunneling microscopic (STM) image
of three glycogen molecules. The corrugated surface
suggests a laminated structure. These molecules have been
purified from tissues by treatment with strong alkali, which
breaks the larger aggregates into particles of M
r
= 110 10
6
and diameter 2530 nm. Courtesy of Fennell Evans.
93
175
dextrans are also produced commercially and are
chemically crosslinked to form gels (Sephadex) which
are widely used in biochemical separation procedures
(Figs. 3-1 and 4-10). A yeast polysaccharide pullulan
is a regular polymer of maltotriose units with -1,4
linkages joined in a single chain by -1,6 linkages.
94
One glucan, called alternan is a linear polymer with
alternate -1,6 and 1,3 linkages.
3. Other Homopolysaccharides
Cell walls of yeasts contain mannans in which
the main -1,6-linked chain carries short branches of
one to three mannose units joined in -1,2, -1,3, and
sometimes -1,6 linkages.
96
These are covalently linked
to proteins.
97
A -1,4-linked mannan forms micro-
fibrils in the cell wall of some algae such as Acetabu-
laria (Fig. 1-11) which do not make cellulose.
98,99
The cell walls of some seaweeds contain a -1,3-linked
xylan instead of cellulose. This polysaccharide forms
a three-stranded right-handed triple helix.
99,100
Even
though xylose is a five-carbon sugar, the polymer
contains the thermodynamically more stable six-
membered pyranose rings. On the other hand, fructose,
a 6-carbon sugar, is present as five-membered furanose
rings in inulin, the storage polysaccharide of the Jeru-
salem artichoke and other Compositae, and also in
sweet potatoes. The difference has to do with biosyn-
thetic pathways. Furanose rings arise both in inulin
and in sucrose because the biosynthesis occurs via the
6-phosphate ester of fructose, making it impossible for
the phosphate derivative to form a 6-membered ring.
The major structural polysaccharide in the exo-
skeletons of arthropods and of other lower animal
forms is chitin, a linear -1,4-linked polymer of N-
C. Polysaccharides (Glycans)
acetylglucosamine whose structure resembles that of
cellulose. In chitin the individual parallel chains
are linked by hydrogen bonds to form sheets in which
parallel chains are held together by NH- - - O= C
hydrogen bonds between the carboxamide groups.
In the more abundant chitin the chains in alternate
sheets have opposite orientations,
101,102
possibly a
result of hairpin folds in the strands. Native chitin
exists as microfibrils of 7.25 nm diameter. These
contain a 2.8-nm core consisting of 1530 chitin chains
surrounded by a sheath of 27-kDa protein subunits.
The microfibrils pack in a hexagonal array, but the
structure is not completely regular. Several proteins
are present; some of the glucosamine units of the
polysaccharide are not acetylated and the chitin core
is often calcified.
103
The commercial product chitosan
is a product of alkaline deacetylation of chitin but it
also occurs naturally in some fungi.
102
Chitin is also
present in cell walls of yeasts and other fungi. It is
covalently bonded to a -1,3-linked glycan which may,
in turn, be linked to a mannoprotein (see Section D,2).
97
4. Heteropolysaccharides of the Animal Body
Many polysaccharides contain repeating units
consisting of more than one different kind of mono-
mer.
104,105
Some of these are composed of two sugars
in a simple alternating sequence. Examples are
hyaluronan (hyaluronic acid) and the chondroitin,
dermatan, keratan, and heparan sulfates. They
are important components of the ground substance
or intracellular cement of connective tissue in animals.
Hyaluronan,
106,107
which is abundant in synovial fluid
and the vitreous humor of the eye, is a repeating poly-
mer of glucuronic acid and N-acetylglucosamine with
the structure shown in Fig. 4-11 and M
r
of several
million. The chondroitin sulfates and dermatan sulfate
are similar polymers but with substitution by N-acetyl-
galactosamine and -L-iduronic acid, respectively, and
with sulfate ester groups in the positions indicated in
Fig. 4-11.
Hyaluronan solutions are remarkably viscous and
at a concentration of only 0.1% can have over 80% of
the typical viscosity of biological fluids. This property
may result from the presence of hydrogen bonds
between the carboxylate, carboxamide, and hydroxyl
groups of adjacent sugar residues as in Fig. 4-12. The
hydrogen bonds stiffen the chain to give a slender rod.
The tetrasaccharide shown in the figure is the repeating
unit in a threefold helix.
109
However, the charged
molecules do not associate to form strong fibrils like
those of cellulose. The chain can be bent easily with
breakage of the H-bonds at various positions to give
a random coil structure.
While hyaluronan is not covalently attached to
proteins, it is usually anchored to cell surfaces and to
Figure 4-10 A schematic representation of the gel networks
of Sephadex (left) and agarose (right). Note that the aggre-
gates in agarose gels may actually contain 1010
4
helices
rather than the smaller numbers shown here. From Arnott
et al.
95
176 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
O
O
O
C
O O
O O
O
N
O
CH
2
OH
H
H O
C
CH
3
O
CH
2
OH
O
N
O
H
C H
O
C
O
O
O
O H
O
H O
CH
3
O
H
H
H

Figure 4-12 Proposed hydrogen-bonding scheme for the native conformation of hyaluronan. See Morris et al.
109
Type I Chain. Alternating 13 and 14 linkages; all , (equatorial).
Figure 4-11 The repeating disaccharide units of hyaluronan and other glycosaminoglycans. See Fransson
108
and Hardingham
and Fosang.
107
Name Repeating unit Variations
Heparan sulfate [4-GlcA14GlcNAc]
n
Some IdoA, sulfation.
Heparin [4-L-IdoA(SO
3
)
14GlcN(SO
3
)
1]
n
Some IdoA-2-SO
3

, GlcNAc.
24 different disaccharides possible.
Name Repeating unit Variations
Hyaluronan [4GlcA13GlcNAc1]
n
None. Homogeneous.
Chondroitin sulfate [4GlcA13GalNAc(SO
3

]1)
n
Some GlcA-2-SO
3

,
GalNAc-4- or 6-SO
3

or both.
Dermatan sulfate [4IdoA13GalNAc(4-SO
3

]1)
n
Some L-IdoA-2-SO
3

.
Keratan sulfate [3Gal14GlcNAc(6-SO
3

]1)
n
Some Gal-6-SO
3
, some Sia, Man, Fuc.
Type II Chain. All 14 linkages, alternating and (axial).
O
OH
O
OOC
O HO
O
NH
O
3
SO
CH
2
O
O
OOC
O
OH
HO
C O
CH
3
OH

4
GlcA GlcA GalNAc-4-Sulfate

Repeating unit
O
O
O
O HO
O
CH
2
O
NH
HO
C O
CH
2
O
OSO
3
O HO
COO
COO
SO
3
OH
IdoA GlcNAc

177
protein receptors within the extracellular matrix.
In contrast, chondroitin, dermatan, keratan, and heparan
sulfates are attached covalently to the proteins at the
reducing ends of the polymer chains (see Section D).
The attached polymers undergo enzyme-catalyzed
chemical alteration. In dermatan most of the glucu-
ronate residues found in chondroitin have been epimer-
ized to iduronate and sulfate groups in ester linkages
have been added. Chondroitin sulfate is especially
abundant in cartilage; dermatan sulfate is concentrated
in skin. Heparan sulfates are more heterogeneous
than the other polymers of this group and have been
described as the most complex polysaccharides on
the surface of mammalian cells.
110
In the mast cells of lungs, liver, and other cells
heparan sulfate chains are modified further and
released into the bloodstream as heparin, a material
with important anticoagulant properties. Both the
amino groups and the 6-hydroxyls of the glucosamine
residues of heparin carry sulfate groups. In some units
D-glucuronic acid or glucuronate-2-sulfate
111
is present
in -1,4 linkage, but more often L-iduronic acid-2-sulfate
is the first unit in the disaccharide.
112114
The iduronate
ring appears to have unusually high conformational
flexibility which may influence the biological activity
of heparin and related polysaccharides.
115
Because of
its anticoagulant property, heparin is an important
drug for prevention of blood clot formation.
116
In the
United States, in 1976 six metric tons of heparin were
used to treat 10 million patients.
117
The anticoagulant
activity resides in large part in a nonrepeating penta-
saccharide (Fig. 4-13).
112,118
This portion of the heparin,
especially if part of a larger octasaccharide,
113
binds
to several proteins including the enzyme inhibitors
heparin cofactor II and plasma antithrombin III
(Chapter 12). Heparin greatly accelerates the rate at
which these proteins bind and inactivate blood clotting
factors. See also Section D,1. Lower invertebrates, as
well as marine brown algae, contain heavily sulfated
fucans which are largely 1,3-linked.
119
5. Plant Heteropolysaccharides
Fibers of cellulose, which run like rods through
the amorphous matrix of plant cell walls, appear to be
coated with a monolayer of hemicelluloses. Predom-
inant among the latter is a xyloglucan, which has the
basic cellulose structure but with -1,6-linked xylose
units attached to three-fourths of the glucose resi-
dues.
104,120,121
L-Fucose may also be present in trisac-
charide side chains: L-Fuc12Gal1 2Xyl1.
122
Pectins of higher plants contain -1,4-linked poly-
galacturonates interrupted by occasional 1,2-linked
L-rhamnose residues. Some of the carboxyl groups of
these rhamnogalacturonan chains are methylated.
123
Arabinans and galactans are also present in pectin.
A possible arrangement of cellulose fibers, hemicellu-
loses, and pectic materials in a cell wall has been
proposed (Fig. 4-14).
Agarose, an alternating carbohydrate polymer
consisting of ~120-kDa chains, is the principal compo-
nent of agar and the compound that accounts for most
of the gelling properties of that remarkable substance.
A solid agar gel containing 99.5% water can be formed.
Agarose molecules form left-handed double helices
with a threefold screw axis, a pitch of 1.90 nm, and a
central cavity containing water molecules.
124,125
A similar structure has been established for the gel-
forming carrageenans from red seaweed. The X-ray
data suggest that three of the disaccharide units form
one turn of a right-handed helix with a pitch of 2.6 nm.
A second chain with a parallel orientation, but displaced
by half a turn, wraps around the first helix.
124
Such
O
NH
O
O
CH
2
HO
OSO
3
C
O
H
3
C
O
OH
COO
O
HO
O
O O
O
O
O
H
2
C
OSO
3
O
3
S NH
O
3
S
HO
OOC
SO
3
O
NH
O
CH
2
HO
OSO
3
SO
3

Figure 4-13 A pentasaccharide segment of heparin


which binds with high affinity to the serum protein
antithrombin causing it to inhibit most of the serine
protease enzymes participating in the blood coagulation
process (see Chapter 12). See Lindahl et al.
118
O
O
O
OSO
3
OH
OH
O
HO
3
n
O
Agarose:
[-3-D-Gal14-(3,6-anhydro)-L-Gal1]
n
1 4
1
OH

C. Polysaccharides (Glycans)
178 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
BOX 4-B SILICON: AN ESSENTIAL TRACE ELEMENT
No one can doubt that
diatoms, which make their
skeletons from SiO
2
, have an
active metabolism of silicon.
They can accumulate as much
as 0.7 M soluble silicon,
possibly attached to
proteins.
a-d
The radiolaria and
sponges often accumulate
silicon and limpets make opal base plates for their
teeth. Silicon may account for as much as 4% of the
solids of certain grasses. Although silicon is usually
not considered an essential nutrient for all plants,
there is much evidence that it is essential to some
and that it is often beneficial.
f
Silicon is found in
soil primarily as silicic acid, H
4
SiO
4
, whose concen-
tration ranges from 0.10.6 mM. Most of the silicon
taken up by plants is deposited within cells, in cell
walls, between cells, or in external layers as hydrated
SiO
2
. Presumably the organic components of the
plant control the deposition. The SiO
2
in some
plants takes the form of sharp particles which may
have a defensive function. They abrade the enamel
surfaces of the teeth of herbivores and can cause
other illnesses.
g
Silicon is essential for growth and development
of higher animals,
h-l
and it has been suggested that
humans may require 520 mg per day.
m
In the chick,
silicon is found in active calcification sites of young
bone.
i
Silicon-deficient animals have poorly calcified
bones and also an elevated aluminum content in
their brains.
m
Silicon is present in low amounts in
the internal organs of mammals but makes up ~0.01%
of the skin, cartilage, and ligaments, in which it is
apparently bound to proteoglycans such as chon-
droitin-4-sulfate, dermatan sulfate, and heparan
sulfate (Fig. 4-11).
m,n
These polymers contain ~0.04%
silicon or one atom of silicon per 130280 repeating
units of the polysaccharides. Plant pectins contain
about five times this amount. The silicon is appar-
ently bound tightly in ether linkage. Perhaps ortho-
silicic acid, Si(OH)
4
, reacts with hydroxyl groups of
the carbohydrates to form bridges between two
chains as follows:
In each of these formulas additional free OH groups
are available on the silicon so that it is possible to
crosslink more than two polysaccharide chains.
Silicon may function as a biological crosslinking
agent in connective tissue. Silaffins, small polypep-
tides containing polyamine side chains of modified
lysine residues, apparently initiate silica formation
from silicic acid in diatoms.
o
a
Robinson, D. H., and Sullivan, C. W. (1987) Trends Biochem. Sci.
12, 151154
b
Round, F. E. (1981) in Silicon and Siliceous Structures in Biological
Systems (Simpson, T. L., and Volcani, B. E., eds), pp. 97128,
Springer, New York
c
Evered, D., and OConnor, M. (1986) Silicon Biochemistry, Wiley,
New York
d
Round, F. E., Crawford, R. M., and Mann, D. G. (1990) The
Diatoms, Cambridge Univ. Press, Cambridge UK
e
Buchsbaum, R., Buchsbaum, M., Pearse, J., and Pearse, V.
(1987) Animals Without Backbones, 3rd ed., Univ. Chicago Press,
Chicago
f
Epstein, E. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 1117
g
McNaughton, S. J., and Tarrants, J. L. (1983) Proc. Natl. Acad.
Sci. U.S.A. 80, 790791
h
Schwarz, K. (1970) in Trace Element Metabolism in Animals
(Mills, F., ed), pp. 2538, Livingstone, Edinburgh, UK
i
Schwarz, K., and Milne, D. B. (1972) Nature 239, 333334
j
Carlisle, E. M. (1972) Science 278, 619621
k
Hoekstra, W. H., Suttie, J. W., Ganther, H. E., and Mertz, W.,
eds. (1974) Trace Element Metabolism in Animals-2, University
Park Press, Baltimore, Maryland
l
Carlisle, E. M. (1988) Science Total Environment 73, 95106
m
Nielsen, F. H. (1991) FASEB J. 5, 26612667
n
Schwarz, K. (1973) Proc. Natl. Acad. Sci. U.S.A. 70, 16081612
o
Krger, N., Deutzmann, R., and Sumper, M. (1999) Science 286,
11291132
Silica skeleton of
a radiolarian
e
double-helical regions provide tie points for the
formation of gels (Fig. 4-10).
104,127,128
Sulfate groups protrude from the structure in
pairs and provide binding sites for calcium ions,
which stabilize the gel. The presence of occasional
extra sulfate groups in these polymers causes kinks
in the chains because the derivatized pyranose rings
reverse their conformation to the other chair form. This
prevents the entire polysaccharide chain from assuming
a regular helical structure.
100,104
Alginates, found in cell walls of some marine
algae and also formed by certain bacteria, consist in
part of a linear -1,4-linked polymer of D-mannuronate
with a cellulose-like structure. Alginates also contain
O
O
O
O
O
OSO
3
OH
OH
O
OSO
3
3
n
-Carrageenan:
[3Gal (4 sulfate) 14 (3,6-anhydro) Gal (2-sulfate) 1]
n

R
1
O
Si
O
R
2
R
1
O
Si
O
Si
O
R
2
OH OH HO HO OH HO
179
Meso-tartrate is joined to glucuronic acid in glycosidic
linkage and by acetal formation to the aldehyde gly-
oxylic acid, HOOCCHO, which is also joined in an
ether linkage to the next repeating unit.
130
Similar
open acetal linkages join monosaccharide units in
some bacterial lipopolysaccharides and may occur
more widely.
130a
Bacteria form and secrete a variety of heteropoly-
saccharides, several of which are of commercial value
because of their useful gelling properties. Xanthan
gum (formed by Xanthomonas campestris) has the basic
cellulose structure but every second glucose residue
carries an -1,3-linked trisaccharide consisting of 6-O-
acetylmannose, glucuronic acid, and mannose in the
following repeating unit:
131,132
The polymer is further modified by reaction of
about half of the mannosyl residues with pyru-
vate to form ketals (Eq. 4-9).
Acetan of Acetobacter xylinum has pentasac-
charide side chains that contain L-rhamnose.
122
A helical structure for the strands has been
observed by atomic force microscopy.
6. Polysaccharides of Bacterial
Surfaces
The complex structure of bacterial cell
walls is discussed in Chapter 8. However, it is
appropriate to mention a few bacterial poly-
saccharides here. The innermost layer of
bacterial cell walls is a porous network of a
highly crosslinked material known as pepti-
doglycan or murein (see Fig. 8-29). The back-
bone of the peptidoglycan is a -1,4-linked
-L-guluronate, sometimes in homopolymeric blocks
and sometimes alternating with the mannuronate
residues. Groups of adjacent guluronate units are
thought to impart calcium-binding properties to algi-
nates.
129
Polysaccharides with calcium-binding sites may
also serve to initiate deposition of calcium carbonate.
For example, the unicellular alga Pleurochrysis carterae
contains an unusual polysaccharide with the following
highly negatively charged repeating unit:
(4-9)
C. Polysaccharides (Glycans)
O
OH
COO
HO
O
C
C
O
C
O
3
2
1
H COO
COO H
COO H
n

[-4GlcA12-meso-tartrate-31-glyoxylate-1]
n
Figure 4-14 Tentative structure of the walls of suspension-cultured
sycamore cells. The wall components are in approximately proper
proportions but the distance between cellulose elementary fibrils is
expanded to allow room to present the interconnecting structure.
There are probably between 10 and 100 cellulose elementary fibrils
across a single primary cell wall. From Albersheim et al.
126
(-4Glc1-4Glc1-)
n
6-O-Acetyl-Man1
3
2
Man1-4GlcA1
O
CH
2
O
OOC
CH
3
O
O HO
O
H
COO
C
CH
3
Mannosyl
residue
O

(as PEP)
180 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
Figure 4-15 Structures of special sugars found in the
antigens of the outer cell walls of gram-negative bacteria.
alternating polymer of N-acetyl-D-glucosamine and N-
acetyl-D-muramic acid. Alternate units of the resulting
chitin-like molecule carry unusual peptides that are
attached to the lactyl groups of the N-acetyl-muramic
acid units and crosslink the polysaccharide chains. In
E. coli and other gram-negative bacteria the peptido-
glycan forms a thin (2 nm) continuous network around
the cell. This baglike molecule protects the organism
from osmotic stress. In addition, gram-negative bac-
teria have an outer membrane and on its outer surface
a complex lipopolysaccharide. The projecting ends of
the lipopolysaccharide molecules consist of long car-
bohydrate chains with repeating units that have anti-
genic properties and are called O antigens. Specific
antibodies can be prepared against these polysaccha-
rides, and so varied are the structures that 1000 differ-
ent serotypes of Salmonella are known. These are
classified into 17 principal groups. For example,
group E3 contains the following repeating unit, where
n may be ~50 on the average. Rha = L-rhamnose.
Polysaccharides of groups A, B, and D contain the
repeating unit
where X is a 3,6-dideoxyhexose: paratose in type A,
abequose in type B, and tyvelose in type D (Fig. 4-15).
The existence of the many serotypes depends on the
variety of components, on the many types of linkage
( and , 12, 13, 14, and 16) in the repeating
units, and on further structural variations at the chain
ends.
At the inner end of the O antigen is a shorter poly-
saccharide core whose structure is less varied than
that of the outer ends but which is remarkable in
containing two sugars found only in bacterial cell
walls: a seven-carbon heptose and an eight carbon
-oxo sugar acid, ketodeoxyoctonate (KDO). The
structures are given in Fig. 4-15 and the arrangement
of these sugars in the Salmonella lipopolysaccharide is
shown in Fig. 8-30. That figure also shows the manner
in which the oligosaccharide that bears the O antigen
is attached to a lipid anchoring group that is embedded
in the outer membrane of the bacteria.
A great variety of polysaccharides are present in
the outer layers of other types of bacteria. For example,
the mycobacteria have an alternating 5- and 6-linked
-D-Galf polymer attached to their peptidoglycan.
Attached to this galactan are branched penta-D-arabinose
units:
(Araf12raf1)
2
3,5Araf
These are further modified by esterification with my-
colic acids.
133
Information on some other cell wall
components is given in Chapter 20.
D. Glycoproteins and Proteoglycans
Many proteins, including almost all of those that
are secreted from cells and many that are components
of cell surfaces, carry covalently attached oligosaccha-
rides.
134139
These glycoproteins may carry just one
or a few, often highly branched, oligosaccharide chains.
For example, ribonuclease B has a structure identical
to that of ribonuclease A (Fig. 12-25) except for the
presence of an oligosaccharide on asparagine 34.
140
In other instances proteins carry a large number of
O
HO
OH
HO
CH
3
O
HO
OH
HO
CH
3
O
OH
HO
CH
3
HO
O
OH
HO
C
HO
HO
H
2
C
HO
OH
H
O
OH
C
HO
6
The enantiomorph of tyvelose is ascarylose,
found not only in certain bacterial antigens
but also in the eggs of .
3
L-Glycero-D-mannoheptose 3-Deoxy-D-mannooctulosonic acid,
also called 2-keto-3-deoxyoctonate
or KDO
COOH
7
HO
Paratose
(3,6-dideoxy-D-ribohexose)
H
Abequose
(3,6-dideoxy-D-xylohexose)
HO
CH
2
OH
Abequose is
acetylated here
in some antigens.
Tyvelose
(3,6-dideoxy-D-arabohexose)
The enantiomorph of abequose
is colitose, a sugar found in
surface antigens of E. coli.
Ascaris
Glc1
4
(Gal16Man14Rha)
n
X
(ManRhaGal)
n
181
such chains and the carbohydrate may account for
over half of the mass of a glycoprotein. Most carbohy-
drate chains are attached either as O-glycosides with
the hydroxyl groups of the side chains of serine, threo-
nine, or other hydroxyamino acid residues or as N-
glycosyl groups through linkage to the amide groups
of asparagine side chains. Both types of linkage may
be present in a single protein. Here are some examples.
Xyl1O-Ser(Thr) Proteoglycans of connective tissue;
thyroglobulins

NLys Some dermatan sulfates


H
Gal1O-Hydroxylysine Collagen, extension
(Hydroxyproline)
L-Ara1O-4-Hydroxy-proline Plants
GalNAc1O-Ser(Thr) Many glycoproteins
GlcNAc1O-Ser(Thr) Glycoproteins of cytoplasmic
surfaces
GlcNAc1NH-CH
2
-Asn Many glycoproteins
Some dermatan and heparan sulfates
Linkage of a glycosyl group to a carbon atom of an
indole ring of tryptophan has also been demonstrated.
141
1. O-Linked Glycosyl Groups
In the O-linked glycoproteins the sugar that is
attached directly to the protein is usually either xylose,
galactose, or N-acetylgalactosamine, all in the pyranose
ring form. Xylose is found only in the intercellular
proteoglycans which carry the chondroitin, dermatan,
and related sulfated polysaccharide chains of connec-
tive tissues.
141a,b
Since amino sugars are a major constit-
uent, proteoglycans are often called glycosaminoglycans.
Chondroitin, dermatan, and heparan sulfates are all
attached to core proteins by the same linkage, which
is illustrated here for chondroitin
Some IdoA may be present in the terminal unit of
dermatan sulfate.
142
Keratan sulfate has its own core
proteins
143
and has different terminal units including
the following:
107
The large proteoglycan of human cartilage is built
upon the 246 kDa protein aggrecan. In the central
half of the peptide chain are many Ser-Gly sequences
to which about one hundred 10- to 25-kDa chondroitin
sulfate chains are attached. About 30 keratan sulfate
chains as well as other oligosaccharide groups are also
present. These proteoglycan subunits are joined with
the aid of 44- to 49-kDa link protein to molecules of
hyaluronan
144146
(Fig. 4-16). Several types and sizes
of proteoglycan are known.
143,145147
Dermatan sulfates
may be linked to these through either serine or aspar-
agine, depending upon the tissue. The polysaccharide
chains of the proteoglycans also bind to collagen fibrils
to form a fiber-reinforced composite material between
cells. Chondroitin and heparan sulfates may be attached
at different Ser-Gly sites in a single peptide chain.
148
Degradation of heparan proteoglycans may lead to
the shorter free carbohydrate chains found in the
circulating heparin. Commercial heparin preparations
used as anticoagulants are produced by oxidative
destruction of the attached proteins.
113
A quite different situation holds for collagen in
which -galactosyl units and glucosyl--galactosyl
disaccharide units are attached to side chains of hydroxy-
lysine formed by postsynthetic modification of the
original procollagen chain.
A great deal of variation in the amount of glycosylation
is observed from one species to another. The human
-1(II) chains of collagen usually carry four disaccha-
rides and four monosaccharide units. In the related
collagen-like extensins, which are found in plant
cell walls, the hydroxyproline (Hyp) side chains are
O-glycosylated, largely by short oligosaccharides of
arabinose in furanose ring form,
150152
e.g., Araf 1
2Araf 12Araf 14Hyp. There are as many as 25
repeats of Ser-Pro-Pro-Pro-Pro encoded in an extensin
gene. Most of the prolines are hydroxylated and glyco-
sylated. The presence of two or more contiguous pro-
line residues seems to be the signal for the hydroxy-
lation reaction to take place.
152
Another distinct family of O-linked glycoproteins
are the mucins, which are present in saliva and other
D. Glycoproteins and Proteoglycans
Glc12Gal1O CH
(CH
2
)
2
CH
2
NH
3
C

Side chain of
erythro--hydroxylysine
+
Repeating unit of
chondroitin as in Fig. 4-11 Terminal unit
3Gal1
3Gal1
4Xyl1O-Ser
(4GlcA13GalNA1)
n
4GlcA1

KeratanGalGlcNAc
GalNAcOSer

SiaGal

182 Chapter 4. Sugars, Polysaccharides, and Glycoproteins


Figure 4-16 (A) Dark field electron micrograph of a proteo-
glycan aggregate from bovine articular cartilage (from bearing
surfaces of joints). Courtesy of Joseph A. Buckwalter. The
filamentous backbone consists of hyaluronic acid, as in (B).
The proteoglycan subunits extend from the backbone. From
Rosenberg.
149
A
B
mucous secretions. The polypeptides consist largely
of serine, threonine, proline, and glycine, with up to
one-third of the residues being Ser + Thr. Mucins
may contain 70 to 85% carbohydrate linked through
N-acetylgalactosamine residues to the serine and
threonine hydroxyls.
153160a
Salivary mucins of cows,
sheep, and dogs contain largely the following disac-
charide, which may contain either N-acetyl- or N-
glycolyl-neuraminic acid:
NeuNAc26GlcNAcSer(Thr)
Up to 800 disaccharides are present on a single large
protein which is composed of disulfide-linked subunits.
The many negatively charged sialic acid (N-glycolyl-
neuraminic acid) groups are thought to cause expan-
sion and rigidity which increase the viscosity of the
protein. Some other mucins contain predominantly
large oligosaccharides, which often carry blood group
determinants.
161,162
The ABO(H) family of blood group determinants
(Box 4-C) are oligosaccharide groups assembled from
D-galactose, N-acetyl-D-galactosamine, and L-fucose.
They are carried on the nonreducing ends of O-linked
oligosaccharides which may be attached to cell surface
proteins, to mucins, or to the sphingolipid known as
ceramide (Chapter 8). Attachment to proteins is usually
via GalNAc. An example of a blood type A determi-
nant on an O-linked oligosaccharide is the following,
where the linkage between the galactose and N-acetyl-
glucosamine may be either -1,3 or -1,4:
GalNAc13Gal13(4)GlcNAc13GalNAcO-Ser(Thr)
2

Fuc1
2. Asparagine-Linked Oligosaccharides
In many glycoproteins oligosaccharides are linked
through N-acetylglucosamine to the side chain of
asparagine.
134,163,164
This structure also illustrates one of the hydrogen-
bonding possibilities through which the sugar can
O
N
HOCH
2
N
O
HO
R
H
C
CH
3
O
H
CH
2
C

Amide N of
asparagine
0.5 m
183
interact with the protein. Since the site of glycosylation
is often at bends in the surface of the protein, the amide
groups of the N-acetylglucosamine may alternatively
hydrogen bond to amide groups of the peptide.
165
This asparagine linkage is very common. For example,
it is present in 97% of glycoproteins of blood plasma
166
and it is also predominent in the glycoproteins of
tissue surfaces.
167
There are numerous structures for
asparagine-linked oligosaccharides but many contain
the following core to which additional glycosyl groups
may be attached:
Here the Fuc indicates that this residue is present
only on some of the chains.
Notice the three mannose residues on the left side.
In the high mannose type oligosaccharide, Y
1
, Y
2
, and
Y
3
are additional mannose units. In many instances
Y
1
= Y
2
= Y
3
= ManMan. These extra mannose
units are put onto the oligosaccharide during the
original biosynthesis and before it is attached to the
protein (Chapter 20). Some of the mannose units may
then be removed during the processing of the oligo-
saccharide in the endoplasmic reticulum and residues
of glucosamine, galactose, and sialic acid (Sia) may be
added. Thus, Y
1
and Y
2
in the foregoing structure
often become
Sia23(6)Gal14GlcNAc1
Here, the sialic acid may be linked either 2,3 or 2,6 and
the GlcNAc either 1,2 or 1,3. Both Y groups may consist
of trisaccharides of this type in biantennary oligo-
saccharides and a third trisaccharide (Y
3
) may be added
to form a triantennary molecule. An additional
N-acetylglucosamine is often linked by -1,4 linkage
to the central mannose of the core and fucosyl residues
in -1,6 linkage are often linked to the N-acetylgluco-
samine next to the asparagine.
168
The Gal14GlcNAc
disaccharide unit in the above Y group is also called
N-acetyllactosamine because of its relationship to
lactose. It is often repeated in long Y groups, e.g., as
(3Gal14Glc-NAc1)
n
, the oligosaccharides
being called poly-N-acetyllactosamino-glycans. These
structures are also principal carriers of the ABO blood
type determinants on erythrocyte surfaces.
169
Many N-linked oligosaccharides are highly branched.
For example, in ovomucoid, a protease inhibitor of hen
eggs, pentaantennary oligosaccharides have two
and three N-acetylglucosamine rings, respectively,
attached to the terminal mannose units of the oligosac-
charide core in 1,2, 1,4 and 1,6 linkages. Another large
Figure 4-17 A large N-linked glycan from the
fertilized egg of the teleost fish oryzias.
171
On
the average, only two of the antenna contain
the repeated unit in parenthesis, i.e., p + q + r +
s + t = 1.9; an average of about 4.6 residues of
sialic acid are present.
NeuAc23Gal14Gal1 (4GlcNAc13Gal1)
q
4GlcNAc14 Man1
6
2
6
Man1
4
2
Man14GlcNA14GlcNA1Asn
3
Ser
Ala
Ala
Asp
Gln
Thr
Val
Ser
NeuAc23Gal14Gal1 (4GlcNAc13Gal1)
p
4GlcNAc1
3
NeuAc2

3
Fuc1

NeuAc23Gal1

4 3
Fuc1

3
NeuAc2

3
Fuc1

NeuAc23Gal1

4 3
Fuc1

NeuAc23Gal14Gal1 (4GlcNAc13Gal1)
r
4GlcNAc1
3
NeuAc2

3
Fuc1

NeuAc23Gal1

4 3
Fuc1

NeuAc23Gal14Gal1 (4GlcNAc13Gal1)
s
4GlcNAc1
3
NeuAc2

3
Fuc1

NeuAc23Gal1

4 3
Fuc1

NeuAc23Gal14Gal1 (4GlcNAc13Gal1)
t
4GlcNAc1
3
NeuAc2

3
Fuc1

NeuAc23Gal1

4 3
Fuc1

D. Glycoproteins and Proteoglycans


Man14GlcNA14GlcNA1Asn
Fuc1

6
3
Y
1
12Man1

Y
2
13Man1
6
Y
3
1

6
184 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
BOX 4-C THE BLOOD GROUP DETERMINANTS
The role of carbohydrates in biological commu-
nication is well illustrated by the human blood types.
a,b
According to the ABO system first described by
Landsteiner in 1900, individuals are classified into
types A, B, AB, and O. Blood of individuals of the
same type can be mixed without clumping of cells,
but serum from a type O individual contains anti-
bodies that agglutinate erythrocytes of persons of
types A and B. Serum of persons of type B causes
type A cells to clump and vice versa. Individuals of
none of the four types have antibodies against type
O erythrocytes. For this reason, persons with type
O blood are sometimes inaccurately described as
universal donors.
The ABO blood types are determined by specific
blood group determinants which are attached to
the nonreducing ends of O-linked oligosaccharides of
surface glycoproteins, mucins, glycolipids, and, to a
lesser extent, N-linked oligosaccharides. The blood
group determinants are found on erythrocytes and
all endothelial cells of the body. In about 80% of the
population they are also present on glycoproteins of
the saliva and other secretions.
The minimal determinant structures, attached
to carrier R, are as follows:
Here R refers to a carrier oligosaccharide which
can be as simple as
or may be a complex lactosaminoglycan or an oligo-
saccharide such as that in Fig. 4-17. The minimal
determinants can be linked to the carrier oligosac-
charide by either a -1,3 (type I chain) or -1,4 (type
II chain) glycosidic bond.
The genetic basis for the ABO blood groups is
well understood. There are three alleles, variants
of a gene, that encodes a glycosyltransferase. In
A type individuals, this enzyme transfers N-acetyl-
galactosamine from a carrier molecule, called UDP
GalNAc13 Gal2R
Fuc1

2
A determinant
Gal13 Gal1R
Fuc1

2
B determinant
Gal1R
Fuc1

2
H(O) determinant
Gal13GlcNAc1
Fuc1

2
Gal13(4) GlcNAc1
H
GDPFuc
GDP
FUT 3
Gal13GlcNAc1
Fuc1

4
Fuc1

2
Gal14GlcNAc1
Fuc1

3
Le
b
3-Fucosyl-N-acetyllactosamine
Le
x
or SSEA-1
GDPFuc
GDP
FUT 3

Gal13GlcNAc1
4
Fuc1
H-transferase
FUT 2
FUT 1
GDPFuc
GDP
FUT 3
Le
a
(Chapter 17), onto the terminal positions of the
H(O) determinant. The enzyme specified by the B
allele transfers galactose. The two enzymes differ
in only four amino acid residues but the result is an
altered substrate specificity.
a,c,d
The O allele produces
inactive enzyme as a result of a single base deletion
in the gene.
a
The H gene has been identified as that
of an -1,2 fucosyltransferase that transfers -L-fucose
from the carrier GDP to the galactose unit in the
foregoing structure.
e
Persons with an inactive H
transferase gene may have the rare type I, which
results from addition of glucosamine branches to
the repetitive H antigen structures of polylactos-
aminoglycans.
f
More often though, the H antigen
Gal13Gal1
Fuc1

2
Gal13(4) GlcNAc1
Fuc1

2
[3Gal14GlcNAc1]
n
GlcNAc1

3
Gal13(4) GlcNAc1
Precursor; type 1 has Gal-1,3,
type 2, Gal-1,4 linkage
GDPFuc
GDP
H-transferase
H
UDPGalNAc
UDP
A-transferase
GalNAc13Gal1
Fuc1

2
UDPGal
UDP
B-transferase
UDP-GlcNAc UDP
B A
I
3(4)GlcNAc13Gal14Glc1OSer (Thr, ceramide)
is acted upon by a different fucosyltransferase
encoded by the Le gene, which determines the
Lewis blood group. This enzyme places -1,4-
linked L-fucose onto the H antigen to give the Le
b
antigen. The same fucosyltransferase (which is
185
BOX 4-C Continued
designated FUT 3) acting on type 2 precursor chains
forms the Le
a
antigen. The same transferase adds a
-1,3-linked fucose to type 2 chains to give the Le
x
antigen, which is also called SSEA-1.
g,h
Lactosamine-
type precursors carrying -2,3-linked sialic acid at
their nonreducing ends on surfaces of granulocytes,
monocytes, and natural killer cells can be converted
to sialylated Le
x
(s-Le
x
).
i,j
Individuals with active gene Se (for secretion)
secrete glycoproteins bearing the blood group sub-
stance into saliva and other body fluids. The Se
gene is another H-transferase present in epithelial
cells and salivary glands. In individuals with active
Se genes (~80% of most populations) most soluble
H antigens are converted to Le
b
oligosaccharides.
k
There are at least 12 other well established
human blood groups, several of which involve
oligosaccharides attached to glycoproteins or gly-
colipids. The MN antigens consist of a sequence
of amino acids near the N terminus of the protein
glycophorin (Chapter 8) with attached sialic acid-
containing O-linked oligosaccharides,
l
e.g., as in the
following structure:
a
Yamamoto, F., Clausen, H., White, T., Marken, J., and
Hakomori, S. (1990) Nature 345, 229233
b
Frevert, J., and Ballou, C. E. (1985) Biochemistry 24, 753759
c
Feizi, T. (1990) Trends Biochem. Sci. 15, 330331
d
Yamamoto, F., and Hakomori, S. (1990) J. Biol. Chem. 265,
1925719262
e
Larsen, R. D., Ernst, L. K., Nair, R. P., and Lowe, J. B. (1990)
Proc. Natl. Acad. Sci. U.S.A. 87, 66746678
f
van den Eijnden, D., Koenderman, A., and Schiphorst, W.
(1988) J. Biol. Chem. 263, 1246112471
g
Mollicone, R., Reguigne, I., Kelly, R. J., Fletcher, A., Watt, J.,
Chatfield, S., Aziz, A., Cameron, H. S., Weston, B. W., Lowe, J.
B., and Oriol, R. (1994) J. Biol. Chem. 269, 2098720994
h
Nishihara, S., Narimatsu, H., Iwasaki, H., Yazawa, S.,
Akamatsu, S., Ando, T., Seno, T., and Ikuyo, N. (1994) J. Biol.
Chem. 269, 2927129278
i
Natsuka, S., Gersten, K. M., Zenita, K., Kannagi, R., and Lowe,
J. B. (1994) J. Biol. Chem. 269, 1678916794
j
Murray, B. W., Wittmann, V., Burkart, M. D., Hung, S.-C., and
Wong, C.-H. (1997) Biochemistry 36, 823 831
k
Kelly, R. J., Rouquier, S., Giorgi, D., Lennon, G. G., and Lowe, J.
B. (1995) J. Biol. Chem. 270, 46404649
l
Adamany, A., Blumenfeld, O., Sabo, B., and McCreary, J. (1983)
J. Biol. Chem. 258, 1153711545
m
Gillard, B., Blanchard, D., Bouhours, J.-F., Cartron, J.-P., van
Kuik, J. A., Kamerling, J., Vliegenthart, J., and Marcus, D. (1988)
Biochemistry 27, 46014606
n
Redman, C., Avellino, G., Pfeffer, S., Mukherjee, T., Nichols, M.,
Rubinstein, P., and Marsh, W. L. (1986) J. Biol. Chem. 261,
95219525
o
Bloy, C., Hermand, P., Blanchard, D., Cherif-Zahar, B.,
Goossens, D., and Cartron, J.-P. (1990) J. Biol. Chem. 265,
2148221487
The Cad antigen, also found on glycophorin as well
as on gangliosides, has an additional -1,4-linked
GalNAc at the indicated position.
m
The Kell blood
group antigens are carried on a 93 kDa glycoprotein
of erythrocyte surfaces.
n
The P blood group depends
upon surface carbohydrates such as the tetrasaccha-
ride part of the ganglioside called globoside; see
Fig. 20-9. Its characteristic antigenic activities are
destroyed by treatment with dilute periodate.
Other antigens, including those of the Rh and
LW groups, are represented by exposed parts of
proteins on the erythrocyte surfaces.
o
NeuAc23Gal14GlcNAc
Fuc1

2
s-Le
x
NeuAc23Gal13GalNAcOSer (Thr)
GalNAc1

4
6
NeuAc2

in Cad antigen }
N-linked glycan is pictured in Fig. 4-17. Like many
others, it has a number of sialic acid residues at the
nonreducing ends and also contains N-acetyllactos-
amine units. The major component of the cell walls of
yeast (S. cerevisiae) is a mannoprotein that carries long
N-linked oligosaccharides with highly branched outer
chains of over 100 mannose residues
170
(see also Section
C,3).
The special importance of sialic acids in glycopro-
teins may lie in the negative electrical charges and the
resultant Ca
2+
-binding properties that they impart to
cell surfaces. Some glycoproteins carry sulfate or
phosphate groups that have similar effects. The glyco-
proteins of the slime mold Dictyostelium contain both
mannose-6-P and mannose-6-sulfate residues.
163
Oligosaccharides on the protein subunits of flagella
of halobacteria contain sulfate esters of glucuronic
acid.
172
Some N-linked oligosaccharides carry chains
of keratan sulfate in place of the sialic acid in the Y
1
and Y
2
groups of the oligosaccharide.
173
D. Glycoproteins and Proteoglycans
186 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
3. Glycoproteins in Biological Recognition
The clusters of sugar rings that form the oligosac-
charides on glycoproteins play a vital role in many
aspects of biological recognition.
139,174176
A good
example is provided by the human blood groups
whose characteristics are determined largely by oligo-
saccharides (Box 4-C). The adhesion of viruses, bacteria,
and eukaryotic parasites to cell surfaces and of one cell
to another in multicellular organisms is also dependent
on carbohydrates. Recently, it has become clear that
the oligosaccharides of cell surfaces change during
growth and development and provide an important
mechanism by which cells can recognize each other
and respond. Why are carbohydrates used for this
purpose? It has been pointed out by Sharon and Lis
175
that four different nucleotides can make only 24 distinct
tetranucleotides but that four different monosaccharides
can make 35,560 unique tetrasaccharides.
Lectins and other carbohydrate-binding pro-
teins. Much of biological carbohydrate-dependent
recognition is a result of interaction of individual
glycosyl groups or of oligosaccharides on a glycopro-
tein with a second protein. In some cases that protein
is referred to as a receptor; in other cases the glycosyl
groups may be called the receptor. Carbohydrate-
binding proteins include antibodies, enzymes, and
carriers that help sugars to cross cell membranes.
In addition, there is a large group of carbohydrate
binding proteins called lectins (from the latin lectus;
to select).
24,174,177,178
The first lectins discovered were proteins of plant
seeds with specific sugar-binding properties and the
ability to agglutinate erythrocytes. Ricin, a very toxic
protein from castor beans, was isolated in 1888.
178,179
Perhaps the best known lectin is concanavalin A, a
protein crystallized by Sumner in 1919.
180
Concana-
valin A makes up 23% of the protein of the jack bean.
It is one of a family of legume lectins that resemble
favin, whose structure is shown in Fig. 2-15.
177,181,182
Many lectins, including ricin, have quite different
three-dimensional structures but share the common
characteristic of having a selective binding site for one
or more glycosyl rings. Concanavalin A binds to -D-
mannopyranose or -D-glucopyranose with unmodi-
fied hydroxyl groups at C-3, C-4, and C-6.
183,184
Tighter binding is observed if additional mannose
residues are present in an oligosaccharide.
178,183
The
protein also has specific binding sites for Ca
2+
and for
a transition metal ion such as Mn
2+
. Soybean lectin
binds D-N-acetylgalactosamine and D-galactose units,
while wheat lectin is specific for D-N-acetylglucosamine.
Animal cells also produce lectins.
24,105,185193
The amebas of the cellular slime mold Dictyostelium
synthesize a classical lectin called discoidin I that
binds GalNAc or Gal. It is absent from cells until they
are ready to differentiate into an aggregating form
(Chapter 1). Then it is produced in abundance.
178
Discoidin I has a second binding site specific for the
peptide sequence Arg-Gly-Asp (RGD) which is known
to be involved in cell adhesion and which binds to
such surface proteins as fibronectin and laminin
(Chapter 8). Many animal tissues contain soluble
lactose-binding lectins known as galectins or S-Lac
lectins. The best known member is a dimer of 14-kDa
subunits;
187,188
many other related lectins have been
found.
189,192
Another family are Ca
2+
-dependent or
C-type lectins which are specific for mannose, L-fucose,
or other sugars.
105,193
Lectin domains are being
discovered in many proteins.
Carbohydrate-binding sites. The structures of
the sites that recognize and bind sugar rings in lectins,
enzymes, transporter proteins, and other carbohydrate-
binding proteins vary greatly, as does the tightness of
binding. However, there are certain common features:
Sugar rings are bound by hydrogen bonds, which are
often numerous. An example is the galactose chemo-
receptor protein from E. coli. It binds both and
anomers of either D-glucose or D-galactose and is
utilized by the bacteria in searching for food (see
Chapter 19). The structure of D-glucose bound to this
protein is shown in Fig. 4-18. Notice the many hydrogen
bonds. Two of the OH groups have the maximum of
three hydrogen bonds apiece. There are three negatively
charged aspartate side chains and one positively charged
guanidinium group. These provide strong iondipole
interactions which add strength to the bonds. The
presence of ionized groups in varying numbers and
constellations is another common feature of protein
carbohydrate interactions. A third common feature is
the presence of aromatic rings, which often lie against
one face of the sugar. The stereoscopic drawing of
Fig. 4-18A shows an indole ring of a tryptophan residue
in front and a phenylalanine side chain behind the
sugar.
194
Sugars bind to lectins,
181,184,195
to enzymes
(Chapter 12), and to antibodies
196
through similar
interactions.
Binding of viruses and bacteria to cells. The
cholera toxin and a related toxic protein from E. coli
bind to SiaGal groups attached to glycolipids (gan-
gliosides, Chapter 8) of erythrocytes and other cells.
197
The influenza virus gains access to our body cells by
first binding through a viral surface hemagglutinin.
This is a protein that binds specifically to NeuAc2
6Gal or NeuAc23Gal of cell surface oligosaccha-
rides.
174,175,198
Removal of sialic acid from erythrocyte
surfaces abolishes the ability of the influenza and some
other viruses to bind. It seems somewhat surprising
that a second surface protein on the influenza virus
is a neuraminidase (or sialidase) which catalyzes
the removal of sialic acid from cell surface proteins,
187 D. Glycoproteins and Proteoglycans
O
O
O
O
O
O
D14
H
O
H
H
H H
N
O
O
H
O
O
H
N
H
H
N
R156
H
N
H
+
N236
D154
O
O
O
H152
N91
6
4
H
H
N
N
N
H
H
D236
N211

H
H
N
H
H
O
Figure 4-18 (A) Stereoscopic view of the interactions between the E. coli
galactose chemoreceptor protein and a bound molecule of D-glucose. (B)
Schematic drawing showing many of these interactions and the state of ioniza-
tion deduced for the aspartate and arginine side chains. From Vyas et al.
194
A
B
destroying unoccupied virus receptors.
199,200
This may
facilitate movement of a virus particle through the
mucin layer surrounding a cell. Bacteria and other
invading parasites also produce neuraminidases.
201
Trypanosamas cruzi, the causative agent of chagas
disease, employs a transsialidase to transfer sialic
acid from a Gal on the host cell onto a protein on the
parasite surface. This is essential for successful inva-
sion of the host.
202
The adherence of cells of E. coli to mannosyl units
of cell surface proteins may initiate the infections that
sometimes occur with this bacterium.
174,203
However,
cells of E. coli from strains that cause urinary infections
bind to Gal14Gal on glycolipids that carry the
blood group P antigens (Box 4C).
174,204
Neuropatho-
genic strains of E. coli or of Neisseria meningitidis,
which may cause neonatal meningitis, bind to
-2,8-polysialic acid chains on nerve cells.
135,205,206
Helicobacter pylori, the stomach ulcer
bacterium, binds to the human
Lewis
b
blood group antigen (Box
4-C).
207
Entamoeba histolytica, which
causes amebic dystentery, binds
to Gal and GalNAc-containing
oligosaccharides such as
GalNAc13Gal.
208
Aggregation and adherence
of cells. Differences among cell
surfaces are fundamental to the
formation of multicellular organisms
and to many physiological processes.
Proteins, carbohydrates, and lipids
all contribute material to exposed
cell surfaces. The adhesion of one
cell to another is mediated by a group
of adhesion proteins together with
oligosaccharide groups and some-
times polysaccharides. Families of
adhesion proteins include integrins
and cadhedrins (discussed in
Chapter 8), various members of
the immunoglobulin superfamily,
the cell differentiation antigens,
(often designated CD44, etc.), the
C-type lectins known as selectins,
and proteoglycans.
Here are two of many known
examples of specific cell cell adhe-
sion. The species-specific reaggrega-
tion of dissociated cells of marine
sponges (Chapter 1) depends upon
a 20-kDa proteoglycan of unique
structure
209211
together with a cell
surface receptor protein and calcium
ions. The recognition of egg cell
surfaces by sperm
212214
is species
specific and depends upon interaction of sperm recep-
tors with O-linked oligosaccharides of the extracellular
coat of the ovum.
Growth and differentiation. The exact structures
of the oligosaccharides and polysaccharides of cell
surfaces vary not only with cell and tissue type but
also with the position of a cell and with time. Actions
of numerous glycosyl transferases alter these saccha-
ride groups as an organism grows and develops.
Other enzymes alter them by hydrolytic removal of
sugars, by isomerizaion, oxidation, and addition of
other components such as phospho, sulfo, and acetal
groups. For example, the presence of the H-antigen
determinant, whose structure is shown in Box 4-C,
is strictly regulated, both temporally and spatially,
during vertebrate development.
215,216
The relative
amounts of the H determinant vs Le
x
, sLe
x
(Box 4-C)
188 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
HO
O
O
O O
O
O
HO
HO
O
S
O
O
O
N
O
H
3
C
N
O
HO
O O
HO
OH
HO
O
H
3
C
HO
O
CH
3
HO
OH
Fuc
6'-Sulfate
GlcNAc
Gal
NeuAc
1
1
4
3
1
3
2

H
3'Sialyl6'SulfoLe
x
determinant

(4-10)
C C
H
CH
2
O
N O
H
Oligosaccharide
C C
CH
2
O
N
Glycosidic
linkage
+ Oligosaccharide
H
H
HO

+
and other surface groupings is controlled by fucosyl-
transferases, sialyltransferases, etc. Human cancers
often accumulate large amounts of fucosylated glyco-
proteins and glycolipids carrying Le
a
, Le
x
, and sialyl-Le
x
antigens
175,216,217
and sialomucins.
218
Glycoproteins
help to control many metabolic processes. For example,
a protein calnexin (a chaperonin, Chapter 10) helps
glycoproteins to fold correctly.
219
On the other hand,
removal of terminal sialic acid residues from blood
plasma proteins leads to rapid removal of the proteins
from circulation and to catabolism by liver cells.
175
This
process depends upon a receptor protein specific for
oligosaccharides with terminal galactosyl residues
(Chapter 20).
Recognition and adhesion by leukocytes. A
group of three calcium-dependent lectins known as
selectins bind the sialyl Lewis x (sLe
x
) antigen and
play important roles in adhesion to cells of the vascular
endothelium and leukocytes
175,220224
and also to
platelets. Although all of the selectins bind the sLe
x
antigen, the binding is weak and these multidomain
proteins may simultaneously bind to other ligands
such as heparan sulfate.
225
The leukocyte L-selectin
(CD62L) binds very tightly to the 3'-sialyl-6'-sulfo-Le
x
determinant
220,226
which occurs on mucin-like glyco-
proteins. The interaction with P selectin helps leuko-
cytes to bind to surfaces on endothelial cells in lymph
nodes and sites of chronic or acute inflammation. In a
similar way E selectin is synthesized in vascular endo-
thelial cells that have been transiently activated by
cytokines in response to injury and other inflammatory
stimuli. The E selectin binds the sulfated sLe
x
antigen
on surfaces of neutrophils, monocytes, eosinophils,
and basophils.
220,222,227230
P selectin is stored in secre-
tory granules of platelets and endothelial cells and
is released to the cell membrane upon activation by
thrombin.
220
E. Some Special Methods
Small monosaccharides and oligosaccharides can
be separated readily from polymeric constituents and
can be purified further by chromatographic procedures
including gel filtration as is illustrated in Fig. 3-3.
However, polysaccharides and complex oligosaccha-
rides are harder to purify. A few of these, such as
cellulose and glycogen, are sufficiently stable that
other materials can be dissolved away from them by
prolonged boiling in strongly basic solutions. Complex
carbohydrates are usually cut into smaller oligosaccha-
rides or glycopeptides. These may be separated by
HPLC, capillary electrophoresis, or thin-layer chroma-
tography
134,231235
(Fig. 4-19) or by chromatography on
immobilized lectins.
234
Quantities of less than 25 pico-
moles can be separated by use of mass spectrometry with
liquid chromatography.
236,237
High resolution Fourier
transform mass spectroscopy is very useful in the study
of posttranslational glycosylation of proteins.
237a,b
1. Release of Oligosaccharides from
Glycoproteins
The O-linked oligosaccharides of glycoproteins or
glycolipids can be split off from the proteins by
elimination (see also Chapter 13):
189
1 ascent
Glc
Mal (G2)
G3
G4
G5
G6
G7
G8
G9
G10
G11
G12
G13
G14
Solvent: MeCN/EtOAc/PrOH-1/H
2
O
85 20 50 50
2 ascents 3 ascents 4 ascents
O
NH
RO
HOH
2
C
N HO C
O
CH
2
Peptide
Ac
H
2
N NH
2
O
NH
RO
HOH
2
C
NH
2
HO
Ac
H
2
N NH C
O
CH
2
Peptide
O
NH
RO
HOH
2
C
N HO
+
a
H
Oligosaccharide
b
Ac
Acetic anhydride
Ac
H
(4-11)
Figure 4-19 Ascending thin-layer chromatography of a
mixture of maltooligosaccharides obtained by the hydrolysis
of linear starch. G2 (maltose), G3 (maltotriose), G4 (malto-
tetraose), G5 (maltopentaose) G14 represent oligosaccha-
rides with the indicated number of glucose residues, all in
-1,4 linkage. In the multiple ascent technique the chroma-
tographic solvent, whose composition (by volume) is indicated,
is allowed to ascend the thin-layer plate repeatedly with the
plate allowed to dry between ascents. The separation of the
higher oligosaccharides is distinctly improved by a larger
number of ascents. Photograph courtesy of John Robyt.
E. Some Special Methods
Treatment with 0.10.5 M NaOH for several hours
will completely liberate the oligosaccharides, whose
released carbonyl groups may then be reduced with
NaBH
4
, NaB
2
H
4
, or
3
H-labeled borohydride to form
stable sugar alcohols (Eq. 4-2).
238
Asparagine-linked
oligosaccharides are often recovered as glycopeptides
prepared by complete proteolytic digestion of the
denatured glycoprotein. These can be separated by
high voltage electrophoresis in borate buffers.
239
The
oligosaccharides can be released from the glycopep-
tides by enzymes such as endo-N-acetylglucosamini-
dase
240
or glycopeptidyl amylase of almonds.
241,242
These release the oligosaccharide as a 1-amino deriva-
tive.
When a dry glycoprotein is heated at 105 with
anhydrous hydrazine for 812 h all of the asparagine-
linked oligosaccharide chains are released (Eq. 4-
11).
239,243245
In addition to the glycosylamine product
of step a, the corresponding 1OH and NHNH
2
compounds are also formed. These are all converted
to the stable acetyl derivative (step b), after which the
oligosaccharides may be characterized by mass spec-
trometry and high-field NMR spectoscopy.
234,246248
Crystallization of oligosaccharides in complex mixtures
is difficult to impossible.
2. Hydrolysis
Most glycosidic linkages are hydrolyzed readily
by heating with 1 N mineral acids. The mechanism of
the hydrolytic reaction is similar to that employed by
the enzyme lysozyme (Chapter 12). Some linkages are
unusually sensitive to acid and a few are very resistant.
Thus, a variety of conditions may be applied for partial
acid hydrolysis as an aid to characterization.
249
Aceto-
lysis, cleavage by acetic anhydride, is also of value.
250
A battery of hydrolytic enzymes specific for sugars
that are joined in a given type of glycosidic linkage
are available.
251
These are useful in determining
sequences of oligosaccharides released from glyco-
proteins. Radioactive tracer techniques can also be
applied.
252
3. Methylation
An important general method in characterization
of carbohydrates is the classical exhaustive methyl-
ation (permethylation). Repeated treatment with a
methylating agent such as methyl iodide converts all
free OH groups to OCH
3
groups. Then, complete acid
190 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
Figure 4-20 Separation of a mixture of O-methylated
glucoses by ascending thin-layer chromatography. Whatman
K6 TLC plates were used with two ascents of the solvent
acetonitrile/chloroform/methanol in the ratio 3/9/2, V/
V/V. Courtesy of John Robyt.
(4-12)
2,3,4,6-Me
4
-Glc
2,3-Me
2
-Glc
2,4-Me
2
-Glc
3,4-Me
2
-Glc
2,3,4,6-Me
4
-Glc
2,3,6-Me
3
-Glc
2,4,6-Me
3
-Glc
2,3,4-Me
3
-Glc
2,3-Me
2
-Glc
2,4-Me
2
-Glc
3,4-Me
2
-Glc
1 2 3
2,3,6-Me
3
-Glc
2,4,6-Me
3
-Glc
2,3,4-Me
3
-Glc
O
OH
OR'
CH
2
RO
HO
OH
CH
HC
O
CH
2
RO
OH
O
O
HIO
4
OR'
hydrolysis, followed by separation of the methylated
sugars and their quantitative determination, reveals
the relative amounts of end units (containing four
methoxyl groups), straight chain units (containing
three methoxyl groups), and branch points (contain-
ing two methoxyl groups). In addition, the structure
of the methylated derivatives provides information on
the positions of the linkages in the sugar rings.
After a methylated polysaccharide is subjected to
partial hydrolysis, the newly exposed hydroxyl groups
(or those created by borohydride reduction of carbonyl
groups) can be labeled by ethylation or propylation.
One procedure for sequencing complex carbohydrates
makes use of high-resolution reversed-phase liquid
chromatography to separate the many alkylated oligo-
saccharides produced by methylation followed by
partial acid hydrolysis, reduction, and ethylation.
233
Reductive cleavage of the glycosidic linkages in methy-
lated polysaccharides allows uniquivocal determina-
tion of ring size.
253
Branch points may be located by
methanolysis of the permethylated polysaccharide
followed by conversion of free OH groups to p-bromo-
benzoate esters. The latter are separated and the
circular dichroism (Chapter 23) is measured. Mass
spectrometry has also been applied successfully.
246
A simple procedure that can be conducted in any
laboratory using thin-layer chromatography is illus-
trated in Fig. 4-20.
254
4. Periodate Oxidation (Smith Degradation)
One of the most important reagents in investiga-
tions of carbohydrate structure is periodic acid (or
sodium periodate).
255
This reagent oxidatively cleaves
CC bonds bearing adjacent OH or NH
2
groups to
form dialdehydes (Eq. 4-12). The method is quanti-
tative. After some hours of reaction, excess periodate
not consumed in the oxidation can be determined.
If three consecutive carbon atoms bear hydroxyl or
amino groups, formic acid is liberated from the central
atom and can also be measured quantitatively. After
destruction of excess periodate the dialdehyde can be
reduced by addition of solid sodium borohydride to
form stable CH
2
OH groups. Following mild acid
hydrolysis to split the acyclic acetal linkages, the frag-
ments can be separated and identified. The sequence
of reactions is known as the Smith degradation. If
sodium borotritide (NaB
3
H
4
) is used for the reduction
the fragments will be radioactive and can be located
on chromatograms by fluorography. Periodate oxi-
dation can also be used to alter all surface oligosaccha-
rides.
99
Removal of O-linked oligosaccharides from
glycoproteins can be accomplished by periodate
oxidation and alkaline elimination from the dialde-
hyde product of Eq. 4-12 if OR is part of a serine or
other hydroxyamino acid side chain in a protein.
256
5. Nuclear Magnetic Resonance
As in other areas of biochemistry, NMR has
become an extremely important tool in carbohydrate
research. The mixtures of anomers of various ring
191
BOX 4-D ANTIFREEZE AND ICE-NUCLEATION PROTEINS
E. Some Special Methods
Fish living in Arctic and Antarctic waters may
encounter temperatures as low as 1.9C. The
freezing point depression provided by dissolved
salts and proteins in the blood is insufficient to pro-
tect the fish from freezing. As winter approaches,
they synthesize and accumulate in their blood
serum a series of eight or more special antifreeze
proteins.
a d
One type of antifreeze glycoprotein
from winter flounder contains the following unit
repeated 17 50 times.
Destruction of the galactosyl residues by oxidation
with periodate, acetylation of the free hydroxyl
groups of the oligosaccharides, or their removal by
elimination all lead to loss of antifreeze activity.
The same fish contain a second series of alanine-
rich antifreeze polypeptides that are not glycosylated
but which exist as amphipathic helices. One of these
(Type I) contains ~40 residues in a single helix.
e i
A
third family of antifreeze proteins (Type II), found
in the sea raven are globular proteins, rich in cysteine
and structures. They are members of the lectin
family.
d,j
A fourth type (Type III) found in the sea pout
and some other fishes are 62- to 66-residue globular
proteins containing an orthogonal sandwich struc-
ture.
k,l
Messenger RNA molecules coding for the
antifreeze proteins are found in the livers of flounder
in the winter but are absent in the summer.
m
Antifreeze proteins, that are 3 4 times as effec-
tive as those in fish, have been isolated from some
insects and other arthropods.
m,n,o
They help beetle
larvae to overwinter.
m
The insect proteins have a
parallel helix structure resembling that in Fig. 2-17
and stabilized by S S bridges.
o,p
Some plants also
synthesize antifreeze proteins.
n,q,r
One of these,
isolated from carrots, is a member of the leucine-
rich-repeat family.
q
How do antifreeze proteins work? The major
effect is to greatly slow the freezing rather than to
decrease the freezing point. The proteins apparently
accomplish this by binding to the surfaces of small
ice crystals and preventing their growth.
d,f,h,i,k,l,s,t
This provides the fish with enough time for the
blood to pass back into the liver, in which a high
enough temperature is maintained to melt any
microcrystals before the blood again circulates
through the colder tissues. Some of the proteins
have clusters of polar side chains that bind to
specific faces of the ice crystals and inhibit growth.
s
(AlaAlaThr)
n
O GalNAc1
3
Gal1

A few fishes tolerate a high internal osmotic


pressure and accumulate glycerol in their blood up
to a concentration of 0.4 M.
t
Insects may accumulate
up to 3 M glycerol and some species utilize various
other cryoprotectants, such as mannitol, sorbitol,
erythritol, threitol, trehalose, glucose, fructose, proline,
and alanine.
u
Some amphibians and reptiles can
survive freezing and recover fully. For the most
studied wood frog, rapid freezing is fatal, but slow
freezing leaves the frog, whose heart ceases to func-
tion, with a 200-fold increased glucose concentration
and a decreased water content in its organs. It resumes
normal activities within 14 24 h of thawing.
t
Having an effect opposite to that of the anti-
freeze proteins are surface proteins of some bacteria
of the genera Pseudomonas, Erwinia, and Xanthomonas.
These proteins provide nuclei for growth of ice
crystals from supercooled water.
v,w
a
Feeney, R. E., Burcham, T. S., and Yeh, Y. (1986) Ann. Rev.
Biophys. Biophys. Chem. 15, 5978
b
Eastman, J. T., and DeVries, A. L. (1986) Sci. Am. 255 (Nov), 106114
c
Davies, P. L., and Hew, C. L. (1990) FASEB J. 4, 24602468
d
Gronwald, W., Loewen, M. C., Lix, B., Daugulis, A. J., Snnichs-
en, F. D., Davies, P. L., and Sykes, B. D. (1998) Biochemistry 37,
47124721
e
Chakrabartty, A., Ananthanarayanan, V. S., and Hew, C. L. (1989)
J. Biol. Chem. 264, 1130711312
f
Yang, D. S. C., Sax, M., Chakrabartty, A., and Hew, C. L. (1988)
Nature (London) 333, 232237
g
Madura, J. D., Wierzbicki, A., Harrington, J. P., Maughon, R. H.,
Raymond, J. A., and Sikes, C. S. (1994) J. Am. Chem. Soc. 116,
417418
h
Sicheri, F., and Yang, D. S. C. (1995) Nature (London) 375, 427431
i
Wierzbicki, A., Taylor, M. S., Knight, C. A., Madura, J. D.,
Harrington, J. P., and Sikes, C. S. (1996) Biophys. J. 71, 818
j
Ng, N. F. L., and Hew, C. L. (1992) J. Biol. Chem. 267, 1606916075
k
Chao, H., Snnichsen, F. D., DeLuca, C. I., Sykes, B. D., and
Davies, P. L. (1994) Protein Sci. 3, 17601769
l
Jia, Z., DeLuca, C. I., Chao, H., and Davies, P. L. (1996) Nature
(London) 384, 285288
m
Gourlie, B., Lin, Y., Price, J., DeVries, A. L., Powers, D., and
Huang, R. C. C. (1984) J. Biol. Chem. 259, 1496014965
n
Li, N., Chibber, B. A. K., Castellino, F. J., and Duman, J. G.
(1998) Biochemistry 37, 63436350
o
Graether, S. P., Kuiper, M. J., Gagn, S. M., Walker, V. K., Jia, Z.,
Sykes, B. D., and Davies, P. L. (2000) Nature (London) 406, 325 328
p
Liou, Y.-C., Tocilj, A., Davies, P. L., and Jia, Z. (2000) Nature
(London) 406, 322324
q
Worrall, D., Elias, L., Ashford, D., Smallwood, M., Sidebottom,
C., Lillford, P., Telford, J., Holt, C., and Bowles, D. (1998) Science
282, 115117
r
Sidebottom, C., Buckley, S., Pudney, P., Twigg, S., Jarman, C.,
Holt, C., Telford, J., McArthur, A., Worrall, D., Hubbard, R., and
Lillford, P. (2000) Nature (London) 406, 256
s
Knight, C. A., (2000) Nature (London) 406, 249251
t
Costanzo, J. P., Lee, R. E., Jr., DeVries, A. L., Wang, T., and Layne,
J. R., Jr. (1995) FASEB J. 9, 351352
u
Storey, K. B., and Storey, J. M. (1990) Sci. Am. 263 (Dec), 9297
v
Wolber, P., and Warren, G. (1989) Trends Biochem. Sci. 14, 179182
w
Gurian-Sherman, D., and Lindow, S. E. (1993) FASEB J. 7, 1338 1343
192 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
1. Pigman, W. W., and Horton, D., eds. (1972)
The Chemistry of Carbohydrates, 2nd ed.,
Academic Press, New York
2. Guthrie, R. D. (1974) Guthrie and Honeymans
Introduction to Carbohydrate Chemistry, 4th ed.,
Oxford Univ. Press (Clarendon), London and
New York (p. 17)
3. Binkley, R. W. (1988) Modern Carbohydrate
Chemistry, Dekker, New York
4. Shalenberger, R. S. (1982) Advanced Sugar
Chemistry: Principles of Sugar Stereochemistry,
AVI Publ. Co., Westport, Connecticut
5. El-Khadem, H. S. (1988) Carbohydrate
Chemistry, Academic Press, San Diego,
California
6. Prince, R. C., Gunson, D. E., Leigh, J. S., and
McDonald, G. G. (1982) Trends Biochem. Sci. 7,
239240
7. Wertz, P. W., Garver, J. C., and Anderson, L.
(1981) J. Am. Chem. Soc. 103, 39163922
8. Ha, S., Gao, J., Tidor, B., Brady, J. W., and
Karplus, M. (1991) J. Am. Chem. Soc. 113,
15531557
9. Angyal, S. J. (1979) in Asymmetry of Carbo-
hydrates (Harmon, R. E., ed), p. 15, Decker,
New York
10. Gray, G. R. (1976) Acc. Chem. Res. 9, 418
11. Pierce, J., Serianni, A. S., and Barker, R. (1985)
J. Am. Chem. Soc. 107, 24482456
12. Maple, S. R., and Allerhand, A. (1987) J. Am.
Chem. Soc. 109, 31683169
13. Goux, W. J. (1985) J. Am. Chem. Soc. 107, 4320
4327
14. Barker, S. A., and Baggett, N. (1978) Trends
Biochem. Sci. 3, 140141
15. Benson, A. A. (1963) Adv. Lipid Res. 1, 387394
References
16. Benning, C., Beatty, J. T., Prince, R. C., and
Somerville, C. R. (1993) Proc. Natl. Acad. Sci.
U.S.A. 90, 15611565
17. Dijkema, C., Kester, H. C. M., and Visser, J.
(1985) Proc. Natl. Acad. Sci. U.S.A. 82, 1418
18. Tarczynski, M. C., Jensen, R. G., and
Bohnert, H. J. (1992) Proc. Natl. Acad. Sci.
U.S.A. 89, 26002604
19. Yamanouchi, T., Tachibana, Y., Sekino, N.,
Akanuma, H., Akaoka, I., and Miyashita, H.
(1994) J. Biol. Chem. 269, 96649668
20. Nadano, D., Iwasaki, M., Endo, S., Kitajima,
K., Inoue, S., and Inoue, Y. (1986) J. Biol.
Chem. 261, 1155011557
21. Schauer, R. (1985) Trends Biochem. Sci. 10,
357360
22. Faillard, H. (1989) Trends Biochem. Sci. 14,
237241
23. Roth, J., Kempf, A., Reuter, G., Schauer, R.,
and Gehring, W. J. (1992) Science 256, 673
675
24. Powell, L. D., and Varki, A. (1995) J. Biol.
Chem. 270, 1424314246
25. Stoddart, J. F. (1971) Stereochemistry of
Carbohydrates, Wiley-Interscience, New York
26. Brady, J. W. (1986) J. Am. Chem. Soc. 108,
81528160
26a. Marszalek, P. E., Pang, Y.-P., Li, H., Yazal, J.
E., Oberhauser, A. F., and Fernandez, J. M.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 7894
7898
27. Venkataraman, G., Sasisekharan, V., Cooney,
C. L., Langer, R., and Sasisekharan, R. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 61716175
28. Hajduk, P. J., Horita, D. A., and Lerner, L. E.
(1993) J. Am. Chem. Soc. 115, 91969201
Figure 4-21 Stereoscopic view of an
N-linked oligosaccharide whose structure
has been deduced by two-dimensional
NMR spectroscopy and energy calcula-
tions. This is one of a range of allowed
conformations. From Homans et al.
257
29. Wolfe, S., Whangbo, M.-H., and Mitchell, D. J.
(1979) Carbohydrate Res. 69, 126
30. Kirby, A. J. (1983) The Anomeric Effect and
Related Stereoelectronic Effects at Oxygen,
Springer, Berlin
31. Perrin, C. L., Armstrong, K. B., and Fabian, M.
A. (1994) J. Am. Chem. Soc. 116, 715722
32. Cui, W., Li, F., and Allinger, N. L. (1993) J. Am.
Chem. Soc. 115, 29432951
33. Westhof, E., and Sundaralingam, M. (1980) J.
Am. Chem. Soc. 102, 14931500
33a. French, A. D., Dowd, M. K., and Reilly, P. J.
(1997) J. Mol. Struct. (Theochem), 395 396
34. Gabb, H. A., and Harvey, S. C. (1993) J. Am.
Chem. Soc. 115, 42184227
35. Plavec, J., Tong, W., and Chattopadhyaya, J.
(1993) J. Am. Chem. Soc. 115, 97349746
36. Ellervik, U., and Magnusson, G. (1994)
J. Am. Chem. Soc. 116, 23402347
36a. French, A. D., and Murphy, V. G. (1973)
Carbohydr. Res. 27, 391 406
37. Robyt, J. F., and White, B. J. (1996) Biochemical
Techniques, Theory and Practice, 2nd ed.,
Waveland Press, Prospect Heights, Illinois
38. Liptk, A., Fgedi, P., Szurmai, Z., and
Harangi, J. (1990) CRC Handbook of Oligo-
saccharides. Vol. I: Disaccharides, CRC Press,
Boca Raton, Florida
39. Cerami, A. (1986) Trends Biochem. Sci. 11,
311314
40. Watkins, N. G., Neglia-Fisher, C. I., Dyer, D.
G., Thorpe, S. R., and Baynes, J. W. (1987)
J. Biol. Chem. 262, 72077212
41. Sarez, G., Rajaram, R., Oronsky, A. L., and
Gawinowicz, M. A. (1989) J. Biol. Chem. 264,
36743679
sizes (Eq. 4-1) can be analyzed with proton or
13
C
NMR.
10,257
A variety of newer NMR techniques, some
of which have been described in Chapter 3, have been
applied.
28,258267
A problem with NMR spectroscopy
of these compounds has been the lack of the large
number of nuclear Overhauser enhancements (NOEs)
that can be observed.
268,269
Nevertheless, when com-
bined with energy calculations it is possible to use
NMR measurements to deduce conformations of sugar
rings, three-dimensional structures, and the degree of
conformational flexibility in various parts of N-linked
oligosaccharides (Fig. 4-21). Measurement of COC
C spin-coulping constants is also of value.
270
Use of
multidimensional NMR has permitted analysis of
mixtures of cellulose oligosaccharides.
269
193
101. Minke, R., and Blackwell, J. (1978) J. Mol. Biol.
120, 167181
102. Roberts, G. A. F. (1992) Chitin Chemistry,
MacMillan, London
103. Blackwell, J., and Weih, M. A. (1980) J. Mol.
Biol. 137, 4960
104. Rees, D. A., and Welsh, E. J. (1977) Angew.
Chem. Int. Ed. Engl. 16, 214224
105. Lee, Y. C. (1992) FASEB J. 6, 31933200
106. Laurent, T. C., and Fraser, J. R. E. (1992)
FASEB J. 6, 23972404
107. Hardingham, T. E., and Fosang, A. J. (1992)
FASEB J. 6, 861870
108. Fransson, L. (1987) Trends Biochem. Sci. 12,
406411
109. Morris, E. R., Rees, D. A., and Welsh, E. J.
(1980) J. Mol. Biol. 138, 383400
110. David, G. (1993) FASEB J. 7, 10231030
111. Yamada, S., Murakami, T., Tsuda, H., Yoshida,
K., and Sugahara, K. (1995) J. Biol. Chem. 270,
86968705
112. Lane, D. A., and Bjrk, I., eds. (1992) Heparin
And Related Polysaccharides, Plenum, New York
113. Edens, R. E., Fromm, J. R., Linhardt, R. J., and
Weiler, J. M. (1995) Biochemistry 34, 24002407
114. Linhardt, R. J., Ampofo, S. A., Fareed, J.,
Hoppensteadt, D., Mulliken, J. B., and
Folkman, J. (1992) Biochemistry 31, 12441
12445
115. Casu, B., Petitou, M., Provasoli, M., and Sina,
P. (1988) Trends Biochem. Sci. 13, 221225
116. Oates, J. A., and Wood, A. J. J. (1991) N. Engl.
J. Med. 324, 15651574
117. Rosenberg, R. D., and Lam, L. (1979) Proc.
Natl. Acad. Sci. U.S.A. 76, 12181222
118. Lindahl, U., Kusche-Gullberg, M., and Kjelln,
L. (1998) J. Biol. Chem. 273, 2497924982
119. Mulloy, B. A., Ribeiro, A.-C., Alves, A.-P.,
Vieira, R. P., and Mourao, P. A. S. (1994) J. Biol.
Chem. 269, 2211322123
120. Gibeaut, D., and Carpita, N. C. (1994) FASEB
J. 8, 904915
121. Hayashi, T., and Matsuda, K. (1981) J. Biol.
Chem. 256, 1111711122
122. Millane, R. P. (1992) in Frontiers in Carbohy-
drate Research-2 (Chandrasekaran, R., ed), pp.
168190, Elsevier, London
123. Rolin, C. (1993) in Industrial Gums, 3rd ed.
(Whistler, R. L., and BeMiller, J. N., eds), pp.
257293, Academic Press, San Diego,
California
124. Arnott, S., Scott, W. E., Rees, D. A., and
McNab, C. G. A. (1974) J. Mol. Biol. 90, 253
267
125. Selby, H. H., and Whistler, R. L. (1993) in
Industrial Gums, 3rd ed. (Whistler, R. L., and
BeMiller, J. N., eds), pp. 87103, Academic
Press, San Diego, California
126. Albersheim, P., Bauer, W. D., Keestra, K.,
Talmadge, K. W. (1973) Biogenesis of Plant Cell
Wall Polysaccharides, (Loewus, F., ed.),
Academic Press, New York
127. Rees, D. A. (1972) Biochem. J. 126, 257273
128. Therkelsen, G. H. (1993) in Industrial Gums,
3rd ed. (Whistler, R. L., and BeMiller, J. N.,
eds), pp. 145180, Academic Press, San
Diego, California
129. Clare, K. (1993) in Industrial Gums, 3rd ed.
(Whistler, R. L., and BeMiller, J. N., eds), pp.
105143, Academic Press, San Diego,
California
130. Marsh, M. E., Chang, D. C., and King, G. C.
(1992) J. Biol. Chem. 267, 2050720512
130a. Vinogradov, E., and Bock, K. (1999) Angew.
Chem. Ed. Engl. 38, 671674
131. Sutherland, I. W. (1979) Trends Biochem. Sci. 4,
5559
References
42. Takata, K., Horiuchi, S., Araki, N., Shiga, M.,
Saitoh, M., and Morino, Y. (1988) J. Biol. Chem.
263, 1481914825
43. Hayase, F., Nagaraj, R. H., Miyata, S.,
Njoroge, F. G., and Monnier, V. M. (1989)
J. Biol. Chem. 264, 37583764
44. Goldberg, R. N., and Tewari, Y. B. (1989)
J. Biol. Chem. 264, 98979900
45. Tewari, Y. B., and Goldberg, R. N. (1989)
J. Biol. Chem. 264, 39663971
46. Goldberg, R. N., Tewari, Y. B., and Ahluwalia,
J. C. (1989) J. Biol. Chem. 264, 99019904
47. Weissborn, A. C., Rumley, M. K., and Kennedy,
E. P. (1991) J. Biol. Chem. 266, 80628067
48. Reymond, P., Grnberger, S., Paul, K., Mller,
M., and Farmer, E. E. (1995) Proc. Natl. Acad.
Sci. U.S.A. 92, 41454149
49. Nacharaju, P., and Acharya, A. S. (1992)
Biochemistry 31, 1267312679
50. Baynes, J. W., and Monnier, V. M., eds. (1989)
the Maillard Reaction in Aging, Diabetes and
Nutrition, Liss, New York
51. Larsen, M. L., Hrder, M., and Mogensen, E.
F. (1990) N. Engl. J. Med. 323, 10211025
52. Luthra, M., and Balasubramanian, D. (1993) J.
Biol. Chem. 268, 1811918127
53. Urbanowski, J. C., Cohenford, M. A., and
Dain, J. A. (1982) J. Biol. Chem. 257, 111115
54. Grandhee, S. K., and Monnier, V. M. (1991) J.
Biol. Chem. 266, 1164911653
55. Brownlee, M., Cerami, A., and Vlassara, H.
(1988) N. Engl. J. Med. 318, 13151320
56. Papoulis, A., Al-Abed, Y., and Bucala, R.
(1995) Biochemistry 34, 648655
57. Wells-Knecht, K. J., Zyzak, D. V., Litchfield, J.
E., Thorpe, S. R., and Baynes, J. W. (1995)
Biochemistry 34, 37023709
58. French, D. (1975) in International Review of
Biochemistry, Vol. 5 (Whelan, W. J., ed), p. 269,
Univ. Park Press, Baltimore, Maryland (Series
One)
59. Atkins, E. D. T., ed. (1986) Polysaccharides,
Topics in Structure and Morphology, VCH
Publisher, New York
60. Whistler, R. L., and BeMiller, J. N., eds. (1993)
Industrial Gums, 3rd ed., Academic Press, San
Diego, California
61. Rao, V. S. R., and Sathyanarayana, B. K. (1972)
Biopolymers 11, 13791394
62. French, A. D., and Dowd, M. K. (1993) in
Cellulostics: Chemical,Biochemical and Materials
Aspects (Kennedy, J. F., Phillips, G. O., and
Williams, P. A., eds), pp. 5156, Ellis
Harwood, New York
63. Duda, C. A., and Stevens, E. S. (1993) J. Am.
Chem. Soc. 115, 84878488
64. Libecq, C., ed. (1992) Biochemical Nomencla-
ture, Portland Press, London and Chapel Hill,
North Carolina (for the International Union of
Biochemistry and Molecular Biology)
65. Preston, R. D. (1986) Trends Biochem. Sci. 11,
377380
66. Ross, P., Mayer, R., and Benziman, M. (1991)
Microbiol. Rev. 55, 3558
67. Sugiyama, J., Vuong, R., and Chanzy, H.
(1991) Macromolecules 24, 41684175
68. Dudley, R. L., Fyfe, C. A., Stephenson, P. J.,
Deslandes, Y., Hamer, G. K., and
Marchessault, R. H. (1983) J. Am. Chem. Soc.
105, 24692472
69. Gebler, K., Kraub, N., Steiner, T., Betzel, C.,
Sandmann, C., and Saenger, W. (1994) Science
266, 10271029
69a. Langan, P., Nishiyama, Y., and Chanzy, H.
(1999) J. Am. Chem. Soc. 121, 9940 9946
70. Grover, J. A. (1993) in Industrial Gums, 3rd ed.
(Whistler, R. L., and BeMiller, J. N., eds), pp.
475504, Academic Press, San Diego,
California
71. French, D. (1973) J. Animal Sci. 37, 10481061
72. Maddelein, M. L., Libessart, N., Bellanger, F.,
Delrue, B., DHulst, C., Van den Koornhuyse,
N., Fontaine, T., Wieruszeski, J. M., Decq, A.,
and Ball, S. (1994) J. Biol. Chem. 269, 25150
25157
73. Jane, J., Kasemsuwan, T., Leas, S., Zobel, H.,
and Robyt, J. F. (1994) Starch/Strke 46, 121
129
74. Meyer, K. H. (1943) Adv. Enzymol. 3, 109136
75. French, D. (1969) in Symposium on Foods:
Carbohydrates and their Roles (Schultz, H. W.,
ed), pp. 2654, AVI Publ. Co., Westport,
Connecticut
76. Rundle, R. E., and French, D. (1943) J. Am.
Chem. Soc. 65, 17071710
77. Hybl, A., Rundle, R. E., and Williams, D. E.
(1965) J. Am. Chem. Soc. 87, 27792788
78. Teitelbaum, R. C., Ruby, S. L., and Tobin, J. M.
(1980) J. Am. Chem. Soc. 102, 33223328
79. Winter, W. T., and Sarko, A. (1974) Biopolymers
13, 14611482
80. Kainuma, K., and French, D. (1972)
Biopolymers 11, 22412250
81. Wu, H.-C., H, and Sarko, A. (1978) Carbohydr.
Res. 61, 725
82. Gidley, M. J., and Bociek, S. M. (1988) J. Am.
Chem. Soc. 110, 38203829
83. Lomako, J., Lomako, W. M., Whelan, W. J.,
Dombro, R. S., Neary, J. T., and Norenberg, M.
D. (1993) FASEB J. 7, 13861393
84. Chen, W., Avison, M. J., Zhu, X. H., and
Shulman, R. G. (1993) Biochemistry 32, 11483
11487
85. Ohana, P., Delmer, D. P., Steffens, J. C.,
Matthews, D. E., Mayer, R., and Benziman, M.
(1991) J. Biol. Chem. 266, 1374213745
86. Mol, P. C., Park, H., Mullins, J. T., and Cabib,
E. (1994) J. Biol. Chem. 269, 3126731274
87. Yamamoto, R., and Nevins, D. J. (1978)
Carbohydrate Res. 67, 275280
88. Marchessault, R. H., and Deslandes, Y. (1979)
Carbohydrate Res. 75, 231242
89. Kang, M. S., and Cabib, E. (1986) Proc. Natl.
Acad. Sci. U.S.A. 83, 58085812
90. Harada, T., Terasaki, M., and Harada, A.
(1993) in Industrial Gums, 3rd ed. (Whistler, R.
L., and BeMiller, J. N., eds), pp. 427445,
Academic Press, San Diego, California
91. Walker, G. J. (1978) in International Review of
Biochemistry, Vol. 16 (Manners, D. J., ed), p. 75,
Univ. Park Press, Baltimore, Maryland
92. de Belder, A. N. (1993) in Industrial Gums, 3rd
ed. (Whistler, R. L., and BeMiller, J. N., eds),
pp. 399425, Academic Press, San Diego,
California
93. Yang, X., Miller, M. A., Yang, R., Evans, D. F.,
and Edstrom, R. D. (1990) FASEB J. 4, 3140
3143
94. Tsujisaka, Y., and Mitsuhashi, M. (1993) in
Industrial Gums, 3rd ed. (Whistler, R. L., and
BeMiller, J. N., eds), pp. 447460, Academic
Press, San Diego, California
95. Arnott, S., Fulmer, A., Scott, W. E., Dea, I. C.
M., Moorehouse, R., and Rees, D. A. (1974) J.
Mol. Biol. 90, 269284
96. Shibata, N., Ikuta, K., Imai, T., Satoh, Y.,
Satoh, R., Suzuki, A., Kojima, C., Kobayashi,
H., Hisamichi, K., and Suzuki, S. (1995) J. Biol.
Chem. 270, 11131122
97. Kollr, R., Petrkov, E., Ashwell, G., Robbins,
P. W., and Cabib, E. (1995) J. Biol. Chem. 270,
11701178
98. Preston, R. D. (1968) Sci. Am. 218(Jun), 102108
99. Turvey, J. R. (1978) in International Review of
Biochemistry, Vol. 16 (Manners, D. J., ed), Univ.
Park Press, Baltimore, Maryland
100. Kirkwood, S. (1974) Ann. Rev. Biochem. 43,
401417
194 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
References
132. Kang, K. S., and Pettitt, D. J. (1993) in
Industrial Gums, 3rd ed. (Whistler, R. L., and
BeMiller, J. N., eds), pp. 341397, Academic
Press, San Diego, California
133. Wolucka, B. A., McNeil, M. R., de Hoffmann,
E., Chonjnacki, T., and Brennan, P. J. (1994) J.
Biol. Chem. 269, 2332823335
134. Kobata, A. (1984) in Biology of Carbohydrates,
Vol. 2 (Ginsburg, V., and Robbins, P. W., eds),
pp. 87161, Wiley, New York
135. Lennarz, W. J., ed. (1980) The Biochemistry of
Glycoproteins and Proteoglycans, Plenum, New
York
136. Montreul, J. (1982) in Comprehensive Biochemistry,
1st ed., Vol. 19B, Part II (Florkin, M., and Stotz,
E. H., eds), pp. 1188, Elsevier, Amsterdam
137. Rademacher, T. W., Parekh, R. B., and Dwek,
R. A. (1988) Ann. Rev. Biochem. 57, 785838
138. Roberts, D. D., and Mecham, R. P., eds. (1993)
Cell Surface and Extracellular Glycoconjugates:
Structure and Function, Academic Press, San
Diego, California
138a. Varki, A., Cummings, R., Esko, J., Freeze, H.,
Hart, G., and Marth, J., eds. (1999) Essentials of
Glycobiology, Cold Spring Harbor Lab. Press,
Cold Spring Harbor, New York
139. Fukuda, M., and Hindsgaul, O., eds. (1994)
Molecular Glycobiology, Oxford Publishing,
Oxford
140. Williams, R. L., Greene, S. M., and McPherson,
A. (1987) J. Biol. Chem. 262, 1602016031
141. Hofsteenge, J., Mller, D. R., de Beer, T.,
Lffler, A., Richter, W. J., and Vliegenthart, J.
F. G. (1994) Biochemistry 33, 1352413530
141a. Iozzo, R. V. (1998) Ann. Rev. Biochem. 67, 609
652
141b. Bernfield, M., Gtte, M., Park, P. W., Reizes,
O., Fitzgerald, M. L., Lincecum, J., and Zako,
M. (1999) Ann. Rev. Biochem. 68, 729 777
142. Sugahara, K., Ohkita, Y., Shibata, Y., Yoshida,
K., and Ikegami, A. (1995) J. Biol. Chem. 270,
72047212
143. Funderburgh, J. L., Funderburgh, M. L.,
Brown, S., Vergnes, J.-P., Hassell, J. R., Mann,
M. M., and Conrad, G. W. (1993) J. Biol. Chem.
268, 1187411880
144. Neame, P. J., Christner, J. E., and Baker, J. R.
(1987) J. Biol. Chem. 262, 1776817778
145. Scott, J. E. (1987) Trends Biochem. Sci. 12, 318
321
146. Rosenberg, L., Tang, L.-H., Pal, S., Johnson, T.
L., and Choi, H. U. (1988) J. Biol. Chem. 263,
1807118077
147. Choi, H. U., Johnson, T. L., Pal, S., Tang, L.-H.,
Rosenberg, L., and Neame, P. J. (1989) J. Biol.
Chem. 264, 28762884
148. Kokenyesi, R., and Bernfield, M. (1994) J. Biol.
Chem. 269, 1230412309
149. Rosenberg, L. (1975) in Dynamics of Connective
Tissue Macromolecules (Burleigh, P. M. C., and
Poole, A. R., eds), p. 107, North-Holland
Publ., Amsterdam
150. Chen, J., and Varner, J. E. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 43994403
151. Kieliszewski, M., deZacks, R., Leykam, J. F.,
and Lamport, D. T. A. (1992) Plant Physiol. 98,
919926
152. Kieliszewski, M. J., ONeill, M., Leykam, J.,
and Orlando, R. (1995) J. Biol. Chem. 270,
25412549
153. Eckhardt, A. E., Timpte, C. S., Abernethy, J. L.,
Toumadje, A., Johnson, W. C., and Hill, R. L.
(1987) J. Biol. Chem. 262, 1133911344
154. Timpte, C. S., Eckhardt, A. E., Abernethy, J. L.,
and Hill, R. L. (1988) J. Biol. Chem. 263, 1081
1088
155. Gupta, R., and Jentoft, N. (1989) Biochemistry
28, 61146121
156. Carter, S. R., Slomiany, A., Gwozdzinski, K.,
Liau, Y. H., and Slomiany, B. L. (1988) J. Biol.
Chem. 263, 1197711984
157. Jentoft, N. (1990) Trends Biochem. Sci. 15, 291
294
158. Butenhof, K. J., and Gerken, T. A. (1993)
Biochemistry 32, 2650 2663
159. Toribara, N. W., Roberton, A. M., Ho, S. B.,
Kuo, W.-L., Gum, E., Hicks, J. W., Gum, J. R.,
Byrd, J. C., Siddiki, B., and Kim, Y. S. (1993) J.
Biol. Chem. 268, 58795885
160. Sadler, J. E. (1984) in Biology of Carbohy-
drates, Vol. 2 (Ginsburg, V., and Robbins, P.
W., eds), pp. 199288, Wiley, New York
160a. Perez-Vilar, J., and Hill, R. L. (1999) J. Biol.
Chem. 274, 3175131754
161. Fukuda, M., Carlsson, S. R., Klock, J. C., and
Dell, A. (1986) J. Biol. Chem. 261, 1279612806
162. Gerken, T. A., and Jentoft, N. (1987)
Biochemistry 26, 46894699
163. Freeze, H. H., and Wolgast, D. (1986) J. Biol.
Chem. 261, 127134
164. Carver, J. P., and Brisson, J.-R. (1984) in
Biology of Carbohydrates, Vol. 2 (Ginsburg, V.,
and Robbins, P. W., eds), pp. 289329, Wiley,
New York
165. Bush, C. A., Duben, A., and Ralapati, S. (1980)
Biochemistry 19, 501504
166. Finne, J., and Krusius, T. (1979) Eur. J. Biochem.
102, 583588
167. Jrnefelt, J., Finne, J., Krusius, T., and Rauvala,
H. (1978) Trends Biochem. Sci. 3, 110114
168. Kornfeld, R., and Kornfeld, S. (1980) in The
Biochemistry of Glycoproteins and Proteoglycans
(Lennarz, W. J., ed), Plenum, New York
169. Fukuda, M., Dell, A., and Fukuda, M. N.
(1984) J. Biol. Chem. 259, 47824791
170. Hernndez, L. M., Olivero, I., Alvarado, E.,
and Larriba, G. (1992) Biochemistry 31, 9823
9831
171. Taguchi, T., Seko, A., Kitajima, K., Muto, Y.,
Inoue, S., Khoo, K.-H., Morris, H. R., Dell, A.,
and Inoue, Y. (1994) J. Biol. Chem. 269, 8762
8771
172. Wieland, F., Paul, G., and Sumper, M. (1985) J.
Biol. Chem. 260, 1518015185
173. Funderburgh, J. L., Funderburgh, M. L.,
Mann, M. M., and Conrad, G. W. (1991) J. Biol.
Chem. 266, 1422614231
174. Sharon, N., and Lis, H. (1989) Science 246,
227229
175. Sharon, N., and Lis, H. (1993) Sci. Am.
268(Jan), 82 89
176. Opdenakker, G., Rudd, P. M., Ponting, C. P.,
and Dwek, R. A. (1993) FASEB J. 7, 13301337
177. Sharon, N. (1993) Trends Biochem. Sci. 18, 221
226
178. Barondes, S. H. (1988) Trends Biochem. Sci. 13,
480482
179. Sphyris, N., Lord, J. M., Wales, R., and
Roberts, L. M. (1995) J. Biol. Chem. 270, 20292
20297
180. Sumner, J. B., and Howell, S. F. (1936) J.
Bacteriol. 32, 227237
181. Bourne, Y., Rougs, P., and Cambillau, C.
(1990) J. Biol. Chem. 265, 18161 18165
182. Sharon, N., and Lis, H. (1990) FASEB J. 4,
31983208
183. Mandal, D. K., and Brewer, C. F. (1993)
Biochemistry 32, 5116 5120
184. Derewenda, Z., Yariv, J., Helliwell, J. R., Kalb,
A. J., Dodson, E. J., Papiz, M. Z., Wan, T., and
Campbell, J. (1989) EMBO J. 8, 2189 2193
185. Drickamer, K. (1988) J. Biol. Chem. 263, 9557
9560
186. Krusius, T., Gehlsen, K. R., and Ruoslahti, E.
(1987) J. Biol. Chem. 262, 1312013125
187. Gitt, M. A., and Barondes, S. H. (1991)
Biochemistry 30, 8289
188. Barondes, S. H., Cooper, D. N. W., Gitt, M. A.,
and Leffler, H. (1994) J. Biol. Chem. 269,
2080720810
188a. Varela, P. F., Sols, D., Daz-Maurino, T.,
Kaltner, H., Gabius, H.-J., and Romero, A.
(1999) J. Mol. Biol. 294, 537549
189. Chiu, M. L., Parry, D. A. D., Feldman, S. R.,
Klapper, D. G., and OKeefe, E. J. (1994) J. Biol.
Chem. 269, 3177031776
190. Madsen, P., Rasmussen, H. H., Flint, T.,
Gromov, P., Kruse, T. A., Honor, B., Vorum,
H., and Celis, J. E. (1995) J. Biol. Chem. 270,
58235829
191. Cho, M., and Cummings, R. D. (1995) J. Biol.
Chem. 270, 52075212
192. Gitt, M. A., Wiser, M. F., Leffler, H.,
Herrmann, J., Xia, Y.-R., Massa, S. M., Cooper,
D. N. W., Lusis, A. J., and Barondes, S. H.
(1995) J. Biol. Chem. 270, 50325038
193. Weis, W. I., Kahn, R., Fourme, R., Drickamer,
K., and Hendrickson, W. A. (1991) Science 254,
16081615
194. Vyas, N. K., Vyas, M. N., and Quiocho, F. A.
(1988) Science 242, 12901295
195. Shaanan, B., Lis, H., and Sharon, N. (1991)
Science 254, 862865
196. Cygler, M., Rose, D. R., and Bundle, D. R.
(1991) Science 253, 442445
197. ngstrm, J., Teneberg, S., and Karlsson, K.-
A. (1994) Proc. Natl. Acad. Sci. U.S.A. 91,
1185911863
198. Sauter, N. K., Hanson, J. E., Glick, G. D.,
Brown, J. H., Crowther, R. L., Park, S.-J.,
Skehel, J. J., and Wiley, D. C. (1992)
Biochemistry 31, 96099621
199. Janakiraman, M. N., White, C. L., Laver, W.
G., Air, G. M., and Luo, M. (1994) Biochemistry
33, 81728179
200. Bossart-Whitaker, P., Carson, M., Babu, Y. S.,
Smith, C. D., Laver, W. G., and Air, G. M.
(1993) J. Mol. Biol. 232, 10691083
201. Crennell, S. J., Garman, E. F., Laver, E. G.,
Vimr, E. R., and Taylor, G. L. (1993) Proc. Natl.
Acad. Sci. U.S.A. 90, 98529856
202. Colli, W. (1993) FASEB J. 7, 12571264
203. Ofek, I., Beachey, E. H., and Sharon, N. (1978)
Trends Biochem. Sci. 3, 159160
204. Lindberg, F., Lund, B., and Normark, S. (1986)
Proc. Natl. Acad. Sci. U.S.A. 83, 18911895
205. Cho, J.-W., and Troy, F. A. (1994) Proc. Natl.
Acad. Sci. U.S.A. 91, 1142711431
206. Baumann, H., Brisson, J., Michon, F., Pon, R.,
and Jennings, H. J. (1993) Biochemistry 32,
40074013
207. Falk, P. G., Bry, L., Holgersson, J., and
Gordon, J. I. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 15151519
208. Adler, P., Wood, S. J., Lee, Y. C., Lee, R. T.,
Petri, W. A., and Schnaar, R. L. (1995) J. Biol.
Chem. 270, 51645171
209. Misevic, G. N., and Burger, M. M. (1990) J.
Biol. Chem. 265, 2057720584
210. Spillmann, D., Hrd, K., Thomas-Oates, J.,
Vliegenthart, J. F. G., Misevic, G., Burger, M.
M., and Finne, J. (1993) J. Biol. Chem. 268,
1337813387
211. Spillmann, D., Thomas-Oates, J. E., van Kuik,
J. A., Vliegenthart, J. F. G., Misevic, G., Burger,
M. M., and Finne, J. (1995) J. Biol. Chem. 270,
50895097
212. Rosati, F., and De Santis, R. (1980) Nature
(London) 283, 762764
213. Seppo, A., Penttil, L., Niemel, R.,
Maaheimo, H., and Renkonen, O. (1995)
Biochemistry 34, 46554661
214. Litscher, E. S., Juntunen, K., Seppo, A.,
Penttil, L., Niemel, R., Renkonen, O., and
Wassarman, P. M. (1995) Biochemistry 34,
46624669
195
References
215. Hitoshi, S., Kusunoki, S., Kanazawa, I., and
Tsuji, S. (1995) J. Biol. Chem. 270, 88448850
216. de Vries, T., Srnka, C. A., Palcic, M. M.,
Swiedler, S. J., van der Eijnden, D. H., and
Macher, B. A. (1995) J. Biol. Chem. 270, 8712
8722
217. Stroud, M. R., Levery, S. B., Mrtensson, S.,
Salyan, M. E. K., Clausen, H., and Hakomori,
S. (1994) Biochemistry 33, 1067210680
218. Hull, S. R., Sugarman, E. D., Spielman, J., and
Carraway, K. L. (1991) J. Biol. Chem. 266,
13580 13586
219. Ware, F. E., Vassilakos, A., Peterson, P. A.,
Jackson, M. R., Lehrman, M. A., and Williams,
D. B. (1995) J. Biol. Chem. 270, 46974704
220. Leppnen, A., Mehta, P., Ouyang, Y.-B., Ju, T.,
Helin, J., Moore, K. L., van Die, I., Canfield,
W. M., McEver, R. P., and Cummings, R. D.
(1999) J. Biol. Chem. 274, 24838 24848
221. Chandrasekaran, E. V., Jain, R. K., Larsen, R.
D., Wlasichuk, K., and Matta, K. L. (1995)
Biochemistry 34, 29252936
222. Kogan, T. P., Revelle, B. M., Tapp, S., Scott, D.,
and Beck, P. J. (1995) J. Biol. Chem. 270, 14047
14055
223. Waddell, T. K., Fialkow, L., Chan, C. K.,
Kishimoto, T. K., and Downey, G. P. (1995) J.
Biol. Chem. 270, 1540315411
224. Bertozzi, C. R., Fukuda, S., and Rosen, S. D.
(1995) Biochemistry 34, 1427114277
225. Norgard-Sumnicht, K., and Varki, A. (1995) J.
Biol. Chem. 270, 1201212024
226. Hemmerich, S., Leffler, H., and Rosen, S. D.
(1995) J. Biol. Chem. 270, 1203512047
227. Yuen, C.-T., Bezouska, K., OBrien, J., Stoll,
M., Lemoine, R., Lubineau, A., Kiso, M.,
Hasegawa, A., Bockovich, N. J., Nicolaou, K.
C., and Feiz, T. (1994) J. Biol. Chem. 269, 1595
1598
228. Patel, T. P., Goelz, S. E., Lobb, R. R., and
Parekh, R. B. (1994) Biochemistry 33, 14815
14824
229. Graves, B. J., Crowther, R. L., Chandran, C.,
Rumberger, J. M., Li, S., Huang, K.-S., Presky,
D. H., Familletti, P. C., Wolitzky, B. A., and
Burns, D. K. (1994) Nature (London) 367, 532
538
230. Lasky, L. A. (1995) Ann. Rev. Biochem. 64, 113
139
231. Ginsburg, V. E. (1982) Meth. Enzymol. 83, all
232. Churms, S. C. (1990) CRC Handbook of
Chromatography Carbohydrates, Vol. 2, CRC
Press, Boca Raton, Florida
233. Valent, B. S., Darvill, A. G., McNeil, M.,
Robertsen, B. K., and Albersheim, P. (1980)
Carbohydrate Res. 79, 165192
234. Chaplin, M. F., and Kennedy, J. F., eds. (1994)
Carbohydrate Analysis A Practical Approach,
Oxford University, Oxford
235. Robyt, J. F., and Mukerjea, R. (1994) Carbohydr.
Res. 251, 187202
236. Green, E. D., and Baenziger, J. U. (1989) Trends
Biochem. Sci. 14, 168172
237. Carr, S. A., Huddleston, M. J., and Bean, M. F.
(1993) Protein Sci. 2, 183196
237a. Fridriksson, E. K., Beavil, A., Holowka, D.,
Gould, H. J., Baird, B., and McLafferty, F. W.
(2000) Biochemistry 39, 3369 3376
237b. Burlingame, A. L., Carr, S. A., and Baldwin,
M. A., eds. (2000) Mass Spectrometry in Biology
and Medicine, Hamana Press, Totowa, New
Jersey
238. Downs, F., and Pigman, W. (1969) Biochemistry
8, 17601766
239. Narasimhan, S., Harpaz, N., Longmore, G.,
Carver, J. P., Grey, A. A., and Schacter, H.
(1980) J. Biol. Chem. 255, 48764884
240. Cohen, R. E., and Ballou, C. E. (1980)
Biochemistry 19, 43454358
241. Plummer, T. H., Jr., and Tarentino, A. L. (1981)
J. Biol. Chem. 256, 1024310246
242. Risley, J. M., and Van Etten, R. L. (1985) J. Biol.
Chem. 260, 1548815494
243. Takasaki, S., Mizuochi, T., and Kobata, A.
(1982) Meth. Enzymol. 83, 263268
244. Parente, J. P., Wieruszeski, J.-M., Strecker, G.,
Montreuil, J., Fournet, B., van Halbeek, H.,
Dorland, L., and Vliegenthart, J. F. G. (1982) J.
Biol. Chem. 257, 1317313176
245. Patel, T., Bruce, J., Merry, A., Bigge, C.,
Wormald, M., Jaques, A., and Parekh, R.
(1993) Biochemistry 32, 679693
246. Sweeley, C. C., and Nunez, H. A. (1985) Ann.
Rev. Biochem. 54, 765801
247. Dua, V. K., Roa, B. N. N., Wu, S.-S., Dube, V.
E., and Bush, C. A. (1986) J. Biol. Chem. 261,
15991608
248. Tsai, P.-K., Dell, A., and Ballou, C. E. (1986)
Proc. Natl. Acad. Sci. U.S.A. 83, 41194123
249. Lindberg, B., Lnngren, L., and Svensson, S.
(1975) Adv. Carbohydr. Chem. Biochem. 31,
185240
250. Guthrie, R. D., and McCarthy, J. F. (1967) Adv.
Carbohydr. Chem. Biochem. 22, 1123
251. Edge, C., Parekh, R., Rademacher, T.,
Wormald, M., and Dwek, R. (1992) Nature
(London) 385, 693694
252. Varki, A. (1991) FASEB J. 5, 226235
253. Rolf, D., and Gray, G. R. (1982) J. Am. Chem.
Soc. 104, 35393541
254. Mukerjea, R., Kim, D., and Robyt, J. F. (1996)
Carbohydrate Res. 292, 1120
255. Bobbitt, J. M. (1956) Adv. Carbohydr. Chem.
Biochem. 11, 141
256. Rich, A., Nordheim, A., and Wang, A. H.-J.
(1984) Ann. Rev. Biochem. 53, 791846
257. Homans, S. W., Dwek, R. A., and Rademacher,
T. W. (1987) Biochemistry 26, 65716578
258. Homans, S. W., Pastore, A., Dwek, R. A., and
Rademacher, T. W. (1987) Biochemistry 26,
6649 6655
259. Cumming, D. A., Shah, R. N., Krepinsky, J. J.,
Grey, A. A., and Carver, J. P. (1987) Biochemistry
26, 66556663
260. Reuben, J. (1985) J. Am. Chem. Soc. 107, 1747
1755
261. Cumming, D. A., Dime, D. S., Grey, A. A.,
Krepinsky, J. J., and Carver, J. P. (1986) J. Biol.
Chem. 261, 32083213
262. Vuister, G. W., de Waard, P., Boelens, R.,
Vliegenthart, J. F. G., and Kaptein, R. (1989)
J. Am. Chem. Soc. 111, 772774
263. Gronenborn, A. M., and Clore, G. M. (1989)
Biochemistry 28, 59785984
264. Abeygunawardana, C., and Bush, C. A. (1991)
Biochemistry 30, 85688577
265. Rivire, M., and Puzo, G. (1992) Biochemistry
31, 35753580
266. van Duynhoven, J. P. M., Goudriaan, J.,
Hilbers, C. W., and Wijmenga, S. S. (1992) J.
Am. Chem. Soc. 114, 1005510056
267. Wu, J., Bondo, P. B., Vuorinen, T., and
Serianni, A. S. (1992) J. Am. Chem. Soc. 114,
34993505
268. Homans, S. W. (1990) Biochemistry 29, 9110
9118
269. Bose, B., Zhao, S., Stenutz, R., Cloran, F.,
Bondo, P. B., Bondo, G., Hertz, B., Carmichael,
I., and Serianni, A. S. (1998) J. Am. Chem. Soc.
120, 1115811173
270. Flugge, L. A., Blank, J. T., and Petillo, P. A.
(1999) J. Am. Chem. Soc. 121, 7228 7238
196 Chapter 4. Sugars, Polysaccharides, and Glycoproteins
1. A nonreducing disaccharide gives an octamethyl
derivative with dimethyl sulfate and alkali. On
acid hydrolysis, this derivative yields 1 mol of
2,3,4,6-tetramethyl-D-glucose and 1 mol of 2,3,4,6-
tetramethyl-D-galactose. The disaccharide is
hydrolyzed rapidly by either maltase or lactase (a
-galactosidase).
Give an adequately descriptive name of the disac-
charide, and draw its Haworth projection formula.
2. An aldopentose (A) of the D-configuration on
oxidation with concentrated nitric acid gives a
2,3,4-trihydroxypentanedioic acid (a
trihydroxyglutaric acid) (B) which is optically
inactive. (A) on addition of HCN, hydrolysis,
lactonization, and reduction gives two stereoiso-
meric aldohexoses (C) and (D). (D) on oxidation
affords a 2,3,4,5-tetrahydroxy-hexanedioic acid (a
saccharic acid) (E) which is optically inactive. Give
structures of compounds (A)-(E).
3. What products are formed when periodic acid
reacts with sorbitol?
4. A 10.0 g sample of glycogen gave 6.0 millimol of
2,3-di-O-methylglucose on methylation and acid
hydrolysis.
a. What percent of the glucose residues in glyco-
gen have chains substituted at the 16
position?
b. What is the average number of glucose residues
per chain?
c. How many millimols of 2,3,6-tri-O-methyl-
glucose were formed?
d. If the molecular weight of the polysaccharide is
2 10
6
, how many glucose residues does it
contain?
e. How many nonreducing ends are there per
molecule or equivalently how many chains are
there per molecule?
5. D-Mannitol is a symmetric molecule, yet it is
optically active. Explain.
6. When D-glucose is treated with acidic methanol,
the first products which can be isolated are mainly
methyl furanosides, but after extensive reaction the
furanosides disappear and methyl glucopyrano-
sides accumulate. Why?
7. Write the structural formulas for (a) 1,6 anhydro
-D-glucopyranose; (b) 1,6 anhydro -D-altrose.
Compound (b) is many times more stable than
compound (a). Explain this on stereochemical
grounds.
8. The disaccharide nigerose is -D-Glup-(1 3)-D-glu.
Write out its structure. How would you prove this
structure using methylation, periodate oxidation,
and other methods.
9. Inositol is 1,2,3,4,5,6-hexahydroxycyclohexane.
Draw configurational formulas for all possible
stereoisomers and indicate which would be
expected to be optically active.
10. Why do you suppose that the major form of
D-fructose in solution is the pyranose form but
D-fructose in sucrose is in the furanose form?
11. D-Xylose is an easily prepared sugar, potentially
available in enormous quantity. What is a common
source? How can it be obtained from this source?
12. How can xylitol be obtained from xylose? Discuss
the stereochemical properties of xylitol.
13. The enzyme xylose isomerase is important indus-
trially. Why?
14. Glucose reacts non-enzymatically with amino acids
and proteins, including hemoglobin, egg-white
proteins and serum albumin. For example, if
glucose is not removed prior to drying, dried egg
whites slowly turn brown and develop off-flavors
and odors. What do you propose as the most likely
first step in the non-enzymatic glucose-protein
chemical reaction? How can the first product
transform spontaneously into a ketose derivative?
15. What characteristics would you expect in a binding
site for a sugar ring in an enzyme, lectin, or other
proteins?
16. Using structural formulas, descirbe the two major
types of linkage of carbohydrate chains or clusters
to proteins to form glycoproteins or proteoglycans.
17. What products would you expect from cellulose as
a result of methylation analysis? Periodate oxida-
tion? The Smith degradation? The action of an
alpha amylase?
Study Questions
197
Study Questions
18. When glycoproteins are treated with alkaline brohy-
dride, amino acid analysis often indicates a decrease in
the amount of serine and a corresponding increase
in the amount of alanine, or a decrease in threonine
with the appearance of -aminobutyric acid.
Explain.
198 Chapter 5. The Nucleic Acids
Contents
A. Structure and Chemistry of Nucleotides
1. Names and Abbreviations
2. AcidBase Chemistry and Tautomerism
3. Absorption of Ultraviolet Light
4. Chemical Reactions of the Bases
5. Base Pairs, Triplets, and Quartets
Strengths of base pairs
Stacking of bases
Tautomerism and base pairing
6. Conformations of Nucleotides
B. Double Helices
1. The B Form of DNA
2. Other Double-Helical Forms of DNA
3. The Conformational Flexibility of Double Helices
Rotational and translational movements of bases
Bends and bulges
Interactions with ions
C. The Topology and Dynamics of Nucleic Acids
1. Rings, Catenanes, and Knots
2. Supercoiled DNA
3. Intercalation
4. Polynucleotides with Three or Four Strands
H-DNA
R-DNA
Designed third strands
Tetraplex (quadruplex) structures
5. Junctions
D. Ribonucleic Acids (RNA)
1. RNA Loops and Turns
2. Transfer RNAs
3 . Properties and Structures of Some Other
RNA Molecules
4. Modified Nucleotides
5. RNA Aptamers
E. The Genetic Code
1. The Reading Frames
2. Variations
3. Palindromes and Other Hidden Messages
4. The Base Composition of DNA and RNA
F. Interaction of Nucleic Acids with Proteins
1. The HelixTurnHelix Motif
2. Other DNA-binding Motifs
Leucine zipper proteins
Zinc fingers
Beta ribbons
The winged helix family
3. RNA-Binding Proteins
G. Viruses and Plasmids
1. Viruses with Single-Stranded DNA
2. Viruses with Double-Stranded DNA
3. Viruses Containing RNA
4. Viruses without Nucleic Acid?
5. Life Cycles
6. Plasmids and Transposable Genetic Elements
203
203
204
205
207
207
209
209
211
211
213
213
213
216
217
217
218
218
218
219
222
226
226
226
227
227
228
230
230
231
232
234
234
235
236
237
238
239
239
239
241
241
241
241
243
243
244
244
244
247
248
248
248
H. Methods of Study
1. Isolation and Separation of Nucleic Acids
2. Hydrolysis and Analysis
3. Characteristic Reactions of the Bases and Backbone
Reactions of nucleophiles
Reactions with electrophilic reagents
Reactions causing cleavage of the sugar-phosphate
backbone
4. Melting, Hybridization, and Polynucleotide Probes
5. Synthesis of Oligonucleotides and Polynucleotides
6. The Polymerase Chain Reaction (PCR)
7. Sequence Determination
Preparing the DNA
Restriction maps and Southern blots
The Sanger dideoxy method
The method of Maxam and Gilbert
Sequencing RNA
Nearest neighbor analysis
Understanding sequences
8. ProteinDNA Interactions
9. Nuclear Magnetic Resonance
249
249
249
251
251
253
254
255
258
260
260
260
260
262
264
264
265
265
266
266
206
224
235
259
203
210
216
232
236
237
245
Boxes
Box 5-A The Alkali Metal Ions
Box 5-B Antitumor DNA Drugs
Box 5-C The RNA World
Box 5-D DNA Fingerprinting
Tables
Table 5-1 Names of Pyrimidine and Purine Bases,
Nucleosides, and 5-Nucleotides
Table 5-2 Gibbs Energies of Formation G
f
at
25C for Addition of One Base Pair to an
Existing RNA Helix
Table 5-3 Helix Parameters for Three Types of DNA
Table 5-4 Some Specialized RNA Molecules
Table 5-5 The Genetic Code
Table 5-6 The Sixty-Four Codons of the Genetic
Code
Table 5-7 Characteristics of Some Individual
Viruses and Groups of Viruses
270
278
References
Study Questions
DNA spreading from the broken head of a bacteriophage T2 phage. This classic
electron micrograph, published by A. K. Kleinschmidt and coworkers in 1962
(Biochem. Biophys. Acta. 61, 857864, 1962) was prepared by spreading the phage
particles suspended in a proteinsalt solution as a mixed monolayer on a water-
air interface. The resultant osmotic shock burst the head and confined the DNA
as a single thread near the phage ghost. After transfer to a suitable carbon
surface, removal of water, and shadowing with platinum, the micrograph was
obtained. Courtesy of Albrecht K. Kleinschmidt
199
N
N
2
3
4
5
6
1
Pyrimidine
The circled H's are the
ones removed when
the bases are attached to
sugars in nucleosides
and nucleotides
Note: all of these molecules are almost perfectly flat!
Cytosine
N
N
H
O
NH
2
Uracil
N
N
H
H
O
O
N
N
Adenine
N
N
H
NH
2
N
N
2
3
4
5
6
1
Purine
N
N
H
9
7
8
N
N
Guanine
N
N
H
O
H
H
2
N
With a methyl group
here it's thymine,
present in DNA
The phosphorus- and nitrogen-containing materials
that came to be known as nucleic acids were first iso-
lated from cells around 1870 by Friedrich Miescher
but were long regarded as something of a curiosity.
1
Nevertheless, the structures of the monomer units, the
nucleotides, were established by 1909 and the correct
polynucleotide structure of the chains of DNA and
RNA was proposed by Levene and Tipson in 1935.
2,3
The nucleotides are made up of three parts:
1. One of the pyrimidine or purine bases
uracil, cytosine, adenine, or guanine (Fig. 5-1).
All four of these bases are present in RNA,
while DNA contains thymine instead of uracil.
Atoms in the bases are numbered 16 or 19.
2. A sugar, either D-ribose or D-2-deoxyribose.
Carbon atoms in sugars are numbered 1 5.
3. Phosphoric acid
Although the biological synthesis is indirect, we
can imagine that nucleotides are formed from these
parts by elimination of two molecules of water as indi-
cated in Eq. 5-1. In nucleic acids the nucleotides are
combined through phosphodiester linkages between
the 5'-hydroxyl of the sugar in one nucleotide and the
3'-hydroxyl of another. Again, we can imagine that
these linkages were formed by the elimination of water
(Eq. 5-2). The structures of a pair of short polynucleo-
tide strands in DNA are shown in Fig. 5-2. That of a
segment of double-helical DNA is shown in Fig. 5-3
and that of a transfer RNA in Figs. 5-30 and 5-31.
Despite the fact that Levene had deduced the
correct structure for polynucleotides, he was thrown
off the trail of a deeper understanding by the roughly
equal amounts of the four bases found in either DNA
Figure 5-1 Structures of the major pyrimidine and purine
bases of DNA and RNA.
or RNA. He assumed that nucleic acids must be regular
repeating polymers for which there was no obvious
biological function. It was not until 1944 that there
The Nucleic Acids
5
0.5 m
200 Chapter 5. The Nucleic Acids
was concrete evidence that DNA carried genetic infor-
mation (see Chapter 26). However, it was James Watson
and Francis Cricks recognition of the double-helical
structure of DNA
4- 9
in 1953 and the mechanism of rep-
lication that this structure implied that captured the
imagination of biologists and chemists alike and paved
the way for the present-day explosion of knowledge
of DNA, RNA, and of the encoded proteins.
Watson and Crick proposed that DNA is a double
helix of two antiparallel polynucleotide chains (Figs. 5-2
and 5-3). The structure was deduced from model build-
ing together with knowledge of the X-ray diffraction
data of Maurice F. Wilkins and Rosalind Franklin
9a
on
O
H
2
C
O
O
OH HO
H
The sugar
D-ribose
OH is replaced by H
in D-2-deoxyribose,
found in DNA
Uracil,
one of the
pyrimidine
bases
2H
2
O
NH
O
N
O
The heavy green
arrows mark the
two reactive groups
involved in the
polymerization to
form nucleic acids
HO
P
O
O
H
2
C
O
OH O
The nucleotide
uridine monophosphate
(UMP or uridylic acid)
H
NH
O
N
O
H
HO
O
P
O
OH
Phosphate
ion
H

5
4
3 2
1
H
2
O
O Sugar-base
O
H HO P
O
O
O
Sugar-base
OH
HO
3
P O Sugar-base
O
P
O
O O
5'
3'
+
Sugar-base
O
H
3'
5'
5'
3'
5'
3'
HO
3
P

(5-1)
(5-2)
artificially formed DNA fibers. An additional key
piece of information was the discovery by Erwin
Chargaff that in all double-stranded DNA the content of
adenine equals that of thymine and the content of guanine
equals that of cytosine.
The most significant feature of the proposed struc-
ture was the pairing of bases in the center through
hydrogen bonding. The pairs and triplets of hydrogen
bonds (Fig. 5-2) could form in the manner shown only
if adenine (A) was paired with thymine (T) and cyto-
sine (C) with guanine (G) at every point in the entire
DNA structure. Thus, the nucleotide sequence in one
chain is complementary to but not identical to that in the
other chain. It was apparent almost immediately that
the sequence of bases in a DNA chain must convey the
encoded genetic information. The complementarity
of the two strands suggested a simple mechanism for
replication of genes during cell divisions. The two
strands could separate and a complementary strand
could be synthesized along each strand to give two
molecules of the DNA, one for each of the two cells.
Figure 5-2 A distorted (flattened) view of the WatsonCrick
structure of DNA showing the hydrogen-bonded base pairs.
N
N
N
H
N
N
O
H
H
O
H
CH2
O
P
H
H
O
N
N
CH3
O
O
H
H
H2C
H
O
O
5'
O
H
H
H2C
H
O
P
O
O
N
N
N
N
O
N
O
O
P
O
O
H
H
H
N
N
O
N
O
H
H
O
CH2
O
P
O
O
N
N
O
N
N
O
H
H
O
CH2
O
P
H
O
O
N H
N
N
O
O
H
H
O
CH2
O
P
H
O
O
O
H2C
P
O
H
3'
5'
3'
H
H
O
O
Helix axis

A
C
G
T
G
T
5
4
3
2
201
Figure 5-3 The double-helical structure of DNA. The structure shown is that
of the B form and is based on coordinates of Arnott and Hukins.
10
The major
and minor grooves, discussed on p. 213, are marked.
The essential correctness of the
concept has been proved.
Two extremely important develop-
ments came in the 1970s: (1) Methods
were found for cutting and rejoining
DNA fragments and for cloning
them in bacteria and (2) ways were
devised for rapid determination of
nucleotide sequences. The appli-
cation of these techniques is now
providing startling advances in
biology and medicine. In 1970 we
knew virtually nothing about the
sequences of nucleotides in genes
but today, we know the sequences
for many thousands of genes of all
types. By the 1980s sequences had
been established for hemoglobin,
11
-globulins,
12
collagen,
13
and for
many enzymes. An example is the
gene sequence for mitochondrial
aspartate aminotransferase (Fig. 5-4).
14
Its coding regions consist of 1299 pairs
of mononucleotides, the bases being
paired as in Fig. 5-2 in double helical
form. Complete DNA sequences
are known for numerous viruses
including the 9740-base pair (bp)
DNA provirus form of the RNA virus
that causes AIDS
15
and for bacterial
viruses such as T7 bacteriophage
(39,936 bp).
16
Also determined in
the 1980s were sequences of human
mitochondrial DNA (16,598 bp),
17
and of chloroplast DNA from the
tobacco plant (155,844 bp).
18
In 1995 the first complete
sequences of bacterial genomes
were obtained (Table 1-3).
19,20
These
were followed by sequences of many
other bacterial genomes,
21
including
the 4.2 x 10
6
bp E. coli genome (Table
1-3). Sequencing of the 16 chromo-
somes of the 12.07 Mbp genome of
yeast
22,23
containing ~6300 genes was
completed in 1996
24
and that of the
97 Mbp genome of the nematode
Caenorhabditis elegans in 1998. By
2000 the sequence of the 180 Mbp
genome of the fruit fly Drosophila
melanogaster was largely completed,
and most sequences of the 3 x 10
9
bp
human genome were known.
24a
Cloning of DNA has not only provided an essential
step in sequence determination but also has given birth
to a new industry devoted to producing proteins from
genes cloned in bacteria, yeast, or other cells. Human
insulin and the antiviral protein interferon were two of
the first proteins produced in this way. Methods now
in use permit us to introduce at will alterations at any
point in a DNA sequence. We are able to locate and
study the genes responsible for many genetic defects.
O
P
O

O
O
P
O
O

O
O
O
O

O P
O
O
O
O
P
O

O
O
O
O
O

P
O
O
O

P O
O
O
O
O

O
P
O
O
O
O
O
O
P
O

O
P
O
O
O
P
O
P
O
O

P
O
O

O
O
O
O
O

P
O
O
O
O
O

P
O
O

P
O
O
O
O

5
3
H
C
O
5
H
H
H
O
O
P
O

O
H
H
2
C
H
2
C
P
C
O
O
C
O
H
H
O
CH
N
N
O
H
N
N
N
H
H
N
H
N
Major
groove
Minor
groove
Helix axis
Radius
~1.0 nm
van der Waals
contact surface
O
O
O
P
O

O
O
O
O

P O
O
O O
O
O

P
H
O

CH
2
O
O
P
O
O

O
O
P
O
O
O
O

3
O
O

P
O
O
O
O
O
O

P
O
O

P
O
CH
2
O

P
O
O
O
O
O
O
O
O
P O

H
O

0 1.0 nm
3 O

5
CH
2
O
H
3
O

O
O
P
C
C
H
H
C
O
O
O
C
H
2
P
O

O
O
2 N
3
N
3
H
~ 2 twist in center
of base pair
N
4
H
H
H
O
6
N
8
O
4
H N
6
H
N
8
O
6
H
H
N
8
H
H
H
C
H N
4
N
4
N
4
Stacked
base pairs
are in van der
Waals contact
A base pair can be
recognized by groups
facing the grooves.
This is a GC pair
N
3
O
2 H
N
2
H
H
N
3
N NH
G-C
C-G
T-A
G-C
T-A
G-C
T-A
C-G
C-G
A-T
T-A
Major
groove
~2.2 nm
Minor
groove
~1.2 nm
Pitch,
3.46 nm
Top view
Side view
202 Chapter 5. The Nucleic Acids
Figure 5-4 The nucleotide sequence of the gene for mitochondrial aspartate aminotransferase from the mouse. From Tsuzuki et al.
14
The gene encodes a 433-residue protein requiring 1299 nucleotide pairs (1.3 kb). However, 29 residues are cut off from the N terminus
to form the mature mitochondrial protein. In addition, the gene is split by nine introns which vary in length from 0.210.2 kb. The
sequences at the ends of the introns are shown. There is also an upstream region (of which 200 nucleotides are shown) at the 5'-end
of the gene. It contains two binding sites for transcription factor Sp1 (boxed). At the 3' end the 993 additional nucleotides contain
signals (boxed) surrounding the polyadenylation site (green arrow) for 3 processing and termination of transcription. The mature
messenger RNA is about 2400 nucleotides (2.4 kb) in length but the gene, with introns, occupies about 25 kb. The +1 marks the
position of the first nucleotide of the initiation codon ATG (encoding methionine) and the asterisk (*) the termination codon TAA.
These are AUG and UAA in the mRNA. Other codons are indicated by amino acid abbreviations.
200 150
5- CATGACCTCCGTGGAGGCGTCGTCGTTCTACCCCCGACCGCAAGGGGCCGCTGACATGATTCGCTCCAGCGCAGGGCCGCT
100 50 +1
Me
CTAGCCACGCCCAGGGAAGTCACTGTCCTCACCTTTTAGGAGCCCGCGCCTCGGTTCCAGCGGACGCTTCCCCAGATCTCGGCTCTACCACCATCCACTGCCGTCTTACCGCCCACC AT
t Ala Leu Leu His Ser Ser Arg Ile Leu Ser Gly Met Ala Ala Ala Phe His Pro Gly Leu Ala Ala Ala Ala Ser Ala Arg Ala Se
G GCC CTC CTG CAC TCC AGC CGC ATC CTC TCC GGG ATG GCT GCT GCC TTT CAC CCT GGC CTA GCT GCT GCA GCC TCT GCC AGA GCC AG GTG
A
INTRON 1 r Ser
AGCCGAGGGATACAGAGATGCAGCGGCACCGGGCCTGCCCTGCCAGCCGAAGTGTGGACCCTGTGAA----(10.2 kb)----ATGGCTGGGGTAACCTATTTCTCATTTCAG C TCC
Trp Trp Thr His Val Glu Met Gly Pro Pro Asp Pro Ile Leu Gly Val Thr Glu Ala Phe Lys Arg Asp Thr Asn Ser Lys Lys Met Asn
TGG TGG ACC CAT GTT GAA ATG GGA CCT CCA GAT CCC ATC CTG GGC GTT ACC GAA GCC TTC AAG AGA GAT ACC AAC AGC AAG AAG ATG AAC
Leu Gly Val Gly Ala Tyr Arg Asp Asp Asn Gly Lys Pro Tyr Val Leu Pro Ser Val Arg Lys A
CTG GGA GTT GGT GCC TAC CGG GAT GAT AAC GGA AAA CCT TAC GTG CTC CCC AGT GTC CGG AAG G GTGAGCTTGGCACTCGTCTCCTGCCAGCTAGGAT
INTRON 2 la Glu Ala Gln Ile Ala Ala Lys Asn Leu Asp Lys Glu Tyr Leu Pro Ile
GTGGAA----(1.4 kb)----AAAAACTAAGACTTATGATTTTCTTGTCTAG CA GAG GCC CAG ATT GCT GCA AAA AAT TTG GAC AAA GAA TAC CTG CCC ATT
Gly Gly Leu Ala Glu Phe Cys Lys Ala Ser Ala Glu Leu Ala Leu Gly Glu Asn Asn Glu Val Asn Glu Val Leu Lys Ser Gly Arg
GGG GGA CTG GCT GAA TTC TGT AAG GCT TCT GCA GAA CTG GCC CTG GGC GAG AAC AAT GAA GTG AAT GAA GTG TTG AAA AGC GGC CGG GTAA
INTRON 3 Phe Val Thr Val Gln Thr Ile Ser Gly
GCCAGCGGAGTCGGGCTTGAGCTTGATA----(1.2 kb)----TGTAAAGGGTAGAGAGAGTGACTCTGTGTATCCCCTGCAG TTC GTC ACT GTG CAG ACC ATT TCC GGG
Thr Gly Ala Leu Arg Val Gly Ala Ser Phe Leu INTRON 4
ACT GGA GCC TTA AGG GTC GGG GCC AGT TTT CTG GTCAGTGGAACTCTTTCAAGAATGAATCTTTTGGGGTGGC----(2.2 kb)----CTTTCTTCATTTTCTCATTC
Gln Arg Phe Phe Lys Phe Ser Arg Asp Val Phe Leu Pro Lys Pro Ser Trp Gly Asn His Thr Pro Ile Phe Arg
CCTTTTATCCCGACTTTTAG CAA AGG TTT TTT AAG TTC AGC CGA GAT GTC TTT CTG CCC AAA CCA TCC TGG GGA AAT CAC ACG CCC ATC TTC AGG
Asp Ala Gly Met Gln Leu Gln Gly Tyr Arg Tyr Tyr Asp Pro Lys Thr Cys Gly Phe Asp Phe Ser Gly Ala Leu Glu Asp Ile Ser
GAT GCC GGC ATG CAG CTC CAA GGT TAT CGC TAC TAT GAC CCC AAG ACT TGC GGT TTT GAC TTC TCC GGA GCC CTA GAA GAC ATA TCA GTAA
INTRON 5 Lys Ile Pro Glu Gln Ser Val Leu Leu Leu
GTGTGGCTTTCCAGGCCCGACTTCTG----(0.2 kb)----GAAGCTGCACAGCCAAAAATCTCGATGTTTCTCCTTAG AAA ATC CCA GAG CAG AGT GTC CTC CTC CTG
His Ala Cys Ala His Asn Pro Thr Gly Val Asp Pro Arg Pro Glu Gln Trp Lys Glu Ile Ala Ser Val Val Lys
CAT GCC TGC GCT CAC AAC CCC ACC GGC GTG GAC CCG CGT CCC GAG CAG TGG AAG GAG ATA GCG TCC GTG GTG AAG GTGAGGAGGATGAAGCGTCA
INTRON 6 Lys Lys Asn Leu Phe Ala Phe Phe Asp Met Ala Tyr Gln Gly
GGAGCTGGTTGCTTAACC----(0.2 kb)----TCAAACACTGGTCTTCTCATTCCTTCCCAG AAA AAG AAT CTC TTC GCA TTC TTT GAC ATG GCC TAC CAA GGC
Phe Ala Ser Gly Asp Gly Asp Lys Asp Ala Trp Ala Val Arg His Phe Ile Glu Gln Gly Ile Asn Val Cys Leu Cys Gln Ser Tyr Ala
TTT GCC AGC GGT GAT GGT GAT AAG GAT GCC TGG GCC GTG CGG CAC TTC ATC GAG CAG GGC ATC AAT GTC TGC CTC TGC CAA TCG TAT GCC
Lys Asn Met Gly Leu Tyr G INTRON 7 ly Glu
AAG AAC ATG GGC CTG TAC G GTAAGCCAGAGGGCCCAATATAAGGTGTGTT----(1.2 kb)----GTTTCAGCCTGCTAGAAGACAGTATCCTCTGTCTTTTCAG GT GAG
Arg Val Gly Ala Phe Thr Val Val Cys Lys Asp Ala Glu Glu Ala Lys Arg Val Glu Ser Gln Leu Lys Ile Leu Ile Arg Pro Leu Tyr
CGT GTG GGA GCC TTC ACG GTG GTC TGC AAA GAT GCA GAA GAA GCC AAA AGG GTG GAG TCA CAG CTG AAG ATC TTG ATC CGT CCC CTG TAT
Ser Asn Pro Pro Leu Asn Gly Ala Arg Ile Ala Ala Thr Ile Leu Thr Ser Pro Asp Leu Arg Lys Gln Tr
TCC AAC CCA CCT CTC AAT GGG GCC CGG ATC GCA GCA ACC ATC CTG ACT TCT CCA GAC TTC CGG AAG CAA TG GTAACGATTACTAGCTGTATACCGT
INTRON 8 p Leu Gln Glu Val Lys Gly Met Ala Asp Arg Ile Ile Ser Met
GACTACAGCTCCATGA----(2.4 kb)----GAGCACGGCATCCCTCTGCTTTCCTCACAG G TTG CAA GAG GTG AAA GGC ATG GCT GAC CGC ATC ATC AGC ATG
Arg Thr Gln Leu Val Ser Asn Leu Lys Lys Glu Gly Ser Ser His Asn Trp Gln His Ile Thr Asp Gln Ile Gly Met Phe Cys Phe Thr
AGG ACC CAG CTG GTC TCC AAC CTG AAG AAA GAG GGC TCT TCC CAC AAC TGG CAG CAC ATC ACC GAC CAG ATC GGC ATG TTC TGT TTC ACC
Gly Leu Lys Pro Glu Gln INTRON 9 Val Glu
GGC CTA AAG CCA GAG CAG GTGAGTGGGCTCTGGTTCCCCACAGCCAACCCCCGC----(2.0 kb)----ACCAGAAGTGGTTATCTAGCTCTTTTCTTTCTGCAG GTA GAG
Arg Leu Thr Lys Glu Phe Ser Val Tyr Met Thr Lys Asp Gly Arg Ile Ser Val Ala Gly Val Thr Ser Gly Asn Val Gly Tyr Leu Ala
CGG CTG ACC AAG GAG TTC TCG GTC TAC ATG ACA AAG GAT GGC CGA ATC TCC GTG GCA GGG GTC ACC TCT GGC AAT GTG GGC TAC CTT GCC
His Ala Ile His Gln Val Thr Lys
*
CAT GCC ATT CAC CAG GTC ACC AAG TAA TTCCCAAGTGCAAAGGAACAGAGACCACTTTCCCGACAGCCTTTGCGCTCGTGAGCGTCACGTGCAGGGTGAGGGAGGGTGGGT
G
GGTGGTGAATAGATCCGGTTTCCAACCACGGTGCATAACTCAGCGACTGAACTCGCTCCTCGGAAGAGAGGTAGGGCAGAGGCTTCCTCGGCTGGTATCTGGAACTTCGTCGGCTCTAAA
CCAAACTCCCTCATCCTTTTGTCTCCAGCTTTTCTGAAAGTTTACACACGCAAGAAAAATCACAGCACCAAACACCTGTCAGCCATGGCACTGGAATAGG 620 bases
CACCATCTGCTCTAATCATGTAGACGTACTGCCGCCTGGTTTCTCTGTTACAATAAAATTACTATAGACCCAGCCACTGTCTGCTTTTCTACTTACTGAGAGGGAAGGGGTAAGAGTGGA -3
203 A. Structure and Chemistry of Nucleotides
The first successes in using virus-like particles to carry
new pieces of DNA into the cells to help correct these
defects have been reported. Our ability to breed new
varieties of plants and microorganisms has been enor-
mously enhanced. We can foresee the production of
artificially designed enzymes to conduct many industrial
chemical processes. These are among the many reasons
for the excitement today in the fields of nucleic acid
chemistry and molecular genetics.
A. Structure and Chemistry of Nucleotides
1. Names and Abbreviations
The purine and pyrimidine ring compounds
found in nucleic acids are known as bases, even
though some of them have almost no basic character.
Nucleosides are the N-glycosyl derivatives of the bases
with ribose or 2-deoxyribose. The nucleotides are
phosphate esters of nucleosides. Similar names are
applied to related compounds such as adenosine tri-
phosphate (ATP) that are not present in DNA or RNA.
The names of the principal nucleotides from which the
nucleic acids are formed are given in Table 5-1. The
TABLE 5-1
Names of Pyrimidine and Purine Bases, Nucleosides, and 5'-Nucleotides
a
a
From Biochemical NomenclatureLibecq, C., ed.
25
b
Isomers of the 5'-nucleotides, in which the phosphate is attached to the oxygen on C-3', are the 3'-nucleotides. Care must be taken to avoid
ambiguity. The simple abbreviations UMP, CMP, AMP, and GMP always refer to the 5'-nucleotides.
These contain 2-deoxyribose and the nucleosides and
nucleotides are called deoxyadenosine (dAdo or dA), deoxy-
adenosine 5'-phosphate (dAMP), etc.
DNA contains thymine (Thy) rather than uracil. The
deoxyribose derivatives are thymidine (dThd or dT) and
thymidine 5'-phosphate. The ribose derivatives of thymine
are the nucleoside ribosylthymidine (Thd) and ribosylthy-
midine 5'-phosphate (Thd-5'-P).
U,T,C,A,G Uracil, thymine, cytosine, adenine, guanine
Y or Pyr Pyrimidine (T or C)
R or Pur Purine (A or G)
M Amino base (A or C)
K Keto base (G or T)
S Strongly pairing (G or C)
W Weakly pairing (A or T)
H Not G (any other base)
B Not A
V Not T or U
D Not C
N Any base
Base: Uracil (Ura) Cytosine (Cyt) Adenine (Ade) Guanine (Gua)
Nucleoside: Uridine (Urd or U) Cytidine (Cyd or C) Adenosine (Ado or A) Guanosine (Guo or G)
5'-Nucleotide: Uridine Cytidine Adenosine Guanosine
5'-phosphate 5'-phosphate 5'-phosphate 5'-phosphate
or or or or
5'-uridylic acid 5'-cytidylic acid 5'-adenylic acid 5'-guanylic acid
(Urd-5'-P or UMP) (Cyd-5'-P or CMP) (Ado-5'-P or AMP) (Guo-5'-P or GMP)
novice may find these confusing! Even worse than the
names in the table is hypoxanthine (Hyp), which is
derived from adenine by replacement of its NH
2
group with OH and tautomerization:
The nucleoside formed from hypoxanthine and ribose
is known as inosine (Ino or I) and the corresponding
nucleotide as inosinic acid. Further substitution at
C-2 of H by OH and tautomerization yields xanthine
(Xan). Its nucleoside is xanthosine (Xao, X). A similar
hydroxylation at C-7 converts xanthine to uric acid,
an important human urinary excretion product derived
from nucleic acid bases.
A Nucleotide units of RNA (abbreviations in parentheses)
b
B Nucleotide units of DNA C Abbreviations used in describing polynucleotide
sequences
C N
OH
C N
O H
N
N
N
N
O
O
H
H
N
N
N
N
O
O
H
H
O
Xanthine Uric acid
H
H H
204 Chapter 5. The Nucleic Acids
Nucleic acid structures are abbreviated in several
ways. For example, the sugar rings may be portrayed
by vertical lines. The abbreviations A, C, U, T, and G
for the individual bases or Pu (purine) and Py (pyrimi-
dine) are placed at the upper ends of the lines, and
slanted lines with P in the centers represent the 3' 5'
phosphodiester linkages in a polynucleotide.
The same structure can be further abbreviated
5' end pApUpGpRpY 3' end
or 5' end A U G R Y 3' end
Here purine is abbreviated R and pyrimidine Y. By
convention the 5' end of a polynucleotide is ordinarily
placed to the left in these formulas. Lengths of nucleic
acid chains are usually given as a number of bases or
kilobases (kb). For double-stranded DNA (dsDNA)
the length is given as base pairs (bp), kilobase pairs
(kbp), or megabase pairs (Mbp). However, in most
places, including this book, the abbreviations kb and
Mb will be used for a length of DNA whether single
or double stranded.
For double-stranded DNA, one strand, usually the
coding strand, from which the amino acid sequence
can be read using the code in Tables 5-4 or 5-5 (Section
C), has the 5' end at the left while the complementary
strand has the 3' end at the left, e.g.,
2. AcidBase Chemistry and Tautomerism
The ionized phosphate groups of the polymer
backbone give nucleic acid molecules a high nega-
tive charge. For this reason DNA in cells is usually
associated with basic proteins such as the histones
or protamines (in spermatozoa), with polycations of
amines such as spermidine (H
3
+
NCH
2
CH
2
CH
2
CH
2
NH
2
+
CH
2
CH
2
CH
2
NH
3
+
), or with alkaline earth cations
such as Mg
2+.
If the pH of a solution containing double-
helical DNA is either lowered to ~3 or raised to ~12,
the two strands unravel and can be separated.
Over the entire range of pH the alternating sugar-
A
P
U
P
G
P
Pu
P
Py
OH P
Terminal 5' -phosphate Terminal 3' OH
3'
5'
G C C T A T G G A C C T T T C T G C C A G T G A
C G G A T A C C T G G A A A G A C G G T C A C T
5'
3'
Ala Tyr Gly Pro Phe Cys Termination
3'
5'
Gln
phosphate backbone of the polymeric chains remains
negatively charged. However, depending on the pH,
the bases can be protonated or deprotonated with a
resultant breaking of the hydrogen bonds that hold
the pairs of bases together.
26,27
Pyrimidines and purines, which contain the NH
2
group, are weakly basic. The cationic protonated con-
jugate acid forms of cytidine, adenosine, and guanosine
have pK
a
values of 4.2, 3.5, and 2.7, respectively. Similar
values are observed for the 5'-nucleotides. In these
compounds it is not the NH
2
group that binds the
proton but an adjacent nitrogen atom in the ring (Eq.
5-3).
We can understand this if we recognize that the
bases have substantial aromatic character.
26,28
In ani-
line (aminobenzene) electrons are withdrawn from
the amino group into the aromatic ring with a strong
decrease in basicity of this NH
2
group (the pK
a
is 4.6).
Similarly, electrons are withdrawn from the NH
2
groups
of cytosine, adenosine, and guanine into the pyrimidine
and purine rings as is indicated by the small curved
arrows on the left-hand structure of Eq. 5-3. The effect
is even stronger than in aniline, largely because of the
presence of the nitrogen atoms in the rings. In cytosine
it is primarily N-3 that serves as the electron acceptor.
As a consequence this nitrogen becomes more basic
than the NH
2
group and is the major site of protona-
tion. However, as is indicated in Eq. 5-3, the positive
charge on the cation is shared by resonance with the
exocyclic amino group.
Adenosine is similar to cytosine in its acidbase
chemistry; N-1, adjacent to the NH
2
group, is the
principal site of protonation. A tautomer of the cation
protonated at N-3 is formed in smaller amounts. Gua-
nosine is electronically more complex, being protonated
mainly at N-7 and to a lesser extent at N-3
29
. This can
be understood in terms of electronic interaction with
the adjacent oxygen as indicated in the resonance
structure to the right in the following diagram:
N
N O
NH
2
Ribose
N
N O
NH
2
Ribose
N
N O
Ribose
N H
2
H
H
pK
a
= 4.2
+
H
+
+
H N
N
N
N
O
H
2
N
Ribose
H
H N
N
N
N
O

H
2
N
Ribose
H
+
+
+
3
7
(5-3)
205
Under basic conditions the proton on N-3 of uridine
or thymine or on N-1 of guanosine can dissociate with
a pK
a
of ~9.2. These bases are actually weak acids!
Look at the Kekule resonance
structure shown in the center of
Eq. 5-4. The two negative charges
flanking the dissociable proton are
sufficient to hold it firmly to the
ring at low pH. The proton is half
dissociated only when the pH is
raised to 9.2. At still higher values of
pH (pK
a
~12.4) a proton dissociates
from a ribose hydroxyl group.
The tautomerism of pyrimidines
was discussed in Chapter 2, Section
A,6. The tautomeric forms shown
in Fig. 5-1 predominate. However,
it is possible that minor tautomers
such as the following are sometimes
preferentially bound into active
sites of enzymes where the dielectric
constant may be low and where the
geometrical arrangement of amino
acid functional groups may favor
protonation on oxygen rather than
nitrogen.
30,31
3. Absorption of Ultraviolet
Light
Nucleic acids strongly absorb
ultraviolet light of wavelengths below
about 300 nm, with an absorption
maximum at ~260 nm and a stronger
one below 200 nm. This property is
HN
N O
O
Ribose
N
N O
O
Ribose
N
N O
Ribose
O
H
+

H
+
+

(5-4)
N
N O
Ribose
OH
A minor tautomer of uridine
biologically important because of the resultant
induction of mutations, a natural result of exposure
to sunlight. In the laboratory the same property is
useful in identification and quantitative analysis of
nucleotides. The ultraviolet spectra of four nucleo-
sides are shown in Fig. 5-5. Notice the changes that
accompany protonation or deprotonation of the ring.
The absorption bands, which are related to those of
benzene
26,32
(see Chapter 23), provide another indica-
tion of the partial aromatic character of the bases.
Chapter 23 also provides information on photo-
chemical reactions of the pyrimidines.
30 34 38 42 46 50
Wave number (cm
1
) 10
3
300 250 220 200
0
4
8
16
12
20
M
o
l
a
r

e
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t


1
0

3
Cytidine
30 34 38 42 46 50
Wave number (cm
1
) 10
3
300 250 220 200
0
4
8
16
12
20
Adenosine
1
2
5
6
Wavelength (nm)
Wavelength (nm)
30 34 38 42 46 50
Wave number (cm
1
) 10
3
300 250 220 200
0
4
8
16
12
20
Uridine
30 34 38 42 46 50
Wave number (cm
1
) 10
3
300 250 220 200
0
4
8
16
12
20
Guanosine
3
4
7
8
Wavelength (nm)
Wavelength (nm)
M
o
l
a
r

e
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t


1
0

3
Figure 5-5 Near ultraviolet absorption spectra of cytidine, uridine, adenos-
ine, and guanosine. 1. Monoprotonated form of cytidine (for which pK
a
= 4.2).
2. Neutral form (pH ~ 7) of cytidine. 3. Neutral form of uridine (for which pK
a
= 9.2). 4. Monoanionic form of uridine. 5. Monoprotonated form of adenosine
(pK
a
= 3.5). 6. Neutral form of adenosine. 7. Neutral form of guanosine
(pK
a
= 9.2). 8. Monoanion of guanosine.
A. Structure and Chemistry of Nucleotides
206 Chapter 5. The Nucleic Acids
BOX 5-A THE ALKALI METAL IONS
The many negative charges along a nucleic acid
backbone interact with all of the cations in a cell. This
box discusses some of these ions with emphasis on the
group IA metal ions. Although sodium and potassium
occur in similar amounts in the crust of the earth, living
cells all accumulate potassium ions almost to the exclu-
sion of sodium.
a-c
Sodium ions may be required only by
certain marine organisms and by multicellular animals
that regulate their internal body fluids. Most nonmarine
plants have no demonstrable need for sodium.
The tendency to accumulate K
+
is even more remark-
able since seawater is ~0.46 M in Na
+
and only 0.01 M in
K
+
. Other alkali metals occur in even smaller amounts,
e.g., 0.026 mM Li
+
, 0.001 mM Rb
+
, and a trace of Cs
+
.
Soil water is ~0.1 mM in K
+
and 0.65 mM in Na
+
. Again,
strong discrimination in favor of potassium is observed
in uptake by plants.
Intracellular concentrations of K
+
range from 200
mM in E. coli and 150 mM in mammalian muscle to ~30
mM in freshwater invertebrates such as clams, hydra,
and some protozoa. While K
+
cannot be replaced by
Na
+
, a partial replacement by Rb
+
and to a lesser extent
by Cs
+
is usually possible. In many microorganisms
rubidium can almost completely replace potassium, and
even a rat can survive for a short time with almost com-
plete substitution of K
+
by Rb
+
. Protons replace most K
+
in brown algae.
d
The human nutritional requirement
for potassium is high, amounting to ~2 g/day. Present
populations may suffer a chronic deficiency of potassium
as a result of food processing and boiling of vegetables.
e
Sodium is also essential to higher animals, and rats
die on a sodium-free diet. The sodium content of cells
varies among species, but it is usually no more than 0.1
0.2 times that of K
+
. A measurement of the [Na
+
]
within heart cells gave a concentration of ~9 mM, which
was increased by a factor of ~2.5 in a low Ca
2+
insulin-
containing medium.
f
In this measurement the NMR
resonance of the abundant external Na
+
was shifted by
use of a paramagnetic reagent (e.g., a dysprosium (III)
complex), that remained outside the cell. The signal
from the internal Na
+
was then seen clearly. In blood,
the relationship between Na
+
and K
+
concentrations is
reversed from that within cells. Human plasma is 0.15
M in Na
+
and 0.005 M in K
+
. Curiously, the taste for salt
in the diet appears to be largely an acquired one.
g
It is not immediately obvious why K
+
is the preferred
counterion within tissues, but a fundamental reason
may lie in the differences in hydration between Na
+
and
K
+
(Chapter 6). On the other hand, the relationship of
these ions to the excitability of membranes (Chapter 30)
may be of paramount importance, even in bacteria. The
concentration differences in the two ions across mem-
branes represent a readily available source of Gibbs
energy for a variety of membrane-associated activities.
Cells actively pump Na
+
out and K
+
into cells (Chapter 8).
Many intracellular enzymes require K
+
for activity.
b,c
These include those promoting phosphorylation of
carboxyl groups or enolate anions and elimination reac-
tions yielding enols as well as some enzymes dependent
upon the coenzyme pyridoxal phosphate.
hj
In all of
these enzymes NH
4
+
, Rb
+
, or Tl
+
can usually replace K
+
.
This permits study of the binding site for Tl
+
by the very
sensitive
205
Tl NMR spectroscopy.
k
The discovery that
K
+
is preferentially bound in some tetraplex DNA struc-
tures (see Fig. 5-8) further emphasizes the significant
difference in biological properties of the alkali metal
ions. Various synthetic macrocyclic compounds are also
able to selectively bind specific alkali metal ions.
l
The following tabulation gives concentrations not
only of K
+
and Na
+
but also of the other principal ionic
constituents in human blood plasma and within cells of
skeletal muscle.
m
Units are mmol/kg H
2
O.
a
Kernan, R. P. (1965) Cell K, Butterworth, London
b
Suelter, C. H. (1974) in Metal Ions in Biological Systems, Vol. 3
(Sigel, H., ed), pp. 201251, Dekker, New York
c
Suelter, C. H. (1970) Science 168, 789795
d
Steinbach, H. B. (1962) Comp. Biochem. Physiol. 4, 677720
e
Weber, C. E. (1970) J. Theor. Biol. 29, 327328
f
Wittenberg, B. A., and Gupta, R. K. (1985) J. Biol. Chem. 260,
20312034
g
Kaunitz, H. (1956) Nature (London) 178, 11411144
h
Toney, M. D., Hohenester, E., Cowan, S. W., and Jansonius, J. N.
(1993) Science 261, 756759
i
Antson, A. A., Demidkina, T. V., Gollnick, P., Dauter, Z., Von
Tersch, R. L., Long, J., Berezhnoy, S. N., Phillips, R. S.,
Harutyunyan, E. H., and Wilson, K. S. (1993) Biochemistry 32,
41954206
j
Metzler, C. M., Viswanath, R., and Metzler, D. E. (1991) J. Biol.
Chem. 266, 93749381
k
Markham, G. D. (1986) J. Biol. Chem. 261, 15071509
l
Christensen, J. J., Hill, J. O., and Izatt, R. M. (1971) Science 174,
459467
m
Composite data from Muntwyler, E. (1968) Water and Electrolyte
Metabolism and AcidBase Balance, p. 14. Mosby, St. Louis,
Missouri; White, A., Handler, P., and Smith, E. L. (1973)
Principles of Biochemistry, 5th ed., p. 802. McGraw-Hill, New
York; Long, C. (1961) Biochemists Handbook, p. 670. Van
Nostrand, Princeton, New Jersey. Reported ranges for some
constituents are very wide.
Skeletal muscle
* Milliequivalents/kg H
2
O

Phosphates and other nonprotein anions.


Na
+
150 14
K
+
5 150
Mg
2+
0.9 8
Ca
2+
2.5 1
Cl

105 16
HCO
3

27 10
Proteins

17
*
50
*
Other anions

6 146
Ion Blood plasma (intracellular)
207
4. Chemical Reactions of the Bases
The purines and pyrimidines are relatively stable
compounds with considerable aromatic character.
Nevertheless, they react with many different reagents
and, under some relatively mild conditions, can be
completely degraded to smaller molecules. The chem-
istry of these reactions is complex and is made more so
by the fact that a reaction at one site on the ring may
enhance the reactivity at other sites. The reactions of
nucleic acids are largely the same as those of the indi-
vidual nucleosides or nucleotides, the rates of reaction
are often influenced by the position in the polynucleo-
tide chain and by whether the nucleic acid is single or
double stranded. The reactions of nucleosides and
nucleotides are best understood in terms of the electronic
properties of the various positions in the bases.
26,33
Most of the chemical reactions are nucleophilic addition
or displacement reactions of types that are discussed
in Chapters 12 and 13.
Positions 2, 4, and 6 of pyrimidine bases are defi-
cient in electrons and are therefore able to react with
nucleophilic reagents. The 6 position is especially
reactive toward additions, while the 2 position is the
least reactive. The corresponding electron-deficient
positions in the purine bases are 2, 6, and 8. These
positions, which are marked by asterisks on the fol-
lowing structures, have electrophilic character in all
of the commonly occurring pyrimidines and purines.
All of the oxygen and nitrogen atoms in the pyrimidines,
as well as the 5 position of the ring, have nucleophilic
character and can therefore react with electrophilic
centers of various reagents. A number of specific reac-
tions that have been found useful to biochemists are
described in Section H,3.
5. Base Pairs, Triplets, and Quartets
The purine and pyrimidine bases are the side
chains of the nucleic acids. The polar groups that are
present in the bases can form hydrogen bonds to other
nucleic acid chains, e.g., in the base pairs of the DNA
double helix, and also to proteins. Figure 5-6 shows
the shapes and the hydrogen bonding groups avail-
able in the bases. The number of both electron donor
groups and proton donors available for hydrogen
bonding is large and more than one mode of base pair-
ing is possible.
C O, NH, and NH
2
H N
N
O
O
R
4
N
N
N
N
R
NH
2
5
6
*
*
*
1
*
6
2
3
9
8
7
*
*
H H
Adenine
N
H N
O
N
H
H
H
Cytosine
Guanine
N
N
N
H
N
H
N
N
N
N
H
N
N
O
H
H
H
N
H N
O
O
H
Uracil
Thymine
H
Figure 5-6 Outlines of the purine and pyrimidine bases of
nucleic acids showing van der Waals contact surfaces and
some of the possible directions in which hydrogen bonds
may be formed. Large arrows indicate the hydrogen bonds
present in the WatsonCrick base pairs. Smaller arrows
indicate other hydrogen bonding possibilities. The directions
of the green arrows are from a suitable hydrogen atom in the
base toward an electron pair that serves as a hydrogen acceptor.
This direction is opposite to that in the first edition of this book
to reflect current usage.
The base pairs proposed by Watson and Crick are
shown in Fig. 5-2 and again in Fig. 5-7. While X-ray dif-
fraction studies indicate that it is these pairs that usually
exist in DNA, other possibilities must be considered.
For example, Hoogsteen proposed an alternative A-T
pairing using the 6-NH
2
and N-7 of adenine.
34
Here
the distance spanned by the base pair, between the
C-1' sugar carbons, is 0.88 nm, less than the 1.08 nm
of the WatsonCrick pairs. Duplexes of certain sub-
stituted poly (A) and poly (U) chains contain only
Hoogsteen base pairs
35
and numerous X-ray structure
determinations have established that Hoogsteen pairs
A. Structure and Chemistry of Nucleotides
208 Chapter 5. The Nucleic Acids
N
N
O
O
CH
3
H
H
H
N
N
N
N
N
N
T
A
N
CH
3
O
O
H
N
O
N
N
N
N
N
N
N
N
N
N
H
H
H
H
H
H
H
H
O
O
C
G
C
Watson-
Crick
pair
T*
+
H
o
o
g
s
t
e
e
n
p
a
i
r

+
+
+
H
o
o
g
s
t
e
e
n
p
a
i
r
Watson-
Crick
pair
Minor groove
1.08 nm
+
Figure 5-7 Two base triplets that form in triple-stranded
DNA and involve both WatsonCrick and Hoogsteen base
pairing. (A) The triplet T
*
AT, where the T (marked T
*
) of
the third strand is hydrogen bonded as a Hoogsteen pair
( ) to an adenine of a WatsonCrick AT pair (whose
hydrogen bonds are indicated ( ). (B) The triplet
C
+
*
GC, where C
+
is cytosine in its N-1 protonated (low pH)
form. The WatsonCrick strands are antiparallel, as indicated
by the + and signs. The third strand may have either
orientation, but when it contains largely pyrimidines it is
parallel to a purine-rich strand. An example is shown in
Fig. 5-24.
N
N N
H
H
N
N
H N
N
O
CH
3
O
0.88 nm
4
Hoogsteen A
*
T base pair
Figure 5-8 A guanine quartet held together by Hoogsteen
base pairing. This structure is found in the telomeres at the
ends of linear chromosomes. Four segments of DNA, each
of which may be part of a single folded strand, (see p. 227)
give structures in which four or more of these G quartets are
stacked one above the other. Monovalent ions, usually K
+
or Na
+
, are bound in the center, although not always in the
plane of the bases. See Fang and Cech
38
and Gellert et al.
39
N
N
N N
O
R
H
N
N
N
N
O
R
H
N
H
H
H
N
N
N N
O
R
H
N H
H
H
N
N
N
N
O
R
H
N
H
H
H
N H
H
H M
+
do occur in true nucleic acids.
36
Figure 5-7 shows the
structures of two Hoogsteen pairs. In each case the
purine component has formed a WatsonCrick pair
with a third base to give a base triplet. The first of
these triplets may be designated T
*
AT, where
*
repre-
sents the Hoogsteen hydrogen bonding and repre-
sents the WatsonCrick bonding. The second triplet
in Fig. 5-7, C
+
*
GC, can form only with the N-1 proto-
nated form (low pH form) of cytosine.
Reversed WatsonCrick or Hoogsteen AU or AT
pairs are formed if the 2-carbonyl rather than the 4-car-
bonyl of the U or T makes a hydrogen bond with the
amino group of adenine.
27
Because of the resulting
arrangement of the ribose rings the base pairs cannot
fit into the ordinary WatsonCrick double-stranded
DNA structure. A reversed WatsonCrick pair can also
be formed between G and C but with only two hydro-
gen bonds, while a reversed Hoogsteen pair can form
only if a minor tautomer of cytosine is used.
Hoogsteen pairs were first observed in nature in
transfer RNA molecules (Fig. 5-31). These molecules
contain mostly WatsonCrick base pairs but there are
also two reversed Hoogsteen pairs. One of them, be-
tween U8 and A14, is invariant in all tRNAs studied.
Hoogsteen pairing also occurs in four-stranded DNA,
which has important biological functions. A G quartet
from a DNA tetraplex held together by Hoogsteen
base pairs is shown in Fig. 5-8.
A
B
209
Another pairing that occurs in tRNAs allows gua-
nine to pair with uracil, e.g., G4 with U69. This was
originally proposed to account for codonanticodon
interactions betweentRNA molecules and messenger
RNA (Chapter 29). It is commonly called wobble
pairing because the uracil must wobble away from
its orientation in the normal WatsonCrick pair.
27,37
Strengths of base pairs. How strong are the
bonds between pairs of bases in DNA? The question
is hard to answer because of the strong interaction of
the molecules with polar solvents through hydrogen
bonding and hydrophobic effects. Some insight has
come from studies of the association of bases in non-
polar solvents. Thus, 1-cyclohexyluracil forms a dimer
involving either hydrogen bonding or stacking, but
the association is weak with the Gibbs energy of for-
mation G
f
= 5 kJ mol
1
. When the same compound
was mixed with 9-ethyladenine, a base-paired complex
formed between the two compounds with a formation
constant over ten-fold greater than that for the dimer
(Eq. 5-5).
40
When the circled hydrogen atom in Eq. 5-5
was replaced by CH
3
, which blocked the pairing, K
f
fell below 1 kJ/mol (G > 0). The difference in G
f
in
the two cases was only 7 kJ mol
1
. Many other estimates
have been made.
40a,40b
The small energies summed
over the many base pairs present in the DNA molecule
help provide stability to the structure.
Stacking of bases. The purines and pyrimidines
of nucleic acids, as well as many other compounds
with flat ring structures and containing both polar and
nonpolar regions, are sparingly soluble in either water
Cyclohexyl
Cyclohexyl
Dimer
Complex (base pair)
K
f
= [complex]/
= 100 l mol
1
G 11 kJ mol
1
[1-cyclohexyluracil][9-ethyladenine]
K
f
= [dimer]/
= 6 l mol
1
G 5 kJ mol
1
[cyclohexyluracil]
2
N
N
O
O
N
N
N
N
N
Ethyl
H
H
H
N
N
O
O
H
N N
N
N
N
H
H
+
Ethyl
(5-5)
or organic solvents. Molecules of these substances
prefer neither type of solvent but adhere tightly to
each other in solid crystals. Both experimental mea-
surements and theoretical computations
41
suggest that
hydrogen bonding is the predominant force in the
pairing of bases in a vacuum or in nonpolar solvents.
However, in water stacking becomes important.
42
In a
fully extended polynucleotide chain consecutive bases
are 0.7 nm apart, twice the van der Waals thickness of
a pyrimidine or purine ring, but in double-helical DNA
of the B type (Figs. 5-3, 5-12) the distance between con-
secutive base pairs is only 0.34 nm. They are touching.
One effect of stacking is a decrease in the expected
intensity of light absorption. The molar extinction co-
efficient of a solution of double-helical DNA or RNA
is always less by up to 2030% than that predicted
from the spectra of the individual nucleosides (Fig. 5-5).
This hypochromic effect is considered further in
Chapter 23.
Because both hydrogen bonding and stacking
are involved, the thermodynamics of base pairing in
nucleic acids is complicated.
43
The hydrophobic parts
of exposed bases tend to induce an ordering of the sur-
rounding water molecules and therefore a decrease in
their entropy. However, hydrogen bonding of the polar
groups of the bases to the solvent causes a decrease in
water structure. This is greater than the increase in
structure around the hydrophobic regions and the
stacking of bases leads to a net decrease in entropy.
The entropy change S for addition of a base pair to
the end of a double-stranded RNA helix in a hairpin
loop such as that displayed in Fig. 5-9 ranges from
0.05 to 0.15 kJ/degree per base pair.
44,45
The enthalpy change H tends to be small and
positive for association of alkyl groups in water and
nearly zero for association of aromatic hydrocarbons
N
N
N
N N
H
H N
O H
N
O
A14
U8
2
A reversed
Hoogsteen base
pair present in
tRNA molecules
N
N
H
H
O
N
N
N
H
H
O
N
N
O
G4
U69
Awobble base pair
A. Structure and Chemistry of Nucleotides
210 Chapter 5. The Nucleic Acids
to G
f
. The exact amount depends upon whether an A
or a U is at the 5' end in the existing helix. If an AU pair
is added to the helix terminating in CG or GC, about
9 kJ/mol is added. Larger increases in G
f
result
from addition of GC pairs, which contain three hydrogen
bonds between the bases versus the two in AU pairs.
27,41,52
UG pairs provide a very small amount of stabiliza-
tion to an RNA double helix, while the presence of
unpaired bases has a destabilizing effect. The most stable
hairpin loops contain four or five bases. Depending upon
whether the loop is closed by CG or AU, the helix is
destabilized by 2030 kJ/mol. Bulge loops, which pro-
trude from one side of a helix, have a smaller destabiliz-
ing effect. An example of the way in which Table 5-2 can
be used to estimate the energies of formation of a loop in
a straight-chain RNA is illustrated in Fig. 5-9. Similar
analysis of base pairing in DNA can also be done.
5355
(Chapter 2). However, H is distinctly negative for
association of heterocyclic bases. This has also been
attributed to a decrease in the ordering of solvent
around the bases as a result of exclusion of water.
Attraction or repulsion of partial charges on the polar
groups comprising the purine and pyrimidine bases
may also be an important factor.
43,4648
For addition
of a base pair to an RNA helix, the change in enthalpy,
H, varies from about 24 to 60 kJ/mol.
44,45
Since G = H TS, the net result is a negative
value of G, a hydrophobic effect that favors asso-
ciation of bases. Substantial efforts have been made to
estimate quantitatively the Gibbs energies of formation
of helical regions of RNA molecules in hairpin stem-
loops such as that of Fig. 5-9.
44,45,4951
Table 5-2 shows
the observed increments in G
f
of such a helix upon
addition of one base pair at the end of an existing
helix. Addition of an AU pair supplies only 4 to 5 kJ
Figure 5-9 The contribution of base-
paired regions and loops to the Gibbs en-
ergy of a possible secondary structure for
a 55 base fragment from R17 virus. The
stem-loop structure shown here is part of
a larger one considered by Tinoco et al.
58
U
A
A
G
G
U
U
A
A
A
A
U
U
C
C
A
A
U
U
U
A
A
C
U
A
U
G
U
A
C
G
A
A
C
C
C
G
C U
5' 3'
C
A
U
G
C
U
U
G
G
G
U
U
C
+25 kJ mol
1
+ 8
+ 8
21
0
C
7.5
5.0
9
21
9
5.0 + 12
7.5
5.0
5.0
9 + 13
7.5
9
5.0
9
9
13
= 91 kJ mol
1
Hairpin
loop
Bulge
loop
Internal
loop
U
U
Hairpin loops of 4 or 5 bases
Closed by GC +5 +21
Closed by AU +7 +29
Bulge loops
1 base +3 +12
47 bases +5 +21
1.2
1.8
2.2
3.2
5.0
0.3
TABLE 5-2
Gibbs Energies of Formation G
f
at 25C for Addition of One
Base Pair to an Existing RNA Helix
a,b
Base pair at end
of existing helix
Base pair
added
G
f

(kcal mol
1
10%)
G
f

(kJ mol
1
10%)
a
Table modified from that of Tinoco et. al.
58
b
All base pairs in the table are oriented as follows:
c
G
f
is the same for added to an end.
d
G
f
is the same for or added to an end.
A

U
5.0
7.5
9
13
21
1
A

U
A

U
A

U
U
c

A
C

G
C

G
G

C
G

C
G

U
U

G
or
G
d

C
C

G
or
G

C
U

A
A

U
C

G
G

C
A

U
5'A3'

3'U5'
211
Tautomerism and base pairing. Tautomerism
has an interesting relationship to the formation of the
pairs and triplets of hydrogen bonds in DNA or RNA.
Each base exists predominately as one preferred
tautomer, but at any moment a very
small fraction is present as less stable
tautomers. Watson and Crick sug-
gested that this fact may be respon-
sible for the occurrence of some
mutations.
56,57
Thus, tautomer B of
Eq. 5-6 would not be able to pair with
thymine, its proper pairing partner,
but could pair with cytosine.
Similarly, tautomer B of uracil
in Eq. 2-4 could pair with guanine
instead of its proper partner adenine.
If a similar event occurred to an AT
pair during gene replication an in-
correct copy of the gene, differing
in a single code letter, would be
formed. However, because the tau-
tomerism is affected so strongly by
the environment (Chapter 2), the ex-
tent to which it may cause mispairing
while replication enzymes act is un-
certain.
59
Both bases in a pair could
be tautomerized by synchronous
transfer of protons in two parallel
hydrogen bonds. However, theo-
retical calculations predict a high
energy barrier to this process.
60
Proton transfer, which can also be
induced by light, has been studied
on a femtosecond scale.
61
After
photochemical transfer of one proton a second is trans-
ferred within a few picoseconds.
6. Conformations of Nucleotides
The furanose ring of ribose or deoxyribose is flexible
and can be interconverted smoothly among an infinite
number of envelope (E) and skew or twist (T) confor-
mations. See Chapter 4, Section A,2. However, there
are limits set by steric and anomeric effects.
27,6264
Conformations are often described as in Fig. 5-10 by
stating which atom in an envelope conformation lies
mostly out of the plane of the other four atoms. If this
atom lies above the ring, i.e., toward the base, the ring
A. Structure and Chemistry of Nucleotides
N
N
N
H
N
N
H H
N
N
N
H
N
H
N
H
A B
(5-6)
Figure 5-10 Conformational properties of nucleosides. (A) Representations of
several conformations of a ribose or deoxyribose ring in a nucleoside. (B) View
down the NC axis joining a purine base to the sugar in a nucleoside in a syn
conformation. The atom marked C4 is in the six-membered ring, which protrudes
further over the sugar ring. The angle is measured from 0 to 180. Syn confor-
mations are those for which falls in the region of the heavy semicircle. (C) An
anti conformation of a pyrimidine nucleoside. Other anti conformations fall in the
region of the heavy semicircle. (D) Labeling of the conformational angles of the
main chain in a polynucleotide.
25,27,37,37a
See also Fig. 2-2.
syn anti
O
H
2
C
O
P
i+1
O
P
i
Base
()

()
()
(')
(')
(')
3' 2'
1'
Direction
4'
5'
5' to 3'
of chain
C
4

P
u
r
i
n
e
2'
3'
4'
04' 60
120
180
-120
-60
0
sp
-sc
-ac
ap
ac
sc
high
syn

P
y
r
i
m
i
d
i
n
e

C
2
2'
3'
4'
04' 60
120
180
-120
-60
0
high
anti
0

A
B C
C5'
04'
2'
3'
B
2
E
C5'
2'
3'
B
2
T
3
*
C5'
2'
3' B
3
T
2
C(2) exo-C(3)endo
C5'
B
C5'
B
Superimposed silhouettes
Twist conformations
C(2)-endo-C(3)-exo
C5'
04'
2'
3'
B
2
E
C5'
04'
2'
3'
B
2
E
C5'
04'
2'
3'
B
2
E
C(3)-endo C(2)-endo C(3)-exo C(2)-exo
Envelope conformations
*This may be designated
3
T if twist is symmetric
2
D
212 Chapter 5. The Nucleic Acids
Figure 5-11 Views of a nucleoside in syn and anti conformations.
66
(C) Courtesy of Dr. Muttaiya Sundaralingam.
syn anti
syn anti
Pyrimidine nucleosides A
Purine nucleosides B
C Ball and stick models of purine nucleosides
syn
N
N
NH
2
O
O
OH OH
HOCH
2
anti
N
N
NH
2
O
O
OH OH
HOCH
2
anti
HOCH
2
N
N
NH
2
O
OH OH
N
N
syn
N
N
NH
2
O
OH OH
HOCH
2
N
N
conformation is known as endo; when below the ring,
it is known as exo. The C(2')-endo (
2
E) and C(3')-endo
(
3
E) conformations are most commonly approximated
in nucleotides and nucleic acids.
65
The C(3')-exo con-
formation is designated E
3
, the twist conformation
C(2')-endo-C(3')-exo as
2
T
3
, etc. A conformation can be
specified more precisely by the five torsion angles
0
to
4
(which have also been designated
0
to
4
). All of the
envelope and twist conformers can be interconverted
readily. The interconversions can be imagined to occur
in a systematic way by pseudorotation, a rotation of
the pucker around the sugar ring. In a commonly used
convention a pseudorotation phase angle P ranges from
0 to 360 as the pucker moves twice around the ribose
ring to restore the original conformation. P is taken as 0
for the symmetric
3
T
2
twist, 18 for E
2
, +18 for
3
E, +54
for
4
E, +198 for E
3
, etc. The five torsion angles
j
, for j
= 0 4 as defined in the foregoing diagram, are related
to P as follows:

j
=
max
.
cos[P + j
.
]
where = 720/5 = 144. The maxi-
mum torsion angle,
max
, is about
40. See Saenger
27
for details.
Conformational alterations of
ribose and deoxyribose rings can
occur within polynucleotides and
are of biochemical importance. An
interesting consequence of changes
in ring conformation is that the dis-
tance between the C(5') and N atoms
attached to the sugar ring of a nucleo-
side may vary by as much as 0.05
nm (see Fig. 5-10A). The orientation
of a base with respect to the sugar is
specified by the angle . In one con-
vention (Fig. 5-10B,C) its zero value
is taken as that in which the N(1)C(2)
bond of a pyrimidine or the N(9)C(4)
bond of a purine is cis to the C(1')
O(4') bond, [sometimes called the
C(1') O(1') bond]. However, other
definitions have been used.
27
Typi-
cal values of for nucleotides and
nucleosides vary between 75 and
165. In these anti conformations
the CO and NH groups in the 2 and
3 positions of the pyrimidine ring
(or in positions 1, 2, and 6 of the
purine ring) are away from the sugar
ring, while in the syn conformations
they lie over the ring. Anti confor-
mations are more often present than
syn in nucleic acids. A different
view of syn and anti pairs of nucle-
otides is shown in Fig. 5-11.
An additional five torsion angles
are needed to specify the backbone
conformation of a polynucleotide.
According to the convention adopted
by the IUB the six angles are desig-
RO CH
2
Base
OH
H
O
O
H
H
R'

1
(5-7)
213 B. Double Helices
nated , , , , , and , as is indicated in Fig. 5-10D.
25,27
In an older but much used convention,
66
starting at
any phosphorus atom the angles , , and specify
the next three torsion angles as one moves toward the
3' end of the chain, while , , and specify the angles
lying toward the 5' end (these are shown in parentheses
in Fig. 5-10D). Notice that () =
3
+ 120.
B. Double Helices
1. The B Form of DNA
This form of DNA, whose structure is depicted in
Figs. 5-3 and 5-12, is stable at high humidity and is
thought to approximate that of most DNA in cells.
27,37,67
If we look directly at the axis of the double helix and
perpendicular to one of the base pairs, and ignore the
fact that the base pair is asymmetric, we see that the
nucleotide unit in one chain is related to the nucleotide
unit lying across from it in the opposite plane by a two-
fold axis of rotation (dyad axis). This symmetry ele-
ment, which arises from the antiparallel arrangement
of the chains, makes the DNA molecules from the out-
side look nearly identical whether viewed from one
end or the other and whether viewed as a model by
the human eye or through contact with an enzyme
which might act on the molecule. Actually, the two
chains are not identical, and the genetic information can
be read off from the functional groups exposed in two
grooves in the surface of the helix (Figs. 5-3 and 5-7).
The broader groove in the form, which is referred to
as the major groove, is about 0.85 nm deep and 1.11.2
nm wide when allowance is made for the van der Waals
radii of the atoms. In some other forms of DNA the
major groove is narrow, but it can always be identified
by the larger of the arcs that can be drawn between the
two N C bonds of the nucleosidic linkages in a nucleo-
tide pair. The minor groove or narrow groove, which
is defined by the smaller arc between the two N C
bonds, is ~ 0.75 nm deep and 0.6 nm wide in B-DNA.
The diameter of the double helix of B-DNA, mea-
sured between phosphorus atoms, is just 2.0 nm. The
rise per turn, the pitch, is 3.4 nm. There are about
ten base pairs per turn (9.7 and 10.6 in two different
crystal forms).
68,69
Thus, the rise per base pair is 0.34
nm, just the van der Waals thickness of an aromatic
ring (Table 2-1). It is clear that the bases are stacked in
the center of the helix. A 1000-bp (1-kb) gene would be
a segment of DNA rod about 340 nm long, about 1/40
the length of the molecule in the electron micrograph
of Fig. 5-13.
As is appropriate for the cells master blueprint,
DNA in the double helix is stable. Factors contributing
to this stability are (1) the pairs and triplets of hydrogen
bonds between the bases; (2) the van der Waals
attraction between the flat bases which stack together;
(3) the fact that on the outside of the molecule are many
oxygen atoms, some negatively charged, which are
able to form strong hydrogen bonds with water, with
small ions, or with proteins that surround the DNA;
and (4) the ability to form superhelices (see Section
C,3). Nevertheless, the long DNA chains present in
our chromosomes are frequently broken and an elabo-
rate system of repair enzymes is needed to preserve
the reliability of this master code for the cell.
73,74
Today it is generally accepted that most DNA
exists in nature as a double helix resembling the B
form. However, doubts were expressed as recently as
1980. The reason for the uncertainty lay in the fact that
X-ray data from the stretched paracrystalline DNA
fibers used in earlier studies are much less precise than
those from true crystals.
10
The X-ray data for B-DNA
fibers could also be interpreted in terms of an alterna-
tive side-by-side structure which would permit
easier separation of strands during replication.
75
However, numerous high-resolution X-ray structure
determinations on single crystals of synthetic DNA
fragments have confirmed the double-helical structure
and the presence of WatsonCrick base pairs.
68,69,76 84
Similarly, fragments related to the double helices of
RNA have been crystallized and the structures deter-
mined to atomic resolution.
85 87
The right-handed
helical structure of DNA in solution has also been
confirmed by independent methods based on electron
microscopy,
88
scanning probe microscopy,
89,90
fluores-
cence resonance energy transfer,
91
and computation.
92
Dickerson and associates discovered
78,79
that
B-DNA has a spine of water in the minor groove
of regions rich in A-T base pairs. Two water molecules
per base pair are hydrogen bonded to form a long
chain. Half of the water molecules in the chain also
form hydrogen bonds to oxygen and nitrogen atoms
that are exposed in the minor groove while the others
hydrogen-bond to a second chain of water molecules,
forming a ribbon of hydration (Fig. 5-14A).
81,93-95
Additional water binds to polar groups in the major
groove. The hydration pattern is largely local, i.e.,
each base has characteristic hydration sites (Fig. 5-
14B).
94
Some of the hydration sites may be occupied
partially by the nomovalent ions, Na
+
or K
+
, depend-
ing upon the medium. Bound divalent metal ions
are also sometimes seen in X-ray structures.
81,83a
However, most cations are thought to remain mobile.
2. Other Double-Helical Forms of DNA
The B form of fibrous DNA is stable under condi-
tions of high (~ 93%) humidity but at 75% humidity it
is converted into A-DNA in which the base pairs are
inclined to the helix axis by about 13 and in which the
ribose rings are primarily C3'-endo rather than C2'-endo.
214 Chapter 5. The Nucleic Acids
Figure 5-12 (Left) Stereoscopic skeleton models of the B, A, and Z forms of double helical DNA. See Schlick.
70
(Center) End views (Right) Space-filling models of the same three DNA forms: B, A, and Z. Courtesy of Tamar Schlick.
Z-form
A-form
B-form
215
Figure 5-14 (A) Stereoscopic drawing
showing two layers of water molecules that
form a spine or ribbon of hydration in
the minor groove of B-DNA. The inner
layer is shown as larger filled circles; water
molecules of the outer layer are depicted
with smaller dots and are numbered.
Hydrogen bonds are shown as dashed
lines. (B) Electron density map. (A) and
(B) from Tereshko et al.
95
(C) Stereoscopic
representation of the superimposed electron
densities of 101 water molecules observed
to hydrate 14 guanine rings found in 14
B-DNA molecules for which high-resolution
X-ray structures were available. Positions
of 101 water molecules within 0.34 nm from
any atom of the 42 guanines are plotted.
From Schneider and Berman.
94
B. Double Helices
There are 10.9 base pairs per turn
93,96 98
(Fig 5-12; Table
5-3). In the A form the major groove is very deep (~ 1.35
nm) and narrower (~ 0.27 nm) than in the B form and
extensively hydrated.
99
The minor groove is wider
(~ 1.1 nm) and shallower (~ 0.28 nm) than in the B form.
The crystal structure for a form intermediate between
A and B has also been reported.
99a
Paracrystalline
forms of DNA known as B, C, and D have also been
observed
100,101
and others have been proposed.
102,103
The most interesting additional form, Z-DNA, was
discovered by X-ray studies of the alternating oligodeoxy-
ribonucleotides d(CpGp-CpGpCpG) and d(CpGpCpG).
104
The helix of Z-DNA is left-handed.
71,72,105112
The repeat-
ing unit consists of two WatsonCrick base pairs, the
backbone following a zigzag pattern (Fig. 5-12). There
are 12 base pairs per turn. The cytosine groups have
the usual anti conformation but the guanosine groups
are syn. Some of the ribose rings have the C2'-endo
conformation characteristic of B-DNA but some are
C3'-endo. The alternating CpG sequence is not essen-
tial for Z-DNA formation. However, alternating syn
and anti conformations are important and purines
assume the syn conformation more readily than do
pyrimidines. The major groove of Z-DNA is shallow
(Fig. 5-12), allowing it to accomodate bulky substituents
at C8 of purines or C5 of pyrimidines. Such substituents
favor Z-DNA.
Figure 5-13 Electron micrograph of a DNA molecule (from
a bacterial virus bacteriophage T7) undergoing replication.
The viral DNA is a long (~ 14 m) duplex rod containing
about 40,000 base pairs. In this view of a replicating molecule
an internal eye in which DNA has been duplicated is present.
The DNA synthesis was initiated at a special site (origin) about
17% of the total length from one end of the duplex. The DNA
was stained with uranyl acetate and viewed by dark field
electron microscopy. Micrograph courtesy J. Wolfson and
D. Dressler.
A
C
B
216 Chapter 5. The Nucleic Acids
The B form is the most hydrated and most stable
form of DNA under conditions of high humidity but
even in solution it can be converted to A-DNA and
Z-DNA by a high concentration of NaCl.
113
This is
presumably because the salt dehydrates the DNA.
Saenger and coworkers pointed out that the oxygen
atoms of successive phosphate groups in the polynu-
cleotide backbone of B-DNA are at least 0.66 nm apart,
too far apart to be bridged by a water molecule.
114
However, the phosphates are individually hydrated.
On the other hand, in A-DNA and Z-DNA the oxygen
atoms on successive phosphates are as close as 0.53 and
0.44 nm, respectively. This allows one H
2
O molecule
to bridge between two phosphates, stabilizing these
forms in environments of low humidity. This may be
one factor that affects the B to A and Z transitions.
115117
In the narrow Z-DNA helix repulsion between negative
charges on the phospho groups of opposite strands is
strong. By shielding these charges high salt concentra-
tions also help to stabilize Z-DNA.
Since the Z form of DNA is favored in regions rich
in G-C pairs,
118,119
it is reasonable to expect that it may
occur in nature. Antibodies have been prepared which
bind specifically to regions of DNA in the Z form and
have been used to identify many such regions.
111,120
Genetic studies in E. coli have also provided strong
evidence that left-handed DNA sequences are formed
in vivo in that bacterium.
121,122
Z-DNA-forming
sequences have also been found in the Halobacterium
genome.
119
Segments of Z-DNA may occur in control
regions called enhancers (Chapter 28)
121 124
and Z-DNA
may also form behind RNA polymerase molecules that
are moving along a gene while synthesizing mRNA.
The associated negative supercoiling of the DNA could
cause it to assume the Z form. This region of Z-DNA
can, in turn, be a site for interaction with specific
proteins.
In the WatsonCrick structure the two strands are
antiparallel, an essential for replication. However, stable
segments of double-stranded DNA with parallel strands
can also be formed and may occur in specialized regions
of the genome.
125 128
3. The Conformational Flexibility of
Double Helices
Local variations in the sequence of
nucleotides affect the conformation of a DNA
molecule and it is clear that the helix
is not uniformly coiled throughout the entire
length.
80,103,124,129134
While most helix seg-
ments probably have a right-handed twist
others may be left-handed. Most DNA is
probably in the B form but there are segments
in the A form. These may arise from formation
of hybrid duplexes with RNA, which assume
the A conformation and are also favored by
certain base sequences. Rules for predicting
the DNA conformation from the nucleotide
sequence have been proposed.
116,135
In the
simplest case
135
we consider each pair of
adjacent nucleotides in the double helix.
There are 16 possible pairs in one chain. These
can be designated as in the following examples:
(AA,TT), (CG,CG), and (AG,CT). The first
two letters within the parentheses represent
the sequence (from 5' to 3') in one chain while
the second pair of letters represent the sequence
(again from 5' to 3') in the complementary
chain. The rules state that (AA,TT) or (TT,AA)
repeated in a sequence will stabilize the B form
of DNA. Repetition of (CC,GG) or (GG,CC)
will favor conversion to the A form. Repeti-
tions of (CG,CG) favor the Z form, especially
if an alternating sequence of purines and
pyrimidines is present throughout the (G + C)-
rich region.
Even within the regular B structure the
TABLE 5-3
Helix Parameters for Three Types of DNA
a
Parameter B A Z
Helical twist,
degrees 2842 1644 GC 51 2
mean 36 4 33 6 CG 8.5 1
Base pairs per turn 10.0 11-12 12
(9.710.6)
Helix rise per
base pair, nm 0.34 .04 0.29 .04 GC 0.35 .02
CG 0.41 .02
Base inclination, degrees 2.0 5 13 2 8.8 .7
Propeller twist, degrees 12 5 15 6 4.4 3
Base roll degrees 1.0 5 6 5 3.4 2
Predominant
conformation of
deoxyribose C2-endo C3-endo
Depth of grooves (nm)
Major 0.85 1.35 very shallow
Minor ~0.75 ~0.28 very deep
Width of grooves (nm)
Major 1.11.2 0.27 broad
Minor 0.6 ~1.1 narrow
Form of DNA or RNA (A-form)
a
See R. E. Dickerson
78,78a
; based on single-crystal X-ray analysis.
C C2-endo
G C3-endo
217
torsion angles and (Fig. 5-10) are variable. Their
changes are highly correlated.
136,136a
As ranges from
140 to 90, varies between 80 and 160. Other pairs
of torsion angles are also correlated. Besides allowing
for changes between B, A, and Z conformations, this
flexibility of the DNA helix together with cooperativity
with adjacent base pairs may allow transmission of
conformational effects for some distances along a
DNA helix.
137,138
Supercoiling, discussed in Section
C,3, also affects the helical conformation.
DNA can be stretched into yet another form or
forms. Application of a force of 65 70 piconewtons
(pN) stretches B-DNA by 70%. This overstretched
DNA may also be important biologically.
130,139 141a
Rotational and translational movements of
bases. In considering the conformational flexibility of
a polynucleotide it is useful to define the parameters
associated with movement of base pairs or individual
bases. The possible movements are indicated in Fig.
5-15 and in Table 5-3. A long axis is established for
each base pair in a structure for which X-ray data are
available. Making use of these axes, we can move
along the helix and measure the angle of twist from
one base pair to the next.
142 145
While the mean value
Tip () Inclination ()
Opening () Propeller twist () Buckle ()
Twist () Roll () Tilt ()
y displacement (dy) x displacement (dx)
Stagger (Sz) Stretch (Sy) Shear (Sx)
Rise (Dz) Slide (Dy) Shift (Dx)
Coordinate frame
+y
+z
+x 3'
5'
5'
3'
Minor
groove
II
I
Coordinate frame
+y
+z
+x 3'
5'
5'
3'
Minor
groove
II
I
A Rotation B Translation
of the twist for B-DNA is 36, values of 28 42 have
been observed in structures of oligonucleotides deter-
mined by X-ray analysis. The base tilt, which is nearly
zero for B-DNA, is less variable. Within each base pair
there is also a propeller twist around the long axis.
146
It averages about 12 for B-DNA. In addition, the
whole base pair may roll, i.e., be rotated around its
long axis by several degrees. These motions relieve
steric interferences in the center of the helix, for example,
those that would arise between purines in adjacent
base pairs if the same helix parameters were imposed
on each base pair.
142,147
A more detailed analysis reveals
the large range of motions, mostly small, which are
described in Fig. 5-15.
145,148
The mathematics needed
to deal with computer-based modeling of polynucleo-
tide structures has been developed.
80,149,150
Its applica-
tion showed that there is a strong correlation between
twist and roll. Typical average values of base tilt, pro-
peller twist, and roll are given in Table 5-3 for B-, A-,
and Z-DNA.
Bends and bulges. If we overlook the ridges and
grooves on their surfaces the DNA structures shown in
Figs. 5-3 and 5-12 are straight rods. However, real DNA
rods are crooked and may contain distinct bends. The
Figure 5-15 (A) Drawing illustrating various rotational movements of bases in polynucleotides. Upper two rows: rotations of
two bases of a pair. Lower row: rotations involving two successive base pairs. (B) Translational movements. Upper two rows:
involving two bases of a pair. Lower row: two successive base pairs. From Diekmann.
145
B. Double Helices
218 Chapter 5. The Nucleic Acids
existence of bent DNA in nature was first discovered
from study of fragments enzymatically cut from kine-
toplast DNA of tropical parasites.
151,152
The bent frag-
ments moved anomolously slowly during electrophor-
esis in polyacrylamide gels of small pore size. Evidently,
the bent shape impedes movement through the pores.
Since the initial discovery, regions of bent DNA have
been found in origins of replication
152 154
and other
specialized sites. Bending of DNA because of local
sequence variations may also be important to the
positioning of nucleosomes (Fig. 27-3) on DNA.
155
Several causes for DNA bending have been identi-
fied.
124,138,142,156 164
For example, a sharp 26 bend is
expected at the junction between B- and A-DNA
segments.
158
The presence of a thymine photodimer
(Eq. 23-26) may cause a 30 bend.
165
Some naturally
occurring bent DNAs contain repeated (A+T)-rich
sequences. This suggested that the sequence (AA,TT),
containing adjacent thymine methyl groups, can be
thought of as a wedge, consisting of both roll and tilt
components. Such a wedge repeated at intervals aver-
aging 10.5 bp along the helix might cause the helix to
bend.
129,144
However, computer modeling experiments
suggest that thymine methyl groups do not distort the
DNA helix. The bending may be a result of the greater
tendency for AT pairs to roll and to enhance the van der
Waals and electrostatic attractions between the 2'-O
of cytosine and the 2'-NH
2
of guanine in the CG base
pairs of bent DNA.
157,166
Bending can occur in both
(A+T)- and (G+C)-rich regions and is often induced
by binding to proteins or protein complexes that act
in replication, transcription, and recombination.
167
Various errors are made during replication or
recombination of DNA. Incorporation of an incorrect
nucleotide will cause a mismatched base pair in which
proper hydrogen bonds cannot be formed. Most of the
mispaired bases that result from mistakes in replication
are removed by repair processes (Chapter 27). Those
that remain can often assume alternative pairings that
distort the helix only slightly. For example, a G-T wobble
pair fits readily into an oligodeoxyribonucleotide double
helix. Even a bulky G-A base pair causes little pertur-
bation of the helix.
168
Incorporation of an extra nucleo-
tide into one strand of the DNA will create a bulge in
the helix.
169
NMR spectroscopic studies on bulged
oligonucleotides have shown that the extra base can
be stacked into the helix causing a sharp bend.
170,171
However, some oligonucleotides with mismatched
bases crystallize as straight helices with the extra
nucleotide looped out.
171 173
Mismatched base pairs
tend to destabilize helices.
174
Interactions with ions. Because each linking
phospho group carries a negative charge (two charges
per base pair, Fig. 5-2) the behavior of polynucleotides
is strongly affected by cations of all kinds. The pre-
dominant small counterions within cells are K
+
and
Mg
2+
. They are attracted to the negative charges on
the polynucleotide backbone and, although they remain
mobile, they tend to occupy a restricted volume.
175 177
Some may bind in well-defined locations as in Fig. 5-8.
Because of the presence of these positive ions the inter-
actions of nucleic acids with cationic groups of proteins
are strongly affected by the salt concentration.
Organic cations compete with the simple counteri-
ons K
+
and Mg
2+
. Among these, the polyamines are
predominant.
178
Crystal structures have revealed that
spermine binds across the deep grooves of tRNA and
the major groove of B-DNA. Spermine also binds into
the deep groove of A-DNA interacting by hydrogen
bonding with bases in GTG sequence in both strands.
179
It binds tightly to CG-rich sequences in the minor groove
of Z-DNA, where it tightens the structure and shortens
the helix.
110
At higher concentrations of DNA, as occur
in cell nuclei, spermidine induces the conversion of the
DNA into liquid crystalline phases.
180
Heavier metal ions and metal complexes can find
sites on nitrogen atoms of the nucleic acid bases. Exam-
ples are the platinum complex cisplatin and the DNA-
cleaving antibiotic neocarzinostatin (Box 5-B). Can
metals interact with the electrons of stacked DNA
bases? A surprising result has been reported for inter-
calating complexes of ruthenium (Ru) and rhodium
(Rh). Apparent transfer of electrons between Ru (II) and
Rh (III) over distances in excess of 4.0 nm, presumably
through the stacked bases, has been observed,
181
as
has electron transfer from other ions.
181a
Stacked bases
are apparently semiconductors.
182
C. The Topology and Dynamics of Nucleic
Acids
1. Rings, Catenanes, and Knots
While a DNA molecule may exist as a straight rod,
the two ends are often covalently joined. Thus, the
chromosomes of E. coli and of other bacteria are single
closed circles. Circular DNA molecules are also found
in mitochondria, chloroplasts, and many viruses. Fur-
ther complexity arises from the fact that the circles of
DNA are sometimes interlocked in chainlike fashion
(catenated). An unusual example of this phenomenon
is the presence of thousands of small catenated DNA
circles in the single mitochondrion of a trypanosome
(Fig. 5-16).
183
Sometimes circular DNA is knotted as
in Fig. 5-17.
184 186
Knots and catenanes often appear
as intermediate forms during replication and recombi-
nation, especially involving circular DNA.
187,188
Methods have been devised for synthesis of even
very complex DNA knots.
185,186
Lets look briefly at
the topology of knots. The three simple knots shown
here have a chirality beyond that of the nucleotide
219
The node is negative if the crossing is like that in a left-
handed supercoil and positive if like that in a right-
handed supercoil:
191,192
2. Supercoiled DNA
Double-stranded DNA in solutions of low salt
content usually assume the B-DNA conformation with
10.410.5 base pairs per turn. If the two ends are joined
the resulting covalently closed circular DNA will be
relaxed. However, there are topoisomerases that act
on this form of DNA by cutting both strands, holding
the ends, and twisting the two chains (Chapter 27).
Figure 5-17 Electron micrograph of a six-noded knot made
by the Tn3 resolvase which is involved in movement of the
Tn3 transposon (Chapter 27) from one location to another
within the genome. Putative six-noded knot DNA was
isolated by electroelution from an agarose gel. The knots,
which are nicked in one strand, were denatured to allow the
nicked strand to slide away and leave a ssDNA knot. This was
coated with E. coli recA protein (Fig. 27-24) to greatly thicken
the strand and to permit the sign of each node (designated in
the tracing) to be seen. From Wasserman et al.
184
C. The Topology and Dynamics of Nucleic Acids

Trefoil knot () Trefoil knot (+)


+ +
+

+ +
Figure-8 knot
units.
189,190
This can be expressed by indicating the
sign of each node in the knot:
() Node (+) Node
Figure 5-16 (A) Electron micrograph of the network of
catenated DNA circles in the mitochondrion of the trypano-
some Crithidia fasciculata. (B) and (C) The same network
after treatment with a topoisomerase from bacteriophage
T4 that catalyzes a decatenation to form individual covalently
closed circles (Chapter 27). Five times as much enzyme was
added in (C) as in (B). Two sizes of circles are present. Most
are minicircles, each containing about 2300 bp but a smaller
number of larger ~ 35-kb maxicircles are also present. One
of these is marked by the arrow. From Marini, Miller, and
Englund.
183
A
B
C
1 m
220 Chapter 5. The Nucleic Acids
The energy source for the process is provided by
cleavage of ATP. DNA gyrase untwists relaxed circular
dsDNA one turn at a time and reseals the cut ends. A
reverse gyrase from certain bacteria twists the relaxed
DNA more tightly. In both cases the change causes the
DNA to form superhelical turns.
67,193 197
These may
be either solenoidal or plectonemically interwound
(as a twisted thread; Fig. 5-18).
The geometric and topological properties of
closed supercoiled DNA molecues may be described
by three quantities: The linking number (Lk also
called the winding number, ), the twist (Tw), and the
writhe (Wr). If a segment of double helical DNA were
laid on a flat surface and the ends were joined to form
a relaxed circle both Lk and Tw would equal the number
of helical turns in the DNA. The writhe Wr would be
zero. The linking number is a topological property. It
has an integral value which is unchanged if the DNA
molecule is distorted. It can be changed only for DNA
with open ends. When the ends are joined the linking
(winding) number is constant unless one or both
chains are cleaved. However, twist and writhe are
geometric properties, which can change according to
Eq. 5-8 while the Lk remains constant. The twist is
related to the number of helical turns while the writhe
is related to the number of superhelical turns.
27,67,198 200
Lk = Tw + Wr
However, Tw and Wr do not usually have integral values.
It is hard to define the relaxed state for a circular DNA.
For example, changing the ionic composition will alter
Tw and Wr according to equation 5-8. Both Lk and
Tw are taken as positive for a right-handed toroidal
(solenoidal) supercoil or a left-handed interwound twist.
For relaxed B-DNA, Lk is equal to the total number
of base pairs in the circle divided by 10.5, Wr = 0, and
Tw = Lk. Both Lk and Tw are positive in the right-handed
B- and A-DNA forms, but Tw is negative for Z-DNA.
In closed circular DNA the value of Wr is usually nega-
tive, the secondary structure being a fully formed
WatsonCrick helix but with right-handed interwound
superhelical turns or left-handed toroidal superhelical
turns. The helix is said to be underwound (Lk < Tw).
Some of the topological properties of double-
stranded DNA can be demonstrated by twisting together
two pieces of flexible rubber tubing whose ends can
be joined with short rods to form a closed circle.
196
Twist the tubing in a right-handed fashion as tightly as
possible without causing supercoiling (Lk = Tw; Wr = 0).
If the ends are now joined the circle will be relaxed.
Now twist one turn tighter before joining the ends. A
right-handed toroidal supercoil will be formed (Wr = 1;
Lk = 1 + Tw; Tw is the same as before). If twisting is
continued until several supercoils appear before the
ends are joined, two interconvertible forms result. One
form has right-handed toroidal supercoils and the other
left-handed interwound supercoils as in Fig. 5-19. If
relaxed circular DNA is unwound by one turn by cutting
and resealing one chain, a single right-handed inter-
wound supercoil will be formed (Wr = 1, Lk = Tw 1).
On the other hand, if the two chains in a closed relaxed
helix are pried apart, as happens during intercalation
(Section 3), Lk must remain constant, Tw will decrease,
and Wr will increase with appearance of left-handed
supercoiling.
Figure 5-19 Topological equivalence of toroidal (solenoi-
dal) and plectonemically interwound forms of a circular
DNA. These two forms have a constant value of the linking
number Lk (or ), the twist Tw, and writhing number Wr.
Plectonemically
interwound
Toroidal
Figure 5-18 Two forms of supercoiling of a DNA duplex:
(A) Solenoidal. (B) Plectonemically interwound. These are
both negatively supercoiled, as can be deduced from the
arrows at the nodes, but the solenoid is left-handed and the
plectonemic form right-handed. From Wasserman and
Cozzarelli.
187
Plectonemic Solenoidal
A B
(5-8)
221
During replication of DNA (Chapter 27) pairing of
some bases associated with the replication apparatus is
prevented. Upon release of the constraint the newly
replicated DNA forms base pairs and becomes super-
coiled.
Since Lk is constant in circularly closed DNA a
change of one turn of B-DNA (Tw = 1) into a turn of
Z-DNA (Tw = 1) will cause the writhing number Wr
to change by 2. Conversely, if the writhing number is
forced to change by 2, a turn of Z-DNA may develop
somewhere in a suitable (G + C)-rich region of the DNA.
Some of the information about supercoiled DNA
can be summarized as follows:
Relaxed circular DNA Lk = Tw, Wr = 0
Underwound Lk < Tw; Wr is negative; right-
circular DNA handed interwound supercoils or
left-handed toroidal supercoils may
be present. Alternatively Lk = Tw
and left-handed Z-DNA regions
may appear.
DNA with DNA is partially untwisted; Tw is
intercalated lowered; Wr is increased with
molecules decrease in number of negative
supercoils. As Tw approaches Lk
the DNA becomes relaxed.
The superhelix density of a DNA molecule is often
expressed as = Wr/ Tw number of superhelical turns
per 10 bp.
31,198,199a,199b,201
In most naturally occurring
circular DNA molecules is negative, a typical value
being 0.05 (~ 5 negative superhelical turns per 1000
bp). The presence of superhelices in circular DNA
molecules can be recognized readily by its effect upon
the sedimentation constant of the DNA. Naturally
occurring supercoiled DNA from polyoma virus sedi-
ments rapidly but after nicking of one of the strands
of the double helix by brief exposure to a DNA-hydro-
lyzing enzyme the resulting relaxed form of the mole-
cule sediments more slowly. Supercoiling lowers the
viscosity of solutions of DNA and increases the electro-
phoretic mobility (Fig. 5-20) and may also be recognized
by electron microscopy.
Naturally occurring or artificially prepared super-
coiled DNA molecules can often be separated by elec-
trophoresis into about ten forms, each differing from
the other by one supercoiled turn and by Lk = 1
(Fig. 5-20). The relative amounts of these topological
isomers form an approximately Gaussian distribution.
The isomers apparently arise as a result of thermal
fluctuations in the degree of supercoiling at the time
that the circles were enzymatically closed.
202
Why is DNA in cells supercoiled? One effect of
supercoiling is to contract the very long, slender double
helices into more compact forms. In eukaryotic cells
much of the DNA exists in nucleosomes. Each bead-
like nucleosome consists of a core of eight subunits of
proteins called histones around which an ~ 140 bp
length of DNA is coiled into two negative, left-handed
toroidal superhelical turns (Fig. 5-21).
203,204
There is
some spacer DNA between nucleosomes. Otherwise a
nucleosome providing two superhelical turns per 200
bp would produce a superhelix density of 0.1, twice
that observed. A detailed analysis of the geometric
properties of DNA wrapped around nucleosomes or
other protein particles has been developed.
199199c
Nucleosome formation is thought to protect DNA and
to keep it in a more compact state than when it is fully
active and involved in transcription. Nucleosomes,
discussed further in Chapter 27, are also important in
the regulation of transcription. Even though nucleo-
somes as such are absent, bacterial DNA also has a
superhelix density of 0.05, apparently a result of inter-
action with other proteins such as the histonelike HU.
205
Interaction with smaller molecules can also affect super-
coiling.
206
The presence of naturally supercoiled DNA and a
variety of topoisomerases suggests that the control of
DNA supercoiling is biologically important. In fact,
Figure 5-20 Electrophoresis of DNA from the SV40 virus
with varying numbers of superhelical turns. Molecules of
the native DNA (lane 1) move rapidly toward the anode as a
series of bands, each differing from its neighbors by one
superhelical turn. The average number of superhelical turns
is about 25. Incubation with a topoisomerase from human
cells causes a stepwise removal of the superhelical turns by
a cutting and resealing of one DNA strand. The DNA incu-
bated with this enzyme at 0C for periods of 1, 3, 6, 10, and
30 min (lanes 2 6) is gradually converted to a form with an
average of zero supercoils. For details see Keller.
202,212
Electrophoresis was carried out in an 0.5% agarose 1.9%
polyacrylamide slab gel (17 x 18 x 0.3 cm). The bands of
DNA were visualized by staining with the fluorescent inter-
calating dye ethidium bromide. Photograph courtesy of
Walter Keller.
C. The Topology and Dynamics of Nucleic Acids
222 Chapter 5. The Nucleic Acids
topoisomerases play essential roles in both replication
of DNA and in transcription of genes. Supercoiling
requires energy and topoisomerases that induce super-
coiling must provide energy, e.g., by cleavage of ATP.
Conversely, supercoiled DNA can be a source of ener-
gy for biological processes. It has been estimated that
the H, S, and G per mole for formation of a single
superhelical turn are 35.9 kJ, 68 J/ K, and 14.6 kJ,
respectively.
207
A reduction in superhelix density is
accompanied by a decrease in Gibbs energy and can
therefore be coupled to other processes that have
positive values of G. An example is conversion of
(G + C)-rich regions into the Z form of DNA, which is
favored by negative supercoiling.
207,208
Supercoiling
affects binding of various proteins to DNA
197,209,210
as
well as intercalation (discussed in the next section) and
formation of cruciform structures (Section D,3).
The folding of DNA into compact forms, such as
that in chromosomes, is also influenced by supercoil-
ing. In the absence of nucleosomes supercoiled DNA
may assume the plectonemic form (Figs. 5-18, 5-19).
Segments of the resulting rods may aggregate side-
by-side in a liquid crystalline state.
200,211
The relatively
high cation concentration within cells favors this trans-
formation.
215217
Polyamines such as spermidine are
especially effective in promoting aggregation of DNA,
in formation of Z-DNA,
110
and possibly in facilitating
10C 4W 5W 6W 7W 8W
9W
10W 4C 5C 6C 7C 8C
14W
13W
1C
12W 2C
H
1
9C
9C
2W 12C 1W 13C 14C
Figure 5-21 Nucleosomes.
(A) Electron micrographs
of individual nucleosomes
reconstituted from 256-bp DNA
fragments and separated pro-
teins. From Hamiche et al.
213
Courtesy of Ariel Prunell. (B)
Model of a nucleosome core.
The 1.75-turn (145-bp) DNA
superhelix winds around the
histone octomer which consists
of two subunits apiece of
histones H2A, H2B, H3, and
H4. In addition, two elongated
molecules of proteins HMG-14
or HMG-17 are indicated (see
also Chapter 27). (C) Schematic
radial projection of the double-
helical DNA showing areas
protected from cleavage by
hydroxyl radicals (see Fig. 5-50)
by the bound proteins. The
shaded areas are those pro-
tected by HMGs. The zigzag
lines near the dyad axis indicate
the most prominent regions of
protection. (B) and (C) are
from Alfonso et al.
214
cooperative processes that require that two DNA
molecules interact with each other.
218,219
3. Intercalation
Flat, aromatic, hydrophobic rings are often able
to insert themselves between the base pairs of a DNA
duplex. Such intercalation is observed for many anti-
biotics, drugs, dyes, and environmental pollutants.
Among them are proflavine, ethidium bromide, actino-
mycin (Box 28-A), hycanthone (Fig. 5-22), and dauno-
mycin (Fig. 5-23). Hycanthone, employed in the treat-
ment of schistosomiasis, is one of the most widely used
drugs in the world. Since intercalating agents can be
mutagenic, such drugs are not without their hazards.
Intercalation is often used to estimate the amount
of negative supercoiling of DNA molecules. Varying
amounts of the intercalating agent are added, and the
sedimentation constant or other hydrodynamic prop-
erty of the DNA is observed. As increasing intercala-
tion occurs, the secondary turns of DNA are unwound
(the value of Tw in Eq. 5-8 decreases). Each intercalated
ring causes an unwinding of the helix of ~ 26. Since
for a closed covalent duplex the value of Lk in Eq. 5-8
is constant, the decrease in Tw caused by increased
intercalation leads to an increase in the value of Wr,
A B
C
223
Figure 5-22 Structures of some substances that tend to intercalate into
DNA structures. See also Fig. 5-23.
O O
O
O
C
O
CH
3
OH
H
3
C
O
NH
3
HO
H
3
C
S
O
CH
2
OH
HN
(CH
2
)
2
N H
C
2
H
5
C
2
H
5
N
C
2
H
5
NH
2
H
2
N
N
H
CH
3
NH
2
H
3
C
H
2
N
+
Daunomycin (daunorubicin), an antitumor antibiotic
+
Hycanthone, a widely used drug for schistosomiasis
N
H
NH
2
H
2
N
Acridine yellow
+
+
+
Ethidium ion
Proflavine
OH
OH
14
which is usually negative for natu-
ral DNA. When sufficient intercala-
tion has occurred to raise Wr to zero,
a minimum sedimentation rate is
observed. Addition of further inter-
calating agent causes a positive
supercoiling.
When the replicative form of
DNA of the virus X174 (Chapter
27), a small circular molecule con-
taining ~ 5000 bp, was treated with
proflavine,
221
the binding of 0.06 mol
of proflavine per mole of nucleotides
reduced Wr to zero. From this it
was estimated that = 0.055, corre-
sponding to 27 superhelical turns
at 25, pH 6.8, ionic strength ~ 0.2.
Changes in temperature, pH, and
ionic environment strongly influ-
ence supercoiling. In general
becomes less negative by ~ 3.3 x 10
4
per degree of temperature increase.
222
For example, the observed value
221
of for X174 DNA was 0.059 at
15C and 0.040 ( 20 superhelical
turns) at 75C at an ionic strength
of ~ 0.2.
The exact ways in which inter-
calating substances can fit between
the base pairs of nucleic acids are
being revealed by X-ray diffraction
studies of complexes with nucleo-
sides, dinucleotides, and other
oligonucleotides.
220,223 225
The
structure of a complex in which
daunomycin is intercalated between
two GC base pairs in DNA is shown
as in Fig. 5-23. Daunomycin and
other related anthracycline antibio-
tics also have an amino sugar ring
that binds into the minor groove of
the DNA, providing both electro-
static stabilization and hydrogen
bonding. Substituents on this ali-
phatic ring also hydrogen bond to
DNA bases.
226
Does intercalation of flat mole-
cules into nucleic acid chains have a
biochemical function? Aromatic
rings of amino acid side chains in
proteins designed to interact with
nucleic acids may sometimes inter-
calate into nucleic acid helices serv-
ing a kind of bookmark function.
227
Changes in
superhelix density caused by such intercalation may be
important in the orderly handling of DNA by enzymes
within cells.
C. The Topology and Dynamics of Nucleic Acids
Figure 5-23 Stereoscopic drawing showing a molecule of daunomycin
(Fig. 5-22) intercalated between two base pairs in a molecule of double-helical
DNA, d(CGTACG). Nitrogen and oxygen atoms are shown as dots. From
Quigley et al.
220
Both daunomycin and adriamycin (doxorubicin; 14-hydroxy-
daunomycin) are important but seriously toxic anticancer drugs.
224 Chapter 5. The Nucleic Acids
BOX 5-B ANTITUMOR DNA DRUGS
Chemotherapy of cancer at present involves
simultaneous use of two or more drugs. For exam-
ple, antifolates (Chapter 15) or nucleoside analogs
such as 5-fluorouridine may be used together with
a drug that binds directly to DNA and inhibits the
replication of cancer cells. In 1963, it was discov-
ered accidentally that platinum ions released from
supposedly inert platinum electrodes inhibited the
division of E. coli cells. This led Rosenberg and asso-
ciates to test platinum compounds against animal
cells.
a
Among many compounds tested cis-dichloro-
diammineplatinum(II) (cis-DDP or cisplatin) emerged
as an important anticancer drug that is especially
effective against testicular and ovarian cancers.
b d
The trans isomer, however, is inactive.
cis-DDP binds to adjacent deoxyguanosines within
one strand of ds or ssDNA. The stacking of adjacent
bases is disrupted as the platinum binds to N-7
nitrogen atoms of the two guanine rings by replace-
ment of the two chloride ions. The product has the
following structure,
c,e
with the Pt lying in the minor
groove of dsDNA:
The two guanines are no longer stacked, but the
structure is impossible for the trans isomer. The
cisplatin adducts appear to prevent proper DNA
repair and to induce programmed cell death
(apoptosis).
f
Since the 1950s, using a different approach, the
U.S. National Cancer Institute, as well as agencies in
other countries, has sought to find natural anticancer
compounds in plants, fungi, microorganisms, and
marine invertebrates.
g
Among these are many
antibiotics that intercalate into DNA helices, e.g.,
daunomycin (Figs. 5-22, 5-23), menogaril,
h
triostin
A,
i
and the antitrypanosomal drug berenil.
j
Some
of these are also alkylating agents that contain double
bonds to which such groups as the 2-NH
2
of guanine
may add:
Diol epoxides and cyclic imines such as mitomycin
also form adducts specifically with guanine 2-NH
2
groups:
l
Neocarzinostatin is an antitumor protein
with a nonprotein chromophore. After intercala-
tion and binding into the minor groove of bulged
DNA, it undergoes activation by addition of a
thiol group. The enediyne structure undergoes
rearrangement with formation of a reactive diradical
that attacks the DNA.
m
A family of related antitumor
enediynes has also been discovered.
n
Pt
H
3
N Cl
H
3
N Cl
cis-DDP
(Cisplatin)
Pt
Cl NH
3
H
3
N Cl
trans-DDP
(inactive)
O
5
N
N
N
O
N
O
NH
2
H
3
N
H
3
N
O O
O
N
N
N
N
3

O
O
P
Pt
HN
N
N
N
O
H
2
N
N
N
H
H
O
R
Guanine
in DNA
Tomaymycin and related antibiotics
k
Neocarzinostatin chromophore
Enediyne
OH
O
Ome
Me
O
O
MeHN
HO
Me
HO
O
O
O
O
O
225
BOX 5-B (continued)
Another group of drugs occupy extended bind-
ing sites in the minor grooves of DNA double helices,
often with specificity for a particular base sequence.
Examples are the antitrypanosomal drug berenil,
j
toxic Streptomyces antibiotics netropsin, distamycin,
and related synthetic compounds.
o q
Netropsin lies
within the minor groove in regions with two or
more consecutive AT pairs, displacing the spine of
hydration as shown in the following stereoscopic
drawing. Binding depends upon both electrostatic
interactions and formation of specific hydrogen
bonds involving the amide groups of the antibiotics.
O
O
N
H
2
N
H
3
C
NH
OMe
O
NH
2
O
1
2
7
6
9
10
10a
9a
Mitomycin
N
H
N
H6
H5
H3
H2
NH
2
H
2
N
+
N
H6'
H5'
H3'
H2'
NH
2
NH
2
+
Berenil
N
O HN
H2N NH2
+
CH3
HN
O
N
CH3
HN
O
NH
H
2N
H2N
+
The antibiotic bleomycin, which also binds in
the minor groove of B-DNA with some specificity
for G-C Sites, forms an iron (II) complex. It can be
oxygenated to form an Fe(II) O
2
complex (see
Chapter 16) which cleaves the DNA chain.
s,t
Syn-
thetic compounds that do the same thing have been
made by connecting an EDTAiron, or other iron
chelate complex covalently to the DNA-binding
compound.
n,u x
A goal is to direct drugs to selected
target sites in DNA and to induce bond cleavage at
those sites in a manner analogous to that observed
with restriction endonucleases. Most current chemo-
therapeutic agents are very toxic. Present research
is designed to target these drugs more precisely to
specific DNA sequences and to identify target se-
quences peculiar to cancers. See also Designed
third strands in main text.
Netropsin lying in the minor groove of B-DNA
hydrogen-bonded to bases in the central ATAT
tetranucleotide. From Coll et al.
r
Netropsin
N N
CH3 HN
O
O
N
HO
CH3
O
HO
N
CH3
NH S H2N
N
NH2
O
O
NH2
H NH2
O
H
N
N
S
R
O
CH3
H
H
O
N
N
H
H
O
O
OH
HO
OH
O
O
OH
OH
OH
NH2
O
H
H
H
NH S
+
(CH3)2
H
H
R =
Bleomycin A
2
H
H
C. The Topology and Dynamics of Nucleic Acids
226 Chapter 5. The Nucleic Acids
BOX 5-B Continued
a
Rosenberg, B., Van Camp, L., and Krigas, T. (1965) Nature
(London) 205, 698699
b
Zamble, D. B., and Lippard, S. J. (1995) Trends Biochem. Sci. 20,
435439
c
Pilch, D. S., Dunham, S. U., Jamieson, E. R., Lippard, S. J., and
Breslauer, K. J. (2000) J. Mol. Biol. 296, 803812
d
Temple, M. D., McFadyen, W. D., Holmes, R. J., Denny, W. A.,
and Murray, V. (2000) Biochemistry 39, 55935599
e
Huang, H., Zhu, L., Reid, B. R., Drobny, G. P., and Hopkins, P.
B. (1995) Science 270, 18421845
f
Zlatanova, J., Yaneva, J., and Leuba, S. H. (1998) FASEB J. 12,
791 799
g
Booth, W. (1987) Science 237, 969970
h
Chen, H., and Patel, D. J. (1995) J. Am. Chem. Soc. 117, 59015913
i
Wang, A. H.-J., Ughetto, G., Quigley, G. J., Hakoshima, T., van
der Marel, G. A., van Boom, J. H., and Rich, A. (1984) Science
225, 11151121
j
Pilch, D. S., Kirolos, M. A., Liu, X., Plum, G. E., and Breslauer,
K. J. (1995) Biochemistry 34, 99629976
k
Barkley, M. D., Cheatham, S., Thurston, D. E., and Hurley, L. H.
(1986) Biochemistry 25, 30213031
l
Kumar, G. S., Lipman, R., Cummings, J., and Tomasz, M. (1997)
Biochemistry 36, 14128 14136
m
Yang, C. F., Stassinopoulos, A., and Goldberg, I. H. (1995)
Biochemistry 34, 22672275
n
Nicolaou, K. C., Dai, W.-M., Tsay, S.-C., Estevez, V. A., and
Wrasidlo, W. (1992) Science 256, 11721178
o
Goodsell, D. S., Ng, H. L., Kopka, M. L., Lown, J. W., and
Dickerson, R. E. (1995) Biochemistry 34, 1665416661
p
Rentzeperis, D., Marky, L. A., Dwyer, T. J., Geierstanger, B. H.,
Pelton, J. G., and Wemmer, D. E. (1995) Biochemistry 34, 2937
2945
q
Tanious, F. A., Ding, D., Patrick, D. A., Tidwell, R. R., and
Wilson, W. D. (1997) Biochemistry 36, 1531515325
r
Coll, M., Aymami, J., van der Marel, G. A., van Boom, J. H.,
Rich, A., and Wang, A. H.-J. (1989) Biochemistry 28, 310320
s
Burger, R. M., Drlica, K., and Birdsall, B. (1994) J. Biol. Chem.
269, 2597825985
t
Kane, S. A., Hecht, S. M., Sun, J.-S., Garestier, T., and Hlne, C.
(1995) Biochemistry 34, 1671516724
u
Veal, J. M., and Rill, R. L. (1988) Biochemistry 27, 1822 1827
v
Campisi, D., Morii, T., and Barton, J. K. (1994) Biochemistry 33,
41304139
w
Mack, D. P., and Dervan, P. B. (1992) Biochemistry 31, 93999405
x
Han, H., Schepartz, A., Pellegrini, M., and Dervan, P. B. (1994)
Biochemistry 33, 98319844
H-DNA. Strands of DNA contain many homopy-
rimidine tracts consisting of repeated sequences of
pyrimidines, e.g., d(T-C)
n
, which may be abbreviated
more simply as (TC)
n
. In this example the comple-
mentary strand would contain the two purines in the
repeated sequence (GA)
n
. At low pH, where protona-
tion of the cytosine rings occurs, or in negatively super-
coiled DNA, the two strands of the repeating sequence
may separate, with the (TC)
n
strand folding back to
form a triple helix in which the base triplets have the
hydrogen bonding pattern of Fig. 5-7. The resulting
structure, which is shown in Fig. 5-24, is known as
H-DNA.
239 241
A variety of related nodule and
looped forms of DNA can also be formed.
35,242
R-DNA. A different type of triplex DNA may be
formed during genetic recombination. A WatsonCrick
duplex is brought together by one or more proteins
with a single strand that is, for at least a considerable
distance, an exact copy of one of the strands of the
duplex. It is within such a triplex that cutting of a strand
of the duplex takes place to initiate recombination (see
Chapter 27 for a detailed discussion). Can a triplex
structure containing two identical chains be formed?
Possible base triplets include the following CG
*
G
triplet which occurs in a crystalline oligonucleotide
structure.
243
This triplet contains a variation on
Hoogsteen pairing and is related to the first triplet in
Fig. 5-7. The other triplets needed for the proposed
R-DNA recombination intermediates are GC
*
C,
TA
*
A, and AT
*
T. While there is keen interest in
R-DNA
35,244 247
the formation of a stable intermediate
triplex is still uncertain.
35,247
4. Polynucleotides with Three or Four Strands
Some nucleotide sequences in DNA favor the
formation of a regular triple-helical (triplex) structure.
This is possible when there are long stretches of adjacent
pyrimidines having any sequence of C and T in one
strand of double-helical DNA. The other strand of the
DNA will contain the correct purines for formation of
WatsonCrick base pairs. With such a structure it is
always possible to add a third strand of a polypyrimi-
dine using Hoogsteen base pairing. Triads formed in
this way contain either two Ts and one A or two Cs
and one G, as is shown in Fig. 5-7. In the latter case one
of the Cs must be protonated to allow formation of the
pair of hydrogen bonds.
227a
These triplex structures can
be formed only for stretches of DNA containing all
pyrimidines in one strand (a homopyrimidine strand)
and all purines in the other (a homopurine strand). A
poly (AAU) triplex can also be formed, using the hydro-
gen bond pattern of the first triplet in Fig. 5-7. Triple-
stranded synthetic polynucleotides of these types were
prepared by Felsenfeld and others as early as 1957.
228230
The third strand in a triplex can be either parallel or
antiparallel to the homopurine strand. The third strand
is either homopurine or a mixture of purine and thym-
ine. The triplets are GC
*
C, A
*
AT, or T
*
AT and are
formed by WatsonCrick () and reversed Hoogsteen (
*
)
pairing.
231
Recently, there has been a renewed interest in
DNA triplets because of their occurrence in natural
DNA, their possible importance in genetic recombina-
tion, and the potential for design of powerful inhibitors
of replication and transcription that function via triplex
formation.
35,232 238
227
Designed third strands. If triplex DNA segments
can form naturally it should be possible to design oli-
gonucleotides that will bind into the major groove of a
DNA duplex to form a triplex at a specific location or
locations in the genome. Dervan and associates are
studying this approach systematically.
248 250
Such
oligonucleotides may be modified chemically to pro-
vide stronger binding to targeted locations and may
prove to be useful therapeutic agents. Among these
C. The Topology and Dynamics of Nucleic Acids
are synthetic phosphoramidates in which the phos-
phodiester linkage between nucleosides is replaced
by 3'-NH PO
3

O-5'. The resulting oligonucleotide is


resistant to digestion by phosphodiesterases present in
cells. Its geometry favors binding to A-DNA or
RNA.
251
Another oligonucleotide mimic, dubbed PNA
(peptide nucleic acid), contains monomer units of the
following type
252253b
and is able to form a triple helix:
Yet another approach is to synthesize a hairpin poly-
amide that contains pyrrole, hydroxypyrrole, and
imidazole groups in a sequence that favors tight bind-
ing to a specific dsDNA sequence.
254
Some possible
geometries for triplex DNA are illustrated in Fig. 5-25.
These are based on computer-assisted modeling.
238
Tetraplex (quadruplex) structures. The ends
of linear chromosomes, the telomeres, have unusual
nucleotide sequences repeated hundreds or thousands
of times.
255 258
There is usually a guanine-rich strand
running 5' to 3' toward the end of the chromosome and
consisting of sequences such as TTAGGG in vertebrates,
TTTTGGGG in the ciliate Oxytricha, and TG
1 3
in Sac-
charomyces cerevisiae. The complementary strand is
cytosine rich. The guanine-rich strand is longer than
its complement, overhanging by about 2 repeat units.
The significance for the replication of chromosomes is
discussed in Chapter 27.
The thing that has attracted most attention to
telomeric DNA is the unusual structure of the G-rich
strand that was signaled by the first NMR studies.
259
Subsequent investigation
260,261
revealed the presence
of G quartets (Fig. 5-8) which are apparently stacked
in folding patterns such as the following:
255,262 268
These structures are stabilized by the presence of
univalent cations, K
+
being more effective than Na
+
,
0.28
0.29
0.28 0.28
0.29
0.28
N
N
N
N
N
N
N
N
H
H
H
H
H
O
O
C
G
N
N
N
O
H
H
H
N
N
O
O
O
Watson-Crick
pair
G
CH
2
N C
CH
2
O
H
2
C
C
NH O
O
H
2
C
N H
Base
Residue of
a PNA chain.
The phantom
green structure
shows the
relationship to
a nucleotide.
5'
3'
3'
5'
Homopurine
segment
Homopyrimidine segment
Figure 5-24 Proposed structure of H-DNA which can be
formed when a homopyrimidine segment and its comple-
mentary homopurine segment separate as a result of pro-
tonation of the cytosine rings or of negative supercoiling
stress.
239,240
The triple-helical portion contains base triplets
of the kind shown in Fig. 5-7.
228 Chapter 5. The Nucleic Acids
Li
+
, Rb
+
, or Cs
+
.
260,269 271
A protein isolated from Tetra-
hymena binds specifically to the G4-DNA regions.
272
The presence of 5-methylcytosine (m
5
c) also stabilizes
quadruplex structures.
273
The oligoribonucleotide UG
4
U
forms G
4
quartets so stable that it takes days at 40C in
D
2
O for the hydrogen-bonded NH protons forming the
quartet to be exchanged for
2
H.
274
Tetraplex structures have also been observed for
G-rich repeating sequences associated with the human
fragile X syndrome.
275,276
This is the most common
cause of inherited mental retardation and appears to
arise as a result of the presence of an excessive number
of repeats of the trinucleotide sequence (CGG)
n
. For
normal persons n = 60 or less; for healthy carriers n
may be as high as 200 but for sick individuals it may
be much higher.
275
The structure in solution, as deter-
mined by NMR spectroscopy, is shown in Fig. 5-26.
Another variant of four-stranded DNA, which arises
from cytosine-rich DNA, contains CCH
+
pairs such
as the following at low pH.
277 279
In sequences such as d(TC
5
) and d(C
3
T)
these CCH
+
pairs are intercalated as is shown
in Fig. 5-27. This intercalated DNA (I-DNA)
may provide an alternative conformation for
some telomeric sequences.
281,282
An I-DNA
motif has also been identified in oligonucle-
otides from the DNA of human centromeres.
283
The seemingly unusual forms of DNA de-
scribed in this section may represent only a
fraction of the naturally occurring DNA struc-
tures of biological significance.
5. Junctions
Special structural features may be found at
junctions between different types of DNA, e.g.,
between A-DNA and B-DNA.
284 286
However,
the most interesting junctions are branched.
287290
For example, Fig. 5-28 shows a four-way junc-
tion in which all of the bases form Watson
Crick pairs. This junction is better known as a
Holliday junction because it was proposed by
Holliday in 1964 as an intermediate in genetic
recombination.
291
As shown at the top of Fig.
5-28A the junction is formed from two homolo-
gous DNA duplexes. These are identical except for the
boxed and shaded base pairs. The ends of the first
duplex are marked I and II and those of the second
III and IV. The Holliday junction appears to arise by
cleavage of one strand of each duplex with rejoining of
the strands as indicated by the green arrows. Rotation
gives the untwisted Holliday junction structure
Figure 5-25 Two superposed stereoscopic diagrams illustrating
the lowest energy triple helical conformations of one pyrimidine,
one purine, and one Hoogsteen DNA strand (in black). Super-
posed is an RNA purine strand in gray. See Srinivasan and
Olson.
238
Courtesy of Wilma K. Olson.
Figure 5-26 Structure of a GCGC tetrad present in a
quadruplex structure formed by the oligonucleotide d(GCG-
CTTTGCGC) in Na
+
-containing solution. See Kettani et al.
276
N
N
N
N
O
d-ribose
H
G3
N
H
H
G10
C9
H
N
N
H
H
O N
d-ribose
H
N
N
N
N
O
d-ribose
H
N
H
H H
N
N
H
H
O N
d-ribose
H
H
H
C2
N
N
N
H
H
d-ribose
O
H
H H
H
N
O
N
N
H
H
d-ribose
H
+
3
5
5 3
Hemiprotonated d(CH
+
C)
Hemiprotonated d(CH
+
C)
229
shown. The postulated three-dimen-
sional structure of the junction is
indicated in Fig. 5-28B.
292 294
An important characteristic of
Holliday junctions formed from
homologous duplexes is that they
can move by a process called branch
migration.
295
Because of the twofold
symmetry of the branched structure
the hydrogen bonds of one base pair
can be broken while those of a new
base pair are formed, the branch
moving as shown in Fig. 5-28. Notice
that, in this example, the nonhomo-
logous (boxed) base pairs TA and
GC have become mispaired as TG
and AC after branch migration. More
significantly, the junction may be cut
by a resolvase at the points marked
C. The Topology and Dynamics of Nucleic Acids
Figure 5-28 (A) Abbreviated reaction sequence
for formation of a four-way Holliday junction
between two homologous DNA duplexes. In
step a strands are cut and rejoined with movement
of the strands to a roughly antiparallel orientation.
The resulting structure is thought to resemble that
shown below the four-stranded representation.
In step b branch migration takes place, separating
the nonhomologous base pairs TA and CG and
causing mismatched pairs which will be subject
to repair. (B) Proposed three-dimensional
structure (after drawing by Bennett and West).
292
G
C
C
G
5'
3'
I II
T
A
G
C
T
A
C
G
A
T
G
C
T
A
A
T
T
A
G
C
3'
5'
G
C
C
G
5'
3'
III IV
T
A
G
C
T
A
C
G
G
C
C
G
5'
3'
I
T
A
G
C
T
A
C
G
A
T
G
C
C
G
A
T
T
A
G
C
3'
5'
III
G
C
A
T
C
G
A
T
G
C
C
G
3'
5'
C G
A T
5' 3'
II
IV
T A
A T
C G
T A
A T
G C
C G
A T
T A
G C
3' 5'
G
C
C
G
5'
3'
I
T
A
G
C
T
A
C
G
III
G
C
A
T
T
A
C
G
A
C
T
A
A
T
T
G
G
C
A
T
C
G
A
T
G
G
G
G
3'
5'
C G
A T
5'
Branch migration
3'
II
IV
T A
G C
3' 5'
a
b
cut
cut
5
3
3
5
Figure 5-27 Stereoscopic view of a four-stranded intercalated DNA or I-DNA
formed from d(C
4
). Two parallel duplexes with C CH
+
pairs are intercalated
into each other. From Chen et al.
280
B A
230 Chapter 5. The Nucleic Acids
by the green double-headed arrow. If the strand break
is then resealed, and the DNA strands are replicated,
the boxed base pair in duplex III will have been
transferred into a strand replicated from IIIIV and
genetic recombination will have been accomplished.
Branch migration can occur over much longer distances
than are indicated in this figure, so the alteration trans-
ferred may be far from the site of the initial cleavage
and whole genes or groups of genes can be transferred.
Recombination is considered in more detail in Chapter
27.
The Holliday junctions formed during recombina-
tion are mobile, but synthetic immobile Holliday
junctions can be synthesized by using nonhomolo-
gous base sequences or by locking the junctions.
288
This has permitted careful physical study of these and
other more elaborate synthetic junctions. With suitable
choices of base sequences for the oligonucleotides from
which they are made, such junctions will assemble
spontaneously. Double-stranded DNA with immobile
junctions is a very suitable construction material on
a nanochemical scale. It has been assembled into
knots, rings, cubes, and more complex polyhedra.
296 297b
D. Ribonucleic Acids (RNA)
The best known forms of RNA are: (1) the long
chains of messenger RNA (mRNA), which carry genetic
messages copied from DNA to the ribosomes where
proteins are made; (2) the much shorter transfer RNAs
(tRNAs) which participate in reading the genetic code,
correctly placing each amino acid in its sequence in
the proteins; and (3) ribosomal RNAs (rRNA), which
provide both structural material and a catalytic center
for peptide bond formation. In addition there are
numerous small RNAs that function in the splicing and
editing
298 300
of mRNA, processing of tRNA precursors,
methylation of ribosomal RNA,
301
transfer of proteins
across membranes, and replication of DNA.
302
The
genomes of many viruses consist of RNA. There are
doubtless additional as yet undiscovered types of RNA.
Unlike DNA, which exists largely as double helices,
the single chains of RNA can fold into complex forms
containing many bulges and loops of the sort depicted
in Fig. 5-9.
300,303,304
These loops are closed by double-
stranded stems which have the A conformation. The
B conformation is impossible because of the presence
of the 2'-hydroxyl groups on the ribose rings in RNA.
Even one ribonucleotide in a 10-nucleotide oligomer
prevents formation of the B structure.
55,83
The 2'-OH
groups not only keep the RNA in the A form but also
engage in hydrogen-bond formation. Hydrogen
bonds may form between the 2'-OH and the oxygen
atom in the next ribose ring in the 3' 5' direction.
The OH groups also hydrogen bond to water molecules
which form a network within the minor groove.
305,306
These bound water molecules, in turn, can bond to
associated protein and to other atoms of the complex
loops found in RNA molecules. The 2'-OH groups
also act as ligands for divalent metal ions in some
tRNAs and in some RNA catalytic sites. Transient
hybrid DNARNA double helices also exist within
cells and they too usually have the overall shape of
A-DNA.
55,307 309
However, the minor groove is inter-
mediate in width between that expected for the A and
B forms.
1. RNA Loops and Turns
Like polypeptides, polynucleotide chains have
preferred ways of bending or turning. The loops at the
ends of the hairpin turns of RNA molecules sometimes
consist of a trinucleotide such as UUU,
304
but are usually
larger. In tRNA there are typically seven bases that do
not participate in regular WatsonCrick pairing (see Figs.
5-30, 5-31). The tetranucleotide UUCG is frequently
present, and the sequence 5'-GGACUUCGGUCC forms
an unusually stable hairpin.
85,310
Other tetranucleotides,
such as UGAA,
311
CCCG (also found in DNA loops),
312
GCAA,
313
and GAAA,
314
occur often. The latter are
members of a larger group of loop structures with the
consensus sequence GNRA, where N is any nucleotide
and R is a purine.
315,315a
These sequences are very
common in highly folded structures of ribosomal
RNAs. Until recently high-resolution X-ray structures
were available for only a few tRNAs and ribozymes.
To help remedy this deficiency the structures of a
great variety of oligonucleotide stem/loop (hairpin)
structures are being determined, most by NMR spectro-
scopy.
304,316,317
The sharp turns in the loops involve
mostly rotation about the two torsion angles around
the phosphorus atom of the third, from the 5' end, of
the seven nucleotides. Base bulges on stems (Fig. 5-9)
not only introduce kinks and bends in RNA stems
169
but also provide well-defined hydrogen-bonded bind-
ing sites for proteins. Numerous branched three-way
and more complex junctions provide other important
motifs in folded RNA.
318
Among the new RNA structures are those of
RNAantisense RNA pairs in kissing hairpin com-
plexes.
87,319
Another interesting complex folding pattern
in RNA is the pseudoknot, a structural feature that
has been identified in many RNA sequences.
85,320 327
A pseudoknot can be formed if nucleotide sequences
favorable to formation of two short RNA stems are
overlapped as shown in Fig. 5-29. After stem 2 in this
drawing is formed (step a) additional base pairing can
lead to formation of stem 1 (step b). The base pairs of
the two stems can stack coaxially to form the pseudo-
knot (step c).
For the formation of base-paired stems the RNA
must contain antiparallel sequences that allow Watson
231
Crick or wobble (GU) base pairing in the stems. This
requires certain relationships in the sequences of the
DNA in the genes that encode these molecules as
discussed in Section E,3. Because of the base-pairing
requirements, some of the bases in the stems protrude
as bulges and fail to form pairs. Often, it is possible to
find more than one reasonable structure, each having
some bases unpaired.
328
2. Transfer RNAs
In all tRNAs the bases can be paired to form clover-
leaf structures with three hairpin loops and sometimes
a fourth as is indicated in Fig. 5-30.
329 331
This struc-
ture can be folded into the L-shape shown in Fig. 5-31.
The structure of a phenylalanine-carrying tRNA of yeast,
the first tRNA whose structure was determined to atomic
resolution by X-ray diffraction, is shown.
170,332 334
An
aspartic acid-specific tRNA from yeast,
335
and an E. coli
chain-initiating tRNA, which places N-formyl-
methionine into the N-terminal position of proteins,
336,337
have similar structures. These molecules are irregular
bodies as complex in conformation as globular pro-
teins. Numerous NMR studies show that the basic
structure is conserved in all tRNAs. However, animal
mitochondrial tRNAs often lack some of the usual
stem-loop arms as well as the invariant nucleotides in
the dihydrouridine and TC loops (Fig. 5-30).
307,338,339
At the bottom of the structure as shown in Figs. 5-30
and 5-31 is the anticodon, a triplet of bases having
the correct structures to permit pairing with the three
bases of the codon specifying a particular amino acid
(see Table 5-5), in this case phenylalanine.
While tRNAs consist largely of loops and stems
containing WatsonCrick base pairs, they also contain
Hoogsteen pairs, wobble pairs, and triplets such as the
following.
D. Ribonucleic Acids (RNA)
N
O
N
N
N
N
N
N
N
N
N
N
H
H
H
H
H
H
H
H
O
O
C
G

+
+ 22 13
m
7
G
N
N
H
3
C

+
N
N
N
N
N
N
O
N
O
A

+ 23 12
N
N
N
46
N H
H
H

A
H
H
N
U
G
G C
C
C G
C
U
U
A
G
G
U
A
G C
G
5
3
C
C
a
b
c
G G C U U A C G G C G C C U A G C C G
3 5
G G C
U U A
C
G
G
C
G
C
C
U
A
3
5
G
C
C
G
Stem 2
Loop 2
Stem 2
Loop 1
Stem 1
G
G
U
C
A
G
C
C
G
A
G
G
C
C
G
C
U
5
3
U
Pseudoknot
C
Figure 5-29 Formation of a pseudoknot in an RNA chain.
After Puglisi et al.
325
The first of these contains a WatsonCrick AU
pair with a second A bound to form a reversed
Hoogsteen AA pair. The second contains a GC
WatsonCrick pair with a Hoogsteen N-7-guanineG
(m
7
GG) pair. Among the complex base associations
present in tRNAs are some that also involve hydrogen
bonding to the 2'-hydroxyl groups of ribose rings and
to at least one of the phosphate groups. There are over
100 internal hydrogen bonds, a large proportion of
which are relatively invariant among the known tRNAs.
3. Properties and Structures of Some Other
RNA Molecules
A few specialized RNA molecules are listed in
232 Chapter 5. The Nucleic Acids
Table 5-4. There are many others.
302
In addition, there
are thousands of different mRNAs within a cell. The
most abundant RNA molecules in cells are those of the
ribosomes. Ribosomes consist of two elaborate RNA-
protein subunits, a large subunit with sedimentation
constant ~ 30S in bacteria and ~ 40S in eukaryotes.
The small subunit contains 16S or 18S RNA and the
large subunit 23S or 28S as well as smaller 5S and
5.8S RNAs (Table 5-4; Table 28-1). A proposed three-
dimensional structure
341,342
of a bacterial 16S ribosomal
RNA and the corresponding ribosomal subunit with
its 21 proteins are shown in Fig. 5-32. It might seem
impossible that the folding pattern of the RNA was
deduced correctly before an X-ray structure was avail-
able. However, a phylogenetic approach, the com-
parison of nucleotide sequences among several
species, suggested that the stem structures of rRNAs
are highly conserved (see discussion in Chapter 29).
This fact, together with a variety of other chemical
Figure 5-30 Schematic cloverleaf structure of a phenylala-
nine-specific transfer RNA (tRNA
Phe
) of yeast. The dots
represent pairs or triplets of hydrogen bonds. Nucleosides
common to almost all tRNA molecules are circled. Other
features common to most tRNA molecules are also marked.
The manner in which the anticodon may be matched to a
codon of mRNA is indicated at the bottom.
U
U
C
U
G
C
U
G
G
A
G
A
A
C
A
C
C
G
C
U
U
A
A
G
C
G
G
A
U
U
G A C A C
C U G U G
5'
p
3
Amino acid
acceptor end
76
Amino acid
acceptor stem
70
A
C
T

m
1
60
TC loop
Ribothymidine
50
m
5
m
7
m
2
2
C
G
C
U
G
C
G
G
5'
p
G
G
U
C
C
C
A
G
A
m
5
40
30
A
Y
C
U
G A A
C U U
Anticodon loop
Hypermodified base
5
Anticodon
Strand of mRNA
Codon in mRNA Wobble position
m
Methylation
of ribose
Always a pyrimidine
3
m
m
2
C U C G
G A G C
A
G
U
G
G
A
G
Dihydro U loop
20
h
h
Transfer RNAs (Figs. 5-30, 5-31) 60 85
Ribosomal RNAs
5S ~ 120
5.8S (rat) 158
16S (E. coli; Fig. 5-32) 1542
18S (rat) 1874
23S (E. coli) 2904
28S (rat) 4718
Telomerase guide RNA
a-d
159 (Tetrahymena)
M1 RNA of Ribonuclease P
e,f
350 410
377 (E. coli)
Tetrahymena Intron ribozyme (Fig. 12-26)
g
413
Viroid hammerhead ribozyme (Fig. 12-27)
h
~ 55
Signal recognition particle 7S RNA
i
295 (human)
Small nuclear RNAs
j,k
65 1200
RNA-editing guide RNA
l
~ 60
Thermotolerance factor (G8 RNA)
m
~ 300
Viroid RNA (Fig. 28-19) 240 380
Virus MS2 genome (Chapter 29) 3569
TABLE 5-4
Some Specialized RNA Molecules
Kind of RNA Number of nucleotides
a
Greider, C. W., and Blackburn, E. H. (1989) Nature (London) 337,
331337
b
Bhattacharyya, A., and Blackburn, E. H. (1994) EMBO J. 13, 5721
5731
c
Singer, M. S., and Gottschling, D. E. (1994) Science 266, 404409
d
Feng, J., Funk, W. D., Wang, S.-S., Weinrich, S. L., Avilion, A. A.,
Chiu, C.-P., Adams, R. R., Chang, E., Allsopp, R. C., Yu, J., Le, S.,
West, M. D., Harley, C. B., Andrews, W. H., Greider, C. W., and
Villeponteau, B. (1995) Science 269, 12361241
e
Stark, B. C., Kole, R., Bowman, E. J., and Altman, S. (1978) Proc.
Natl. Acad. Sci. U.S.A. 75, 37173721
f
Mattsson, J. G., Svrd, S. G., and Kirsebom, L. A. (1994) J. Mol. Biol.
241, 16
g
Cech, T. R. (1987) Science 236, 15321539
h
Hertel, K. J., Herschlag, D., and Uhlenbeck, O. C. (1994) Biochemis-
try 33, 33743385
i
Li, W.-Y., Reddy, R., Henning, D., Epstein, P., and Busch, H. (1982)
J. Biol. Chem. 257, 51365142
j
Maxwell, E. S., and Fournier, M. J. (1995) Ann. Rev. Biochem. 35,
897934
k
Nicoloso, M., Qu, L.-H., Michot, B., and Bachellerie, J.-P. (1996)
J. Mol. Biol. 260, 178195
l
Kable, M. L., Seiwert, S. D., Heidmann, S., and Stuart, K. (1996)
Science 273, 11891195
m
Fung, P. A., Gaertig, J., Gorovsky, M. A., and Hallberg, R. L. (1995)
Science 268, 10361039
233
Figure 5-31 The three-dimensional structure of a phenylalanine-specific transfer RNA of yeast. (A) Perspective diagram
of folding of polynucleotide chain. The ribose phosphate backbone is drawn as a continuous cylinder with bars to indicate
hydrogen-bonded base pairs. The positions of single bases are indicated by rods which are intentionally shortened. The TC
arm is heavily stippled, and the anticodon arm is marked by vertical lines. Tertiary structure interactions are illustrated by
black rods. Redrawn from Quigley and Rich.
340
(B) Stereoscopic view of the structure of yeast tRNA
Phe
as revealed by X-ray
crystallography. The acceptor stem with the protruding ACCA sequence at the 3' end is to the right. The anticodon GAA is
at the bottom right side of the drawing. The guanine ring is clearly visible at the very bottom. The middle adenine of the
anticodon is seen exactly edge-on as is the hypermodified base Y (see Fig. 5-33) which lies just above the anticodon. Its side
chain is visible at the back of the drawing. Preceding the anticodon on the left (5' side) are two unpaired bases (C and U).
The 2'-hydroxyl groups of the cytosine and of the guanosine in the anticodon are methylated. Moving far up the anticodon
stem one can see two groups of base triplets which utilize both WatsonCrick and Hoogsteen types of base pairing. Drawing
courtesy of Alexander Rich.
A B
D. Ribonucleic Acids (RNA)
Figure 5-32 (A) A three-dimensional computer graphics model proposed by Brimacombe et al.
341
for the single chain of E. coli
16S ribosomal RNA. The helices are depicted as cylinders, which are all connected. The small dark squares denote the posi-
tions of artificially formed RNAprotein crosslinks, marked with the appropriate protein number. For proteins exhibiting
more than one crosslink site (e.g., S17), the sites are denoted A or B, in each case A being the site nearer to the 5' terminus of the
16S RNA. (B) Stereoscopic view of tentative atomic model of 16S RNA in the 30S ribosomal subunit. The viewing direction is
different from that in (A). From Mueller and Brimacombe.
342
Courtesy of Richard Brimacombe.
B A
234 Chapter 5. The Nucleic Acids
evidence,
341,343,344
allowed the prediction of the struc-
ture shown as well as many characteristics of the
rRNA of the large subunit.
345
Culminating decades
of effort
346 350
(Chapter 29), complete structures of
bacterial ribosomes were established by 2000
342,351 354
and the peptidyl transferase center was identified as a
ribozyme
355 357
(discussed in Chapter 12).
4. Modified Nucleotides
The picture of DNA or RNA as chains of only four
kinds of nucleotides is not quite accurate. DNA contains
a significant number of methylated bases among which
are 5-methylcytosine (5mC or m
5
C) and 6-methyl-
adenine (6mA). The former is regularly present in the
nuclear DNA of higher animals and plants. In mam-
mals from two to seven percent of the cytosine is
methylated. It is likely that these methylated bases
mark special points in the genetic blueprint. As is
discussed in Chapter 27, methylation, which is accom-
plished after the synthesis of the polynucleotide, may
block the expression of certain genes.
358
This appears to
happen when one of the two X-chromosomes becomes
inactivated in cells of females. Methylation may also
be used to imprint certain genes, i.e. to mark them as
coming from a specific one of the two parents.
359
Another function of methylation is to protect
DNA from attack by enzymes (restriction endonu-
cleases) formed in response to invading viruses
(Chapter 26). Some viruses, notably the bacteriophage
of the T-even series that attack E. coli (Box 7-C), have
developed their own protective devices. They contain
5-hydroxymethylcytosine (HOMeC) in place of cyto-
sine. The extra hydroxyl groups provided in this
fashion often carry one or two glucose units in glyco-
sidic linkage.
360
A bacteriophage attacking Bacillus
subtilis substitutes hydroxymethyluracil for uracil and
5-dihydroxypentyluracil for thymine, and phage W14
of Pseudomonas acidovorans substitutes the 5-methyl of
thymine with CH
2
NH (CH
2
)
4
NH
3
+
.
360,361
The modifications carried out on RNA molecules
are more varied and more extensive than those of DNA.
Sixty or more modification reactions are known for
tRNA, with the number and the extent of modification
depending upon the species. Structures of some of the
modified bases are indicated in Fig. 5-33. Uridine can
be methylated either on the base or on the 2'-hydroxyl
of the sugar. Methylation at the 5 position of uridine
yields ribothymidine. Cytidine can be modified in the
same positions. Reduction of the 5,6 double bond of
uridine gives dihydrouridine (hU). Replacement of
the oxygen at position 4 by sulfur gives 4-thiouridine
(
4
sU). Positions in the guanosine structure that can be
methylated are also indicated in Fig. 5-33. The symbol
m is commonly used to designate methylation in
nucleic acid bases; m
2
indicates dimethylation, e.g.,
6,6-dimethyladenine is abbreviated m
2
6
A.
A remarkable transformation is that of uridine into
pseudouridine ().
HN NH
O
O
O
CH
2
OH OH
HO
Pseudouridine ()
N
NH
O
O
O
CH
2
OH OH
HO
N
NH
O
NH
2
O
CH
2
OH OH
HO
N
N
N
N
N
N N
O
CH
3
CH
3
Ribose
HN
H
C
C
O
OCH
3
O
OCH
3
N
N
N
N
NH
Ribose
HN N
H
O
COO

CH
3
H
HO
H
5 3
Double bond
is reduced in
dihydro U (hU)
m
O is replaced by S
in 4-thiouridine
Abbrev:
4
S
m
Uridine
OH
OH
NH
2
CH
2
N
NH
N
Ribose
NH
2
O
6
1 7
m
Guanosine
3
m
m
m or m
2
Adenosine is modified in
6 position as follows in
threonylcarbamoyladenosine (tA)
Nucleoside Y
(Abbrev: iA) found at 3' end of anticodons
ending in A (codons starting with U) +
6
found in wobbling positions of anticodons
H
H
N
6
-(
2
-Isopentenyl)adenosine
Nucleoside Q (Queosine)
H
a cytokinin
Figure 5-33 Structures of some nucleosides containing
modified bases and found in tRNA molecules. Positions
where methylation may occur are designated m. Some other
abbreviations are e (ethyl), ac (acetyl), and f (formyl).
25
235 E. The Genetic Code
BOX 5-C THE RNA WORLD
The discovery in the 1980s that RNA molecules
often have catalytic properties and may serve as
true enzymes (ribozymes; Chapter 12) stimulated
new thinking about evolution. Although RNA
catalysts are not as fast as the best enzymes they are
able to catalyze a wide variety of different reactions.
Could it be that in the early evolution of organisms
RNA provided both the genetic material and cata-
lysts? The RNA world would have been indepen-
dent of both DNA and protein.
a,b
Later DNA could
have been developed as a more stable coding mole-
cule and proteins could have evolved as more effi-
cient catalysts. Plausible reactions by which both
cytosine and uracil could have arisen in drying
ponds on early Earth have been demonstrated.
c
A major objection to the RNA world is the lack
of stability of ribose and the inability to demonstrate
the nonenzymatic synthesis of ribose in significant
amounts. Even if ribose were present, it would be
largely in the pyranose ring forms. Initial formation
of the 5-phosphate would be required to allow
formation of a nucleotide with a furanose ring.
These and other obstacles to the RNA world have
led to the suggestion that some other genetic material
preceded RNA and DNA.
d,e
One possibility is a
peptide-like RNA analog.
e
A simple coding system
could also have been used, e.g. one based on only
two bases, such as C and G, instead of four.
f,g
Perhaps it is more probable that formation of
proteins and the present coding system evolved
simultaneously? The major metabolic cycles
(Chapter 10) could also have developed at the same
time. RNAs could not have been the first catalysts.
Hydrogen ions, hydroxyl ions, ammonium, cyanide,
and other simple ions as well as amines and peptides
could all have played a role in prebiotic chemistry.
Another speculation suggests an iron-sulfur world
in which organic materials would be formed on
mineral surfaces through reactions involving
reduction of bicarbonate by iron sulfide and H
2
S.
h
If the RNA world did exist, has it left us with
any real clues? Benner et al. suggest that modern
metabolism is a palimpsest of the RNA world, a
parchment that has been inscribed two or more
times, with previous texts imperfectly erased and
therefore still partially legible.
i
If so, can we find a
way to read the text of the ancient RNAs?
a
Gesteland, R. F., Cech, T. R., and Atkins, J. F., eds. (1999) The
RNA World, 2nd ed. Cold Spring Harbor Lab. Press, Cold
Spring Harbor, New York
b
Orgel, L. E. (1994) Sci. Am. 271 (Oct), 77 83
c
Robertson, M. P., and Miller, S. L. (1995) Nature (London) 375,
772 774
d
Larralde, R., Robertson, M. P., and Miller, S. L. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 8158 8160
e
Bhler, C., Nielsen, P. E., and Orgel, L. E. (1995) Nature (London)
376, 578 581
f
Sievers, D., and von Kiedrowski, G. (1994) Nature (London) 369,
221 224
g
Piccirilli, J. A. (1995) Nature (London) 376, 548 549
h
Maden, E. H. (1995) Trends Biochem. Sci. 20, 337 341
i
Benner, S. A., Ellington, A. D., and Tauer, A. (1989) Proc. Natl.
Acad. Sci. U.S.A. 86, 7054 7058
Pseudouridine is formed by enzymatic rearrangement
of uridine in the original transcript (Eq. 28-3). It can
form a base pair with adenine in the same manner as
does uracil. Pseudouridine is found not only in tRNA
but also in several places in both large and small ribo-
somal RNA subunits. For example, it is present at
position 516 in the E. coli 16S RNA,
364
at a specific
position in the 23S RNA, and at many more locations
in eukaryotic rRNA.
The bases called Y and Q are highly modified
guanines (Fig. 5-33). Q is found at the 5' end of some
anticodons in the wobble position (see Fig. 5-30).
Two hypermodified adenosines are also shown in Fig.
5-33. The N
6
-isopentenyladenosine is found at the 3'
end of the anticodons that pair with codons starting
with U. This compound is also a plant hormone, a
cytokinin (Chapter 30). Another highly modified
purine, threonylcarbamoyladenine, occurs adjacent
to the end of anticodons pairing with codons starting
with A. The function of these hypermodified bases is
uncertain, but they appear to promote proper binding
to ribosomes. The modifications are often not abso-
lutely essential for function.
Another source of modified bases in both DNA
and RNA is spontaneous or accidental alteration.
Nucleic acids encounter many highly reactive and
mutagenic materials including hydroxyl radicals, formed
from O
2
, and are able to convert guanine rings into
7,8-dihydro-8-oxoguanine.
362
Other reactive and carci-
nogenic compounds can form adducts with nucleic
acid bases.
363
See Eq. 5-18 and also Chapter 27.
5. RNA Aptamers
Ellington and Szostak
365
synthesized a random
pool of ~10
15
different oligodeoxyribonucleotides,
each ~100 nucleotides in length. They amplified
these using the polymerase chain reaction (PCR; Sec-
tion H,6) and prepared a mixture of the corresponding
RNAs by in vitro transcription. From the ~10
13
differ-
ent sequences still present they selected individual
236 Chapter 5. The Nucleic Acids
oligonucleotides by affinity chromatography on col-
umns that contained well-defined immobilized ligands
such as organic dyes. They called the selected RNAs
aptamers. Their approach is being used to find RNA
sequences that bind to such ligands as ATP, FMN,
366
the bronchodilator theophylline,
367
aminoglycoside
antibiotics,
368
arginine,
369
etc.
370
Many of the selected
aptamers bind their ligands very tightly and studying
them may shed light on interactions of RNA with
proteins and on the catalytic activities of RNA, which
are discussed in Chapter 12.
E. The Genetic Code
The general nature of the genetic code was sug-
gested by the structure of DNA. Both DNA and pro-
teins are linear polymers. Thus, it was logical to suppose
that the sequence of the bases in DNA codes for the
sequence of amino acids. There are only four bases in
DNA but 20 different amino acids in proteins at the
time of their synthesis. It is obvious that each amino
acid must be specified by some combination of more
than one base. While 16 pairs of bases are possible,
this is still too few to specify 20 different amino acids.
Therefore, it appeared that at least a triplet group of
three nucleotides would be required to code for one
amino acid.
371
Sixty-four (4
3
) such triplet codons
exist, as is indicated in Tables 5-5 and 5-6.
Simplicity argues that the genetic blueprint speci-
fying amino acid sequences in proteins should consist
of consecutive, nonoverlapping triplets. This assump-
tion turned out to be correct, as is illustrated by the
DNA sequence for a gene shown in Fig. 5-5. In addi-
tion to the codons that determine the sequence of
amino acids in the protein, there are stop codons that
tell the ribosomal machinery when to terminate the
polypeptide chain. One methionine codon serves as
an initiation codon that marks the beginning of a
polypeptide sequence. One of the valine codons
sometimes functions in the same way.
How does a cell read the code? This question is
dealt with in detail in Chapters 28 and 29. A key step
is the positioning of each amino acid on the ribosome
in proper sequence. This is accomplished by the pair-
ing of codons of messenger RNA with the anticodons
of the appropriate transfer RNA molecules as is indi-
cated at the bottom of Fig. 5-30. Each tRNA carries the
appropriate activated amino acid at its 3' end ready
to be inserted into the growing peptide.
1. The Reading Frames
It is immediately obvious that there are three ways
of reading the genetic code in mRNA depending upon
which nucleotide is used to start each codon. For
example, in the following mRNA sequence either
codons GCA, CAG, or AGC could be selected as first.
These codons define the three reading frames or phases
in which the code may be read. Here, the term frame
Alanine GCX 4
Arginine CGX, AGA, AGG 6
Asparagine AAU, AAC 2
Aspartic acid GAU, GAC 2
Cysteine UGU, UGC 2
Glutamic acid GAA, GAG 2
Glutamine CAA, CAG 2
Glycine GGX 4
Histidine CAU, CAG 2
Isoleucine AAU, AUC, AUA 3
Leucine UUA, UUG, CUX 6
Lysine AAA, AUG 2
Methionine
(also initiation codon) AUG 1
Phenylalanine UUU, UUC 2
Proline CCX 4
Serine UCX, AGU, AGC 6
Threonine ACX 4
Tryptophan UGG 1
Tyrosine UAU, UAC 2
Valine
(GUG is sometimes
an initiation codon) GUX 4
Termination UAA (ochre)
UAG (amber)
UGA 3
Total 64
TABLE 5-5
The Genetic Code
a
Amino acid Codons
Total number
of codons
a
The codons for each amino acid are given in terms of the sequence
of bases in messenger RNA. From left to right, the sequence is
from the 5' end to the 3' end. The symbol X stands for any one of
the four RNA bases. Thus, each codon symbol containing X
represents a group of four codons.
5 G C A U G C G A U G G U C A C G A U U A A U 3
1
2
3
Ser Val
Met Val Thr Ile Asn
Arg Stop. Ser Trp
237
U(T) Phe Ser Tyr Cys U(T)
Phe Ser Tyr Cys C
Leu Ser Term
c
Term
d
A
Leu Ser Term Trp G
C Leu Pro His Arg U(T)
Leu Pro His Arg C
Leu
a
Pro Gln Arg A
Leu Pro Gln Arg G
A Ile Thr Asn Ser U(T)
Ile Thr Asn Ser C
Ile Thr Lys Arg A
Met
b
Thr Lys Arg G
G Val Ala Asp Gly U(T)
Val Ala Asp Gly C
Val Ala Glu Gly A
Val
b
Ala Glu Gly G
TABLE 5-6
The Sixty-Four Codons of the Genetic Code
Middle base
a
The codon CUA (CTA) encodes threonine and the codon AUA
(ATA) methionine in mammalian mitochondria.
b
Initiation codons. The methionine codon AUG is the most com-
mon starting point for translation of a genetic message but GUG
can also serve. In such cases it codes for methionine rather than
valine.
c
The termination codon UAA (TAA) encodes glutamine in
Tetrahymena.
d
The termination codon UGA (TGA) encodes tryptophan in
mitochondria and selenocysteine in some contexts in nuclear genes.
5OH
Terminal
base
3OH
Terminal
base U(T) C A G
does not designate a single codon, although frame
does designate a single exposure in a motion picture
film. Reading frame designates which of the three
possible sets of codons we are using. In the foregoing
sequence the codons in reading frames 2 and 3 are
labeled. Reading frame 2 contains the initiation codon
AUG (shaded) which could mark the beginning of an
encoded protein sequence. Reading frame 3 contains a
termination (stop) codon which, when the mRNA tran-
script is read by ribosomes, will terminate polypeptide
synthesis. It may be in the position shown but not
have any real function. However, it could represent
the end of the coding sequence that is marked if genes
for the two proteins overlap a little at the ends, a situa-
tion that actually occurs in nature.
A reading frame in a specified part of a DNA
sequence is said to be open if there is an initiation
codon preceded by suitable regulatory signals (an
operator region). This means that it could encode a
protein. The reading frame of a sequence is open until
the next termination codon. Recently another usage
has appeared. Many writers refer to an open reading
frame as a segment of DNA in which any one of the
three reading frames is open. Another complexity in
the reading of genetic messages arises because splicing
of RNA may sometimes cause a shift in the reading
frame. For example, a mRNA being transcribed from
the sequence in reading frame 2 in the foregoing exam-
ple may skip over a nucleotide part of the time to form
an RNA in which the first part is encoded by reading
frame 2 and the second by reading frame 3 (a + 1 frame-
shift). Alternatively, a nucleotide could be read twice
with a 1 frameshift with the sequence of reading
frame 1 for the latter part of the mRNA. Frameshifts
can also occur during protein synthesis as the mRNA
is being read.
In the present example we have examined the
sequence in mRNA. In the DNA there are two strands.
One is the coding strand (also called the nontranscrib-
ing or nontranscribed strand), which has a sequence
that corresponds to that in the mRNA and the one that
is given in Fig. 5-4. The second antiparallel and com-
plementary strand can be called the template strand
or the noncoding, transcribing, or transcribed strand.
372
The mRNA that is formed is sometimes referred to as a
sense strand. The complementary mRNA, which
corresponds in sequence to the noncoding strand of
DNA, is usually called antisense RNA.
2. Variations
Is the genetic code universal or does it vary
from one organism to another? Studies with bacteria,
viruses, and higher organisms including humans have
convinced us that the code is basically the same for all
oganisms. However, there are some variations. For
example, in mitochondria of both humans and yeast
the codon TGA is not a termination codon but repre-
sents tryptophan. In mammalian mitochondria CTA
represents threonine rather than leucine, and ATA
encodes methionine instead of isoleucine. These differ-
ences in the code are related to the fact that mitochon-
dria contain their own piece of DNA. It encodes not
only several proteins but also tRNA molecules whose
anticodon structures are altered to accommodate the
changed meanings of the codons of the mitochondrial
DNA and mRNA.
373,374
Variations in the code for cytoplasmic proteins
have been found. In Tetrahymena and other ciliates the
codon TAA represents glutamine rather than being a
termination codon.
375
A few proteins, including some
in the human body, contain selenocysteine, the sele-
nium-containing analog of cysteine. Selenocysteine is
encoded by termination codon TGA. See Chapter 29
for details. However, even though TGA is occasionally
used in this way, it serves as a termination codon for
most proteins within the same cells.
376
Thus, the
context in which the codon TGA occurs determines
how it is read by the ribosomal machinery.
E. The Genetic Code
238 Chapter 5. The Nucleic Acids
loop of the other to form a triplex base structure. A
related triplex structure may be formed when inverted
repeat sequences occur within a homopyrimidine tract
in one chain of DNA (Fig. 5-34B).
378
These have been
called mirror-repeats or H-palindromes to distin-
guish them from true palindromes.
237,379
Each base
triplex structure contains one set of WatsonCrick
hydrogen bonds and one set of Hoogsteen hydrogen
bonds. The triplexes of H-DNA are all either TAT or
CGC
+
, where one C is protonated (see also Fig. 5-24).
3. Palindromes and Other Hidden Messages
The sentence Madam, Im Adam reads the same
either forward or backward. Such sentences, known
as palindromes,
377
are infrequent in the English lan-
guage. However, most DNA contains many palin-
dromes, sequences of base pairs that read the same in
forward and reverse directions. Consider, for example,
the gene that specifies the sequence of nucleotides for
the tRNA molecules of Fig. 5-30. It
is a double-stranded DNA segment
in which one strand has a sequence
identical to that of Fig. 5-30 except
for the substitution of T for U and
for (pseudouridine) and for the
lack of methylation and other base
modifications. The second strand is
the exact complement. Figure 5-34
shows the part of this gene (residues
49 76) that corresponds to the 3'
end of the tRNA molecule. This
DNA segment could exist in a sec-
ond conformation having a loop on
each side of the molecule (Fig. 5-34).
The stems of the two loops in this
cruciform conformation are iden-
tical and symmetrically disposed
around the center of the molecule.
If we overlook the seven nucleotides
in the center of the loop, the message
in the stems reads the same in both
directions, as is indicated by the
green arrows.
Palindromes are often imperfect
as is the one shown in Fig. 5-34. Here
the two stems in the cruciform struc-
ture are related by an exact twofold
rotational symmetry but the loops
at the ends of the stems are not.
Unpaired bases may bulge at various
points in double-stranded stems of
longer palindromes. These imper-
fect palindromes in the DNA are
responsible for much of the tertiary
structure of the various kinds of
RNA. The tertiary structure, in
turn, often determines the interac-
tion of the RNA with enzymes and
other proteins.
Special properties may be ob-
served for palindromes containing
homopurine tracts in one strand, and
therefore homopyrimidine tracts in
the other. If two identical palindromes
of this type occur close together it is
possible that the pyrimidine-containing
strand of one can join with a hairpin
Figure 5-34 (A) Two conformations of a segment of the yeast phenylalanine
tRNA gene. The segment shown codes for the 3' end of the tRNA molecule
shown in Fig. 5-30, including the TC loop. (B) Formation of H-DNA (Fig.
5-24) proposed for a sequence in plasmid pGG32. The major element of the
structure is the triplex, which is formed from the WatsonCrick duplex (

)
associated with the homopyrimidine loop through Hoogsteen base pairing
(

, +). One of the two possible isomeric forms is shown. See Mirkin et al.
378
Inverted repeats
Triple helix
G A T C C T T C C C T C T T C C C C C T A T A C C C C C T T C T C C C T T G A A
C T A G G A A G G G A G A A G G G G G A T A T G G G G G A A G A G G G A A C T T
5
3
G A T C C T T C C C T C T T C C C C C T
C T A G G A A G G G A G A A G G G G G
A
5
3
T T C C C T C T T C C C C C T
A
T
T
A
T
G
G G G A G A A G G G G
G C
A T
A T
T A
T A
3 5
A
A
3
H- DNA
One of two isomeric forms
A C C A C
5
3
3
G C T T A A G A C A C C T A G C T T G T G T C C T G 5
T G G T G C G A A T T C T G T G G A T C G A A C A C A G G A C
A C C A C
5
3 G C T T A A
C
T
A
G
C
T
T
T G G T G C G A A T T
G
A
T
C
G
A
A
3
C T G 5
G A C
T A
G C
T A
C G
G C
A T
C G
A T
C G
G C
CT
TC loop
Forward
Reverse
A
B
239
Segments of H-DNA might block transcription, and
proteins that bind to the triplex H-DNA may be in-
volved in transcriptional regulation.
379
Until rather recently there had been little to indi-
cate that DNA actually assumes cruciform conforma-
tions in cells. However, strong experimental evidence
suggests that some cruciform structures do form natu-
rally.
380
Their formation from palindromic DNA [like
the formation of Z-DNA from (G + C)-rich sequences]
is a way of relieving torsional strain induced by super-
coiling. Whether or not cruciform structures occur
frequently within cells, there is no doubt that palin-
dromic sequences are of great importance in the inter-
action of nucleic acids with symmetric dimeric and
tetrameric protein molecules such as the gene repressor
protein shown in Fig. 5-35.
381 383
DNA contains numerous other protein binding
sites which are not palindromes but whose sequences
represent additional encoded information. The RNA
transcripts likewise contain sequences that direct the
catalytic machinery involved in splicing, that bind to
ribosomal proteins, that control rates of transcription,
and that cause termination of transcription.
4. The Base Composition of DNA and RNA
The nucleotide composition of DNA is surprisingly
variable. The sum of the percentage cytosine plus the
percentage guanine (C + G) for bacteria varies from 22
to 74%. That for E. coli is 51.7%. Among eukaryotic
organisms, the range is somewhat narrower (28 to
58%; for humans, 39.7%). The fact that bacterial DNA
molecules are more varied than those of higher organ-
isms is not surprising. The prokaryotes have evolved
for just as many more million of years as have we.
Perhaps because of their simpler structure and rapid
rate of division, nature has done more experimentation
with genetic changes in bacteria than in people.
Comparisons of the C + G content of organisms
have been used as a basis for establishing genetic
relatedness.
384
However, since thymine is especially
susceptible to photochemical alteration by ultraviolet
light, bacteria with a high (C + G) content may have
evolved in environments subject to strong sunlight or
high temperatures, whereas those with a low (C + G)
content have developed in more protected locations.
385,386
F. Interaction of Nucleic Acids with Proteins
Most functions of DNA and RNA are dependent
on proteins. Cells contain a vast array of polypeptides
that bind to nucleic acids in many specific ways.
387 390
We have seen (Fig. 5-21) that the histones hold super-
coiled DNA in the form of nucleosomes
391
in eukaryotic
nuclei and similar proteins bind to and protect the
double-stranded DNA of bacteria.
205,392
Sperm cells
package a large amount of DNA into a small space
with the help of small arginine-rich proteins called
protamines.
393
Cells always have some single-stranded
DNA segments as well as single-stranded DNA bind-
ing (SSB) proteins. Among the latter are proteins
from E. coli
394,395
and from viruses.
396 399
Specialized
proteins bind to DNA sequences in telomeres
38,272
and
centromeres.
400
A large number of proteins interact
with RNA in ribosomes, spliceosomes, and other
complexes.
A host of enzymes, which are described elsewhere
in the book, act on DNA and RNA. They include hydro-
lytic nucleases, methyltransferases, polymerases,
topoisomerases, and enzymes involved in repair of
damaged DNA and in modifications of either DNA
or RNA. While most of these enzymes are apparently
proteins, a surprising number are ribozymes, which
consist of RNA or are RNAprotein complexes in
which the RNA has catalytic activity.
1. The HelixTurnHelix Motif
Much current interest in DNAprotein interactions
is focused on regulatory processes. In prokaryotes the
initiation of transcription of a large fraction of the genes
is blocked by the binding of proteins known as repres-
sors. While their structures are varied, one large group
of repressors have DNA-binding domains with a
similar helixturnhelix architectural motif. They
bind with high affinity to specific control regions
which contain palindromic DNA sequences such as
the following one, which defines a binding site for the
E. coli trp (tryptophan) repressor:
These repressor proteins form dimers joined through a
rigid central domain with a pair of arms, each contain-
ing two helices that form the helixturnhelix motif.
One helix of each pair fits into the major groove of the
DNA helix and forms a reading head that carries a
specific arrangement of amino acid side chains able to
locate the symmetric nucleotide sequences flanking
the twofold axis of the palindrome. In the case of the
trp repressor, these are the ACTAGT hexanucleotides
(marked by the arrows in the preceding structure).
The interaction is depicted in Fig. 5-35.
401 405
The
repressor has a high affinity for the DNA only if one
molecule of L-tryptophan is bound to each subunit at
a specific site near the DNA helix. Binding of the
tryptophan causes a conformational change, which
is pictured in Fig. 5-35. If tryptophan is absent the
F. Interaction of Nucleic Acids with Proteins
5
3
3
5
T G T A C T A G T T A A C T A G T A C
T C A T G A T C A A T T G A T C A T G
240 Chapter 5. The Nucleic Acids
repressor binds to the palindromic DNA only weakly
and transcription of genes needed for tryptophan
biosynthesis occurs freely. However, if tryptophan
accumulates within the cell it binds to the repressor
molecules causing them to bind firmly to DNA and
prevent transcription. There are at least three of these
palindromic sequences in E. coli, each one regulating
a set of genes (operons) involved in tryptophan syn-
thesis. In contrast to the effect of tryptophan, the
tryptophan analog indole-3-propionate, which lacks
the amino group of tryptophan, derepresses the same
operons. It also binds to the trp repressor, but with the
indole ring flipped over by 180 so that its carboxylate
group contacts the phosphate groups of the DNA
repelling them through both electrostatic and steric
effects.
406,407
The picture in Fig. 5-35 is an oversimplifi-
cation. In fact, at some binding sites (operator sites)
more than one dimeric repressor binds in a tandem
fashion.
405
The helixturnhelix motif is also found in many
other proteins. One of these is the bacterial lac (lactose)
repressor which controls the lac operon and for which
Figure 5-35 Stereoscopic
drawings illustrating the
binding of a dimeric molecule
of the Trp repressor protein
to a palindromic sequence in
DNA. (A) Schematic view
showing structures of the
aporepressor (partly shaded
gray) and the holorepressor
with bound tryptophan
(unshaded) are superimposed.
Cylinders represent the
helices in (B). From Zhang
et al.
402
(B) MolScript ribbon
diagram with a few side chains
that interact with the DNA
shown. Two tandemly bound
dimeric repressor molecules
are shown. Two bound mole-
cules of tryptophan are visible
in each dimer. The DNA is
drawn as a double helix with
lines representing the base
pairs. From Lawson and
Carey.
405
B
A
241 F. Interaction of Nucleic Acids with Proteins
the terms operator, promoter, repressor, and operon
were first introduced (Chapter 28).
408 410
Some bacteri-
al viruses, such as phage lambda () of E. coli, encode
repressors that allow the virus to reside in the bacteria
without immediately destroying them. The repressor
and other closely related proteins also utilize the helix
turnhelix motif
411 415
as do some proteins that
activate transcription (see Chapter 28).
2. Other DNA-Binding Motifs
Nobody knows how many different DNA-binding
structures may be discovered. However, most of those
that are designed to recognize specific DNA sequences
have some part that fits into the major groove of B-
DNA.
416
Usually the DNA structure must be in a
specific form: B-, A- or Z-, but it may sometimes be
bent or distorted. The DNA recognition motifs in the
proteins may consist of helices, strands, or loops.
Leucine zipper proteins. Several transcription
factors have the leucine zipper structure, which has
been described in Chapter 2 and was illustrated there
by the structure of transcription factor Max (Fig. 2-21).
417
Related structures include those of the transcription
activators c-Jun and c-Fos
418
and of the yeast transcrip-
tion factor GCN4.
419,420
The latter is a dimeric protein
in which the C-terminal halves of the two monomers
form the helices of the leucine zipper. The helices,
which are continuous for over 60 residues, fan out to
interact with the DNA double helix as in Fig. 2-21. A
19-residue basic domain of each protein helix crosses
the major groove of the DNA with side chain groups
interacting with the DNA as is illustrated in Fig. 5-36.
Zinc fingers. Another large group of transcrip-
tion factors contain a bound zinc ion in a finger
motif. The Zn
2+
is held by two cysteine S

groups
present on a loop of structure and by two imidazole
groups present on an helix (Fig. 5-37).
421 424
Amino
acid sequences found in many proteins that regulate
transcription tend to form zinc fingers and to interact
with DNA. These were recognized first in the transcrip-
tion factor TFIIIA from Xenopus laevis in 1985.
421,425,426
This protein contains a zinc finger motif repeated nine
times. X-ray structures, one of which is shown in Fig.
5-38, are known for proteins with three
423
and five
427
zinc fingers. (See Chapter 28.)
Beta ribbons. An antiparallel double-stranded
ribbon can fit into the major groove of DNA and form
A
B
Figure 5-36 Stereoscopic
diagrams showing some of
the interactions between an
N-terminal helical domain of
the yeast transcription factor
GCN4-bZIP, a leucine zipper
protein, and a specific palin-
dromic DNA binding site:
(The bases are numbered
outward from the central C
and G.) The small solid
spheres are water molecules.
Notice the water mediated
interactions of the basic
arginine and lysine side
chains with the nucleic acid
bases and also the interaction
of R240 and R243 (in B) with
a backbone phosphate. The
overall structure of the protein
is similar to that of another
leucine zipper shown in Fig.
2-21. From Keller et al.
419
Drawings courtesy of Timothy
J. Richmond.
5 A T G A C G T C A T
5 1 1 5
242 Chapter 5. The Nucleic Acids
Figure 5-38 (A) Sketch of a complex of a
five-zinc finger protein bound to a high-
affinity site in DNA. This protein is a
human oncogene called GL1. Its gene is
amplified in many tumors, but it is similar
to many other transcription factors. The
cylinders and ribbons highlight the
helices and sheets. Cobalt ions, which
replaced the Zn
2+
in the crystals, are
shown as spheres. (B) Stereoscopic view
of the complex in a similar orientation.
(C) Stereoscopic view emphasizing the
contacts of side chains from fingers 5 and 6
with the DNA. The view is similar to that
in (A) and (B) but the structure has been
tilted back. Drawings courtesy of Nikola
Pavletich.
427
B
C
A
Figure 5-37 Three-dimensional structure of a zinc finger.
This is formed by the binding of Zn
2+
to the following se-
quence in a protein:
Here is a hydrophobic amino acid and X may be any
amino acid. After Krizek et al.
428
Zn
Leu
Phe
Tyr
17
11 2
O
H
His
20 Cys
4
7 Cys His
N
N
N
H
H
24
N
S
S
X C X
24
C X X X F X
5
L X X H X
3,4
H
turn

turn
helix
243
hydrogen bonds and other interactions with the DNA.
This is the basis for specific recognition of the operator
sequence for the E. coli met (methionine) repressor as
is illustrated in Fig. 5-39.
429
The same binding motif is
employed by some other repressors
430,431
and also by
the abundant bacterial DNA-binding protein HU.
432
It has also been utilized in designing the previously
mentioned hairpin polyamide DNA-binding com-
pounds.
The winged helix family. A group of large pro-
tein transcription factors contain an N-terminal DNA-
binding domain with the striking winged helix motif
shown in Fig. 5-40.
433 436
It occurs in proteins from a
wide range of organisms from yeast to human.
3. RNA-Binding Proteins
Among the many proteins that bind to RNA mole-
cules
437 439
are the aminoacyl-tRNA synthetases, a
variety of other well known enzymes,
440
the ribosomal
proteins discussed in Chapter 29, and various proteins
with dual functions of catalysis and regulation of
Figure 5-40 Structure of a protein known as transcription
factor NF-B bound to its DNA target. Each subunit of the
dimeric protein contains two barrel domains. The loops at
the ends of the barrels interact with the DNA in the center.
From Mller et al.
433
Courtesy of Stephen C. Harrison.
Figure 5-39 (A) Stereoscopic
ribbon representation of the E. coli
methionine repressoroperator
complex. Two subunits form a
dimer with a double-stranded
antiparallel ribbon that fits into
the major groove of the DNA in
the B form. One strand is shaded
more darkly than the other.
(B) View of the ribbon and its
interactions with the DNA.
Notice the direct hydrogen bonds
from amino acid side chains of
lysine and threonine residues to
bases in the specific palindromic
sequence AGACGTCT. From
Somers and Phillips.
429
Drawings
courtesy of S. E. V. Phillips.
B
A
F. Interaction of Nucleic Acids with Proteins
244 Chapter 5. The Nucleic Acids
containing virus 25 nm in diameter, is only three times
as thick as the thinnest cell membrane. Its DNA contains
just 5386 nucleotides.
450 452
The similar bacteriophage
G4 contains 5577.
453,454
It is remarkable that such tiny viruses are able to
seize control of the metabolic machinery of the cell and
turn it all in the direction of synthesis of more virus
particles. There are only 11 known proteins encoded
by the genes of X or G4. Three genes encode the
three kinds of protein subunits of the virus coat. Sixty
copies of each are needed, as are 12 copies of a pilot
protein.
451
Eight of the genes are spaced closely together,
occupying most of the DNA. The other three genes
are embedded within some of the first eight but in
different reading frames. In one short region of the G4
chromosome the same nucleotides are part of three
different genes, using all three possible reading frames.
In addition to their own genes, these small viruses make
use of many components of the cell that they infect. A
large group of animal viruses, the parvoviruses, are
similar in size and architecture to bacteriophage X174.
Canine parvovirus, first identified in 1978, is now
endemic.
455,456
Childhood fifth disease is also caused
by a parvovirus.
456,457
When these single-stranded
DNA viruses infect cells a double-stranded replicative
form of DNA arises by synthesis of the complementary
negative strand alongside the original positive DNA
strand. Many copies of the replicative form are then
synthesized. The negative strands of the replicative
forms serve as templates for synthesis of numerous
new positive strands that are incorporated into the
progeny viruses. The whole process may take only 20
minutes. Some parvoviruses are unable to reproduce
unless the cell is also infected by a larger adenovirus.
While most plant viruses contain dsRNA, the
geminiviruses,
458
which cause a number of plant
diseases, contain single-stranded DNA. The virus
particle consists of a fused pair of incomplete icosahedra,
evidently containing a single ~ 2500-bp DNA strand.
Replication may require coinfection with two virus
particles of differing sequence.
2. Viruses with Double-Stranded DNA
One of the smallest viruses containing double-
stranded DNA is the 3180-nucleotide human hepatitis
B virus. It infects millions of people throughout the
world causing chronic hepatitis and often liver can-
cer.
459 461
The circular DNA is surrounded by an
envelope consisting of proteins, carbohydrate, and a
lipid bilayer. Many icosahedral viruses also contain
dsDNA. Among them are the papovaviruses, some
of which cause warts and others malignant tumors.
Much studied by biochemists is the 5386-nucleotide
simian virus 40 (SV40), a monkey virus capable of
inducing tumors in other species.
462,463
Closely related
translation. A widely found RNA-binding
ribonucleoprotein domain (RNP domain), occurs
in hundreds of proteins including many of the RNA
processing proteins considered in Chapter 28.
437,441,442
This 70 90 residue module binds an RNA strand
against a sheet surface. Another ~ 70-residue protein
motif binds ds RNA.
443
G. Viruses and Plasmids
Attacking every living thing from the smallest
mycoplasma to human beings the nucleoprotein particles
known as viruses have no metabolism of their own.
However, they come alive when the nucleic acid
that they contain enters a living cell. Viruses are sig-
nificant to us not only because of the serious disease
problems that result from their activities but also as
tools in the study of molecular biology. A mature
virus particle or virion consists of one or more nucleic
acid molecules in a protein coat or capsid, usually of
helical or icosahedral form. The capsid is made up of
morphological subunits called capsomers. They can
sometimes be seen clearly with the electron microscope.
The capsomers in turn are usually composed of a
number of smaller protein subunits. Some of the larger
viruses are surrounded by membranous envelopes.
Others, such as the T-even bacteriophage which attack
E. coli, have extraordinarily complex structures (Box 7-C).
Most viruses contain a genome of either double-
stranded DNA or single-stranded RNA, but some
small viruses have single-stranded DNA and others
have double-stranded RNA. The number of nucleo-
tides in a virus genome may vary from a few thousand
to several hundred thousand and the number of genes
from 3 to over 200. Sometimes the nucleic acid mole-
cules within the virion are circular, but in other cases
they are linear. Table 5-7 lists a few of the known types
of viruses as well as some individual viruses.
72,444
The
size of the genome, in kilobases (kb), or kilobase pairs
(kbp) is indicated. The number of genes in a virus is
often somewhat more than one per kbp of DNA. A
vast amount of information is available about these
infectious particles. Only a few reference sources are
cited here.
72,444 446
The architectures of some helical
and icosahedral protein coats that surround genomic
DNA or RNA are described further in Chapter 7.
1. Viruses with Single-Stranded DNA
The very small helical bacteriophages of the Ff
family, such as fd, f1, and M13 (see Fig. 7-7) resemble
thin bacterial pili but each virus particle contains a mole-
cule of single-stranded circular ~ 6400 nucleotide DNA
of M
r
~ 2 x 10
6
which encodes only ten proteins.
447 449
Bacteriophage X174 (Fig. 5-41), an icosahedral DNA-
245
DNA, single-stranded
Bacteriophages fd, f1, M13 H ~6 x ~880 (length) 17.6 2.1 6.4
Bacteriophages 174, G4 I 25 6.2 1.8 5.45.6
Parvoviruses I 1825 1.8 5.5
Geminiviruses I (fused pairs)
DNA, double-stranded
Hepatitis B virus I (enveloped) 2.1 3.18
Papoviruses I 4555
SV40 (monkey) I 17.6 3.5 5.22
Polyoma (mouse) I 45 23.6 3.3 4.96
BK virus (human) I 3.4 4.96
Papilloma (human wart) I 56 5.3 8.0
Bacteriophage 29 T 12 19.3
Adenoviruses I 70 2030 3045
Bacteriophage Mu 25 38
Bacteriophage T7 T(short) 39.9
Bacteriophage P22 T 28.5 43.2
Bacteriophage T 32 48.6
T-even bacteriophage T 100 x 80 (head) 215 130 166
Baculoviruses of insects I 70130
Herpesviruses I 100
core 78 ~1000 80120 80140
envelope 150200
Pox viruses, e.g.,
Smallpox, vaccinia (cowpox) C 160 x 250 ~4000 150240 240300
Cauliflower mosaic virus I 8.0
RNA, single-stranded
Unsheathed
Potato spindle tuber viroid 0.116 0.30
Hepatitis delta virus 1.7
Sheathed, plus-strand
Tobacco necrosis satellite I 18 1.7 0.4 1.20
Small bacteriophages
R17, MS2, Q I 2326 3.64.0 1.21.5 3.54.5
Picornaviruses I 8.4 2.6 7.9
Polioviruses I 27 6.4 2 6.1
Rhinoviruses I 2730 78 2.22.8 6.78.5
Turnip yellow mosaic virus I 28 5.06.0 2.0 6.1
Tobacco mosaic virus H 18 x 300 40 2.2 6.7
Togaviruses I 2040 4 11
Negative-strand viruses
Influenza virus I 80100 200 2.0 6.1
Bullet-shaped viruses
Rhabdoviruses C 20 x 130
Retroviruses I 80100 710 2030
RNA, double-stranded
Reoviruses I 5560 1112 1618
Mobillivirus I (enveloped) 38
Rotavirus Wheel-shaped 70
Leishmania RNA virus 5.28
TABLE 5-7
Characteristics of Some Individual Viruses and Groups of Viruses
Masses in daltons x 10
6
Thousands of
bases or base pairs
(kilobases)
Type of genome
a
and group
or individual virus name
Shape
b
Diameter
c
(nm)
Total DNA or RNA
a
Complete nucleotide sequences are known for most of these viruses.
b
Shapes are indicated as I, icosahedral; H, helical; T, a tailed phage; C, complex.
c
The second dimension given for some helical and complex viruses is the length (nm).
G. Viruses and Plasmids
246 Chapter 5. The Nucleic Acids
Figure 5-41 Electron micrographs of some viruses:
(A) Bacteriophage L7, which resembles X174. See also
Fig. 7-28. Courtesy of A. S. Tikhonenko. (B) Adenovirus.
(C) Cytomegalovirus particles being released from a
fibroblast. (B) and (C) courtesy of T. Moninger. (D)
Tobacco mosaic virus. Omikron, Photo Researchers.
(E) A tailed bacteriophage. Courtesy of A. S. Tikhonenko.
B
C
A
E
D
247
is the polyomavirus of the mouse
464
and the BKvirus
of humans,
465
also suspected of causing cancer. The
papillomaviruses cause warts and perhaps cancer
466
and the larger (70 nm diameter) adenoviruses (Fig. 5-
41),
467,468
cause respiratory infections. Some 32 types
infect humans. Many of the details of eukaryotic
transcription were first studied using the adenovirus.
469
The very large herpesviruses are enveloped by a
lipid-containing membrane.
470 472
Among them are
herpes simplex viruses, which infect human mucous
membranes and varicella-zoster virus, which causes
chicken pox and shingles. Other herpesviruses include
cytomegaloviruses (Fig. 5-41), another common human
pathogen, and the EpsteinBarr virus, which causes
mononucleosis and is suspected of causing cancer.
473
Another herpesvirus has been associated with multiple
sclerosis.
474
The very large icosahedral baculoviruses
cause polyhedroses in insects. One that infects the fly
Tipula measures 130 nm in diameter. Poxviruses are
also large and complex.
475
The tailed bacteriophages
(Box 7-D) range in size from the small ~ 29-kDa phage
P22 to the very complex T-even phage. Some plant
viruses also contain dsDNA. One of these, the cauli-
flower mosaic virus,
476,477
is transmitted by aphids.
It has proved useful as a gene-transfer vehicle for
genetic engineering.
3. Viruses Containing RNA
Several plant diseases including the potato spindle
tuber disease are caused by viroids, molecules of
single-stranded RNA only 240 380 nucleotides in
length with a folded structure.
478 481
Such an RNA
could code for a protein containing only about 100
amino acids. However, the known 359-nucleotide
sequence of the potato spindle tuber virus contains no
AUG initiation codon. It seems impossible that the
virus carries any gene for a protein. Conserved features
of viroid sequences suggest a close relationship to the
intervening DNA sequences known as type I introns
(see Fig 28-19). Whatever its genetic message, a plant
viroid causes the plant cell to replicate many copies of
the viroid molecule, which may then be transmitted to
other plants by aphids, or on the surface of tools, by
humans. A larger 1678-nucleotide viroid-like RNA
(hepatitis delta virus) has been identified in human
patients with severe chronic hepatitis and who were
also infected with hepatitis B virus. The ssRNA in this
virus does encode at least one protein.
482 484
One of the smallest of the encapsulated RNA-
containing viruses is the satellite tobacco necrosis
virus. It replicates only when the plant is also infected
with the larger tobacco necrosis virus. The satellite
virus, whose three-dimensional structure is known from
X-ray diffraction studies,
485
contains a 1200-nucleotide
strand of RNA which encodes a 195-residue protein.
Sixty copies of the latter are assembled around the
RNA in an icosahedral array (Fig. 7-14) to form the
virion. The structure of the similar satellite tobacco
mosaic virus has also been described in detail.
486,487
RNAs of the small bacteriophages f2, R17, MS2
and Q contain 3500 4500 nucleotides and are
enclosed in icosahedral shells made up of 90 identical
subunits.
488 490
Initially only three genes were evident
but a fourth small gene in a different reading frame
has since been found in at least one of them.
491
The
RNA molecules in these and other positive-strand
viruses serve as mRNA within the host cells. Another
group of small RNA viruses are the picornaviruses
(picoRNA, meaning very little RNA). Many of these
icosahedral viruses of 15 30 nm diameter attack hu-
mans. Among them are the enteroviruses including
the polioviruses,
492,493
the hepatitis A virus,
494
the
coxsackieviruses, and some of the echoviruses. A
second class of picornaviruses include rhinoviruses
which cause the common cold. More than 100 types
are known.
495 497
The foot andmouth disease
virus,
498 500
which attacks the cloven-footed animals,
and the Mengo virus,
501
which can cause a fatal
encephalitis in mice, are also picornaviruses. The
three-dimensional structures of polio virus, human
rhinoviruses, Mengo virus, and many other eicosahedral
viruses are known. Their architecture is discussed in
Chapter 7.
The togaviruses, which are a little larger than the
picornaviruses, have an icosahedral core surrounded
by a lipid membrane. Yellow fever and rubella (Ger-
man measles) are both caused by togaviruses. Other
togaviruses, such as Sindbis virus
502
and Semliki
Forest virus,
503
have become important in biological
research.
A large number of icosahedral RNA viruses of
diameter 28 30 nm (Fig. 7-14) attack plants, causing
diseases such as tomato bushy stunt,
504
southern bean
mosaic,
505
or turnip yellow mosaic. Best known of the
helical RNA viruses is the tobacco mosaic virus (Figs.
5-41, 7-8).
506-507a
Its genome contains 6395 nucleotides
as linear ssRNA. Many strains are known. Related
viruses cause cucumber green mottle
508
and other
plant diseases.
Large viruses of 80 100 nm diameter bearing 810
spikes at the vertices of the icosahedra cause influen-
za,
509,510
mumps, measles, and related diseases. The
internal structure must be complex. Only 1% of the
virus is RNA, and that consists of several relatively
small pieces. These are negative strand viruses
whose RNA is of the opposite polarity to the mRNA.
The latter must be formed by transcription from the
negative strand. The viruses carry their own RNA
polymerase for this purpose. Of even more complex
structure are the bullet-shaped rhabdoviruses which
cause rabies and vesicular stomatitis.
511
The diameter
of these viruses is 65 90 nm and the length 120 500
G. Viruses and Plasmids
248 Chapter 5. The Nucleic Acids
nm. The internal structure includes a helical arrange-
ment of nucleoprotein. They also are negative strand
viruses.
Among the retroviruses
512 514
are types B and C
oncoviruses which induce malignant tumors in mam-
mals and birds
515
and the human immunodeficiency
virus (HIV), the apparent causative agent of AIDS.
Their RNA functions in a surprising way. Each virion
contains a reverse transcriptase, an enzyme that
transcribes copies of circular dsDNA copies from the
one or two mRNA-like molecules that make up the
virus genome. Following action of the reverse trans-
criptase, one of the transcribed DNA circles becomes
covalently spliced into the hosts own cellular DNA.
There it remains permanently as a provirus. RNA
molecules transcribed from the provirus serve as
mRNA for virus-encoded proteins and also as the
genomes for new virus particles.
Double-stranded RNA is unusual in nature but
constitutes the genome of the reoviruses.
516
The RNA
of these viruses fragments into segments upon infec-
tion. One member of the group is thought to be the
cause of acute diarrhea of infants.
517
4. Viruses without Nucleic Acid?
The cause of the slow, fatal neurological disease of
sheep known as scrapie has been a mystery for many
years. Similar human diseases include kuru and
Creutzfeldt-Jakob disease.
518 520
Scrapie can be trans-
mitted by injection and this has permitted isolation of
the apparent infective agent, a 27- to 30-kDa hydro-
phobic protein particle
521
which is devoid of DNA or
RNA. Prusiner
521
suggested the name prion (protein-
aceous infectious particle) for the scrapie agent. How-
ever, mRNA for the prion is present in normal as well
as infected brains, and protein produced in mouse
cells from cloned prion genes did not cause scrapie
infections. Therefore, there was doubt about the caus-
ative agent for the disease. The prion concept is now
generally accepted and is considered further in Chapter
29. There are still some who are looking for a nucleic
acid component.
519,522 524
5. Life Cycles
Viruses have many modes of life. They enter cells
in various ways. Some enter through coated pits from
which they are taken into lysosomes via endocytosis.
Others are literally injected into the cells (See Box 7-C).
Within cells some viruses are assembled in the nucleus,
some in the cytoplasm, and some in membranes. The
typical life cycle of a virus leads to rapid formation of
large numbers of progeny. Within 20 minutes after
entrance into a bacterial cell, a bacteriophage can
induce the formation of 100 200 new bacteriophage
particles. One of the bacteriophage genes encodes a
protein that is also synthesized by the host and which
induces lysis of the cell membrane and destruction of
the cell. Many animal viruses destroy cells in a similar
fashion.
Temperate bacteriophage, the best known being
phage , have a very different life cycle. Their DNA
usually becomes integrated at a specific point into the
genome of the bacterium (Chapter 27). Only rarely is
an infected cell lysed. The retroviruses that attack
mammals and birds have a similar characteristic.
Their DNA is also integrated into the host genome.
Some viruses that usually produce lysis of cells, e.g.,
SV40, adenoviruses, herpes viruses, and hepatitis B
virus, can occasionally be integrated into the DNA of
the host. If such integration occurs in the middle of a
gene, that gene will be mutated. This is one way in
which such viruses may induce cancers.
One of the most important results of integration of
viral DNA into the host genome is that the integrated
genes are replicated as part of the genome and are
transmitted from one generation to the next. Among
these are the cancer-causing viral oncogenes (v-onc),
which are discussed in Box 11-D and in Chapter 11,
Section H. While viruses are important causes of
cancer in some animals, relatively few human cancers
are thought to result directly from the action of viruses.
However, the EpsteinBarr virus, which causes mono-
nucleosis, can sometimes be integrated into epithelial
cells of nasal regions and can evidently cause cancer. The
same virus appears to be responsible for Burkitts lym-
phoma, a common cancer in certain areas in Africa.
525
6. Plasmids and Transposable Genetic
Elements
In addition to their chromosomal DNA, bacteria
often carry extra small pieces of DNA as permanent
parts of their genome. These plasmids (sometimes
called episomes), which are about the size of the DNA
of viruses, replicate independently of the host chromo-
somes. Each bacterial cell usually contains more than
a single copy of the plasmid. For example, the colici-
nogenic plasmid ColE1, that infects E. coli is a circular
piece of DNA of molecular mass 4.2 x 10
6
Da. Over 20
copies are normally found per cell but in the presence
of a suitable concentration of the drug chloramphenicol
the number may rise to 10002000.
Plasmids carry a variety of genes which are often
useful to bacteria. Some proteins encoded by plasmid
genes confer drug resistance to a bacterium. Some are
antibiotics. For example, a protein encoded by a gene
in plasmid ColE1 is toxic to other strains of E. coli.
Some plasmids carry genes for enzymes needed for
the oxidation of hydrocarbons. Some plasmids contain
249
genes for the restriction endonucleases which have
become essential to present-day molecular biology and
genetic engineering (Section H, 2).
As with some viruses, the DNA of many plasmids
can become integrated into the genome of the host.
An example is provided by the large 62-kDa plasmids
known as sex factors. They contain genes encoding
the protein subunits of the sex pili (Chapter 7) and
can become integrated into the bacterial chromosome.
Bacteria containing integrated sex factors are male
and are able to transfer genes not only of the sex factor
but also of virtually the entire bacterial genome into
other susceptible bacterial cells. This provides bacteria
with the means for sexual reproduction. The transfer
of DNA between the bacteria may occur via the sex
pili (see Chapter 26). In this respect the sex factors are
similar to viruses such as M13 that also appear to gain
entrance to bacteria via sex pili.
72
Integrated viruses are also related to transposable
genetic elements (transposons). These are segments
of DNA that allow genes to move from place to place
within the chromosomes (Chapter 27).
H. Methods of Study
Many of the methods discussed in Chapter 3 are
directly applicable to nucleic acids. A few additional
methods will be considered in this section.
525a
1. Isolation and Separation of Nucleic Acids
RNA is often extracted from lysed cells or tissues,
separated ribosomes, mitochondria, plastids, or nuclei
by warming with aqueous phenol and a detergent
such as sodium dodecyl sulfate (SDS). Proteins are
denatured by this treatment and are dissolved by the
phenol, while RNA remains in the lighter aqueous
layer. Depending on the conditions DNA may either
remain in the aqueous layer or be removed.
526 528
Various precipitation and extraction procedures may
be used to separate the RNA in the aqueous layer from
polysaccharides, from DNA (if present), and from their
components.
526,528,529
DNA may be extracted from cells
or nuclei as a nucleic acidprotein complex using 1 M
NaCl. The protein can then be denatured with an
organic solvent, by detergents, or by phenol. It is
desirable to digest proteins away with a nonspecific
protein-hydrolyzing enzyme such as proteinase K.
528
After removal of proteins DNA is often precipitated
with cold ethanol.
During isolation of RNA, bentonite (a type of clay)
or other inhibitors of ribonuclease are often added.
For the same reason, chelating agents that complex
metal ions needed for the action of deoxyribonucleases
are used to protect DNA. Care is necessary to avoid
shearing of the very long, narrow strands of DNA.
Even rapid pipetting of solutions will cause such
breakage.
Extracted RNA molecules may be separated from
each other by centrifugation in a sucrose gradient
(Chapter 3).
528
Fragments of DNA are purified in the
same way or by equilibrium centrifugation in CsCl
gradients.
37
Concentration gradients in the dense salt
solution are stable, and the sharpness of banding of
particles is ensured by use of a high centrifugal field.
Single-stranded DNA may be separated from double-
stranded DNA, and DNAs of differing G + C content
can be separated. The latter separation is based on
differences in buoyant densities in CsCl which are
approximately shown in Eq. 5-9.
= 1.660 + 0.098 (mole fraction C + G)
One of the most important methods for separating
either RNA or DNA mixtures is zone electrophoresis
through polyacrylamide or agarose gels. The separat-
ed bands may be visualized by scanning in ultraviolet
light or by fluorescence of intercalated dyes such as
ethidium bromide (Figs. 5-20, 5-22). This method is
being displaced to some extent by HPLC using DEAE
type ion exchange columns
530,531
for small lengths of
DNA including plasmids. The procedure called pulsed
field electrophoresis makes it possible to isolate very
large pieces of DNA, up to several million base pairs
in length.
532 534
The separation is carried out in agarose
gels, through which the long DNA rods must move in
a snakelike fashion.
534
The current is delivered in a
pulse and then, after a period of a second to several
minutes, a second pulse in a different direction, usually
at 90 to the first. The procedure is repeated many
times. The size of the DNA seems to affect the time
required to reorient the molecules and to start moving
in the second direction. Intact DNA from small chro-
mosomes can be separated (Fig. 5-42). To prevent
breakage of the DNA by shearing, intact cells are
suspended in liquid agarose and allowed to gel into a
block about 2 x 5 x 10 mm in size. The block is treated
with enzymes and detergents to lyse the cells and to
remove all protein and RNA.
534
The block, containing
the residual DNA molecules, is then embedded in the
electrophoresis gel. Other methods of DNA separation
include chromatography on hydroxyl-apatite and gel
filtration.
2. Hydrolysis and Analysis
Both DNA and RNA are easily broken down by
acid-catalyzed hydrolysis. Thus, heating at 100C for
one hour in 12 M HClO
4
is sufficient to hydrolyze
nucleic acids to their constituent bases. However, for
analysis of RNA it is better to heat in 1 N HCl for 1 h at
H. Methods of Study
(5-9)
250 Chapter 5. The Nucleic Acids
100C. The products are adenine, guanine, cytidine-5'-
phosphate, and uridine-5'-phosphate.
535
As is suggested
by this distribution of products, the glycosylamine
linkages to purines are more labile than those to pyri-
midines. The linkages are also less stable in DNA than
in RNA. A procedure based on these differences and
useful in sequencing by the Maxam-Gilbert method, is
to leave DNA overnight in the cold at pH 2 to cleave
off all of the purine bases. The resulting polymer is
known as an apurinic acid.
In alkaline solutions RNA is hydrolyzed to a mix-
ture of 2'- and 3'-nucleotides.
The mechanism involves participation of the free
2'-OH of the ribose groups and formation of cyclic 2',
3'-phosphates and is similar to that of pancreatic ribo-
nuclease (Chapter 12). Because deoxyribose lacks the
free 2'-OH, the phosphodiester linkages in DNA are
quite stable in base.
Hydrolytic cleavages of nucleic acids by the en-
zymes known as nucleases are of great practical value.
Pancreatic ribonuclease, an endonuclease, cuts a chain
adjacent to a pyrimidine in nearly random fashion,
leaving phospho groups attached to the 3' position in
the nucleotide products (Fig. 5-43). Exonucleases
cleave from the ends of chains. For example, the phos-
phodiesterase of snake venom cleaves from the 3' end,
which must have a free 3'-OH group, to give 5'-nucleo-
tides. On the other hand, the phosphodiesterase from
spleen has the opposite polarity, cleaving chains from
the 5' end to give 3'-nucleotides. Similar variations in
specificity are found among enzymes that cleave DNA.
For example, pancreatic DNAase I, which cleaves
preferentially between adjacent purines and pyrimidines,
yields 5' mononucleotides whereas DNAase II gives
3'-mononucleotides. Various hydrolytic cleavage reac-
tions of polynucleotides are summarized in Fig. 5-43.
The most striking specificity in DNA hydrolysis is
displayed by the restriction endonucleases which
are discussed further in Chapter 26. These fussy cata-
lysts cleave only at points within or close to a defined
sequence of several nucleotides in double-stranded
DNA. For example, the enzyme EcoR I cuts only at
the following palindromic sequence:
The cuts in the two strands are made at the points
indicated by the arrows. This one endonuclease will
cut almost any DNA into long pieces averaging about
5000 base pairs each. These pieces can in turn be
cleaved by other restriction endonucleases to form
smaller fragments. Since there are about 2400 of these
enzymes known, with 188 different specificities,
536
it
is possible to cut any piece of DNA down to a size of
100500 base pairs, ideal for sequencing.
537 539
Each
fragment has known sequences at the two ends. Some
restriction enzymes cleave outside their specific recog-
nition sequence (see Table 26-2). Some recognize 16-
nucleotide palindromes and cut at rare sites.
It is sometimes desirable to cut a large DNA mole-
cule at only a few points. One approach is to protect
most sites of a restriction enzymes action by methyl-
ating them (see Chapter 26) while protecting the desired
cleavage site, for example by a repressor protein
540
or by
a PNA molecule (p. 227) of specific sequence designed to
clamp the site chosen for protection.
541
Ribozymes
(Chapter 12) have been engineered to be as specific or
more specific than endonucleases.
542,543
Other new
approaches are being developed.
544
The base composition of either RNA or DNA can
be determined after hydrolysis catalyzed by 98% formic
acid at 175C for 30 min or by 12 M perchloric acid at
100C for 1 h.
545
The bases can then be separated by
ion exchange chromatography on a sulfonated poly-
styrene resin. RNA can be hydrolyzed to a mixture of
nucleoside 2- and 3-phosphates by 0.3 M NaOH at
O
O OH
H
2
C Base
O
3
P
O
HO O
H
2
C Base HO HO
PO
3
+
2' 3'

Figure 5-42 Intact DNA from the chromosomes of three
strains of the malaria parasite Plasmodium falciparum, ranging
from 750 Kb to 5 Mb, separated by pulsed-field gel electro-
phoresis. Courtesy of C. Smith and T. E. Wellems. Repro-
duced by permission of Amersham Pharmacia Biotech Inc.
5 G A A T T C 3
3 C T T A A G 5

251
37C for 16 h and DNA can be hydrolyzed to nucleotides
enzymatically. The negatively charged nucleotides can
then be separated by ion exchange chromatography on
a quaternary base-type resin (Chapter 3). Periodate
cleavage (Eq. 4-12) and reduction of the resulting dialde-
hydes by [
3
H]NaBH
4
to trialcohols allows introduction of
a radioactive label (Fig. 5-44). Alternatively, the dialde-
hydes can be reductively alkylated by an amine plus
NaCNBH
3
.
546
The total content of RNA + DNA in tissues may
be estimated from the phosphorus content or by color
reactions of the sugars.
37,545
These reactions depend
upon dehydration to furfural or deoxyfurfural by
concentrated sulfuric acid or HCl (Eq. 4-4). Furfural
formed from RNA reacts with orcinol (3,5-dihydroxy-
toluene) and ferric chloride to produce a green color
useful in colorimetric estimation of RNA. A similar
reaction of DNA with diphenylamine yields a blue color.
Quantitative determination of over 90 free nucleotide
compounds found within cells can be accomplished
by thin layer chromatographic procedures on as few
as 10
6
bacterial cells (~ 2 g) labeled by growth in a
32
P
i
-containing medium.
547
3. Characteristic Reactions of the Bases and
Backbone
Reactions of nucleophiles. A number of nucleo-
philic reagents add reversibly at the 6 position of pyrim-
idines. Thus, bisulfite adds to uridine (Eq. 5-10).
528
Hydroxylamine (HONH
2
) adds in a similar fashion to
give a compound with HNOH in the 6 positions.
528
Sodium borohydride (NaBH
4
), which can be viewed
as a donor of a hydride ion (H

), reduces uridine to
the 5,6-dihydro derivative. This presumably occurs
by attack of the hydride ion at position 6 in a manner
analogous to the reaction of bisulfite in Eq. 5-10.
Cytidine reacts in the same way, but the bisulfite
addition compound is unstable. These C5C6 adducts
of cytidine all have a greatly enhanced reactivity at C4,
O
B
O HO
3
P
O
O
H
P
O
O
O
Pu
O
O
O
H
P
O
O
O
Py
O
O
O
H
P
O
O
O
O
O
O
H
P
O
O
O

3'-End 5'-End
The free 2' OH in RNA and other ribonucleotides can
participate in b-type cleavage by nucleophilic attack
on the phosphorus (see Fig 12-25 )
Cleavage here by weak acids (pH 2) yields an apurinic acid
a
b
B
Pancreatic ribonuclease b cleavage
is to the right of pyrimidines
Figure 5-43 Some hydrolytic cleavage reactions of polynucleotides. Reactions of both RNA and DNA are included.
A. Cleavage at point a is catalyzed
1. Throughout the molecule by endonucleases
Pancreatic deoxyribonuclease I
2. Only at the 3' end by exonucleases
Venom diesterase, nonspecific, attacks DNA and RNA. A free 3'-OH is essential
B. Cleavage at point b is catalyzed
1. Randomly throughout the molecule by endonucleases and by bases (nonenzymatically)
Pancreatic ribonuclease cleaves only to the right of a pyrimidine-containing nucleotide
Ribonuclease T1 of Aspergillus oryzae cleaves to the right of a guanine-containing residue (3'-guanylate) in ssRNA
Ribonuclease T2 of Aspergillus oryzae cleaves to the right of an adenine-containing residue (3'-adenylate) in ssRNA
Pancreatic deoxyribonuclease (DNase) II
Micrococcal DNase
2. Only at the 5' end by exonucleases
Bovine spleen phosphodiesterase hydrolyzes both polyribo- and polydeoxyribonucleotides
H. Methods of Study
N
H N
O
O
Ribose
H
SO
3
H
H
Uridine + HSO
3
5
6

(5-10)
252 Chapter 5. The Nucleic Acids
N
N O
R
HN
N O
N
R
H N
OH (OCH
3
) OH (OCH
3
)
(5-12)
presumably because of the lessened aromatic character
of the ring. Cytidine is slowly deaminated by base,
presumably as a result of attack by hydroxyl ion on the
electrophilic center at C4 and subsequent elimination
of NH
3
(Eq. 5-11). The reaction is catalyzed by buffer
salts and by bisulfite and hydroxylamine. Catalysis
probably occurs, at least in part, as a result of addition
of these nucleophiles at the 6 position to form com-
pounds with increased nucleophilic reactivity at C4.
548
Hydroxylamine and methoxyamine (NH
2
OCH
3
) par-
ticipate in reactions parallel to that of the hydroxyl ion
in Eq. 5-11. Products contain NHOH or NHOCH
3
in place of NH
2
but tautomerize to the more stable
forms shown in Eq. 5-12. Similar substitution reactions
occur with other amines.
33
The C6 adduct with hydra-
zine can undergo ring cleavage (Eq. 5-13). The initial
product then undergoes elimination, leaving ribo-
sylurea or deoxyribosylurea. The same reaction can
be carried out on intact strands of DNA and is widely
used in determination of nucleotide sequences.
The conversion of 5-hydroxymethylcytosine to the
Figure 5-44 Fluorographs of 23'-[
3
H] nucleoside trialcohols from Bacillus subtilis grown in the absence (A) and presence (B)
of 5-fluorouracil. About 2.3 nmol of nucleosides from each sample was chromatographed and exposed to X-ray film for 90 h.
at 80 C. Or (origin) B
1
, B
2
, and B
3
contain unidentified material present in a reaction mixture lacking RNA. Abbreviations
used: FU, 5-fluorouridine; FC, 5-fluorocytidine; U, uridine; C, cytidine; G, guanosine; A, adenosine; I, inosine; mG, 1-meth-
ylguanosine; m
7
G, 7-methylguanosine; mA, 1-methyladenosine; m
6
A, 6-methyladenosine; m
6
6
A, 6,6-dimethyladenosine; t
6
A,
N-[9-(-D-ribofuranosyl) purin-6-yl carbamoyl] threonine; H
5
6
U, 5,6-dihydrouridine; , pseudouridine;
D
, decomposition
product of ; m
5
U, 5-methyluridine (ribosylthymine); mo
5
U, 5-methoxyuridine; N, a nucleoside trialcohol obtained by reduc-
tion of a nucleoside dialdehyde with [
3
H]NaBH
4
; FU-5 and FU-20 samples correspond to tRNAs from cells grown at that final
concentration of 5-fluorouracil in g / ml. Courtesy of Ivan Kaiser.
N
N O
NH
2
R
H N
N O
R
H N
N O
O
R
NH
3
H
2
N
H
OH
H
OH
Adduct
+

+
(5-11)
A B
253 H. Methods of Study
5-methylenesulfonate (5-CH
2
SO
3

) by reaction with
bisulfite should also be mentioned.
549
This is a nucleo-
philic displacement on the electron-deficient methylene
group of this base.
Reactions with electrophilic reagents. Reactions
of nucleic acids with the simplest electrophile, the proton,
have been considered in Section A2. Somewhat similar
are the reactions by which metal ions bind at many sites
on both the bases and the phosphate groups of the back-
bone.
550
An important reaction is the deamination of amines
by dilute nitrous acid. This reagent, by a complex
mechanism, converts the amino groups of cytidine,
adenosine, and guanosine to hydroxyl groups; hydroxy
compounds tautomerize to the corresponding amides
(Eq. 5-14).
Cytidine reacts more rapidly than does adenosine
which in turn reacts more rapidly than guanosine. The
reaction converts cytosine into uracil and adenine into
hypoxanthine. The changes are mutagenic because
during replication the modified bases of the DNA pair
differently than do the original bases. Guanine is con-
verted to xanthine but this is not likely to be highly
mutagenic. Nitrous acid can also convert uridine to
5-nitrouridine.
The amino groups of the bases react reversibly
with aldehydes but to a lesser extent than do the more
strongly basic amino groups of the amino acids. Form-
aldehyde forms adducts containing either one or two
molecules of the aldehyde (Eq. 5-15).
N
H N
O
O
R
H
H
NH
H
NH
2
H
N
H
NH
O
N O
H
2
N
R
H
H
H
N
N
O
H
O NH
H
2
N
R
elimination
Ribosylurea
H
+
(5-13)
These are reversible reactions. A more nearly irrevers-
ible crosslinking can occur by elimination of water
between one of these products and a nucleophilic
group in another base. A dicarbonyl reagent that is
widely used because of its specificity toward guanine
is kethoxal (Eq. 5-16).
Formation of the cyclic product is a consequence of the
presence of the adjacent amino and NH groups in the
guanine ring.
Pyrimidines undergo halogenation at position 5
(Eq. 5-17), while guanine reacts at position 8. Adenine
is quite unreactive. Elemental halogens or a variety of
other halogenating reagents may be used. Of special
Br
2
+ cytidine 5-Br-cytidine + H
+
+ Br

value is iodination with


131
I or
125
I, by which a high
level of radioactivity may be introduced into nucleic
acids.
Alkylation reactions are not only of use in struc-
tural studies but also provide the basis for the action
of a large class of mutagenic compounds.
528
Treatment
of a nucleoside, nucleotide, or nucleic acid with an
alkyl iodide or a dialkylsulfate converts residues of
guanosine to an N
7
-alkyl-guanosine (Eq. 5-18).
N
NH
2
HN
O
HNO
2
(5-14)
NH
2
N
CH
2
OH
HOH
2
C
N
CH
2
OH
H
C
H
O
H
2
CO
H
+
(5-15)
N
N
N
N C
C
N
O
R
C
HO
H
3
C
HN
N
N
N
H
2
N
O
R
C
C
C
H
H
3
C
OC
2
H
5
H
high pH pH 7
= O
O
H
= O
+
C
2
H
5
H
H HO
(5-16)
(5-17)
254 Chapter 5. The Nucleic Acids
Reaction occurs at other nitrogen atoms as well as the
oxygen atom of the base and of the ribose ring to a
lesser extent. Adenine is alkylated preferentially at
N-1 and cytosine and thymine at the corresponding
position (N-3) almost exclusively. Uridine and thymi-
dine react very slowly. Adenine is also alkylated at
N-3, N-7 and at the exocyclic N-6.
Other alkylating reagents include the powerful
mutagens dialkylnitrosamines and alkylnitrosoureas.
Epoxides alkylate by a nucleophilic displacement
reaction that opens the ring (Eq. 5-19).
The nitrogen and sulfur mustards undergo internal
ring closure to an iminium ion (Eq. 5-20) which can
then open by attack of a nucleophilic atom of the
nucleic acid.
Other alkylating agents react through nucleophilic
addition to a carboncarbon double bond. Thus,
acrylonitrile reacts with the nitrogen or oxygen atoms
of nucleic acids in the same manner as does the SH
group in Eq. 3-25. The water-soluble carbodiimides
react as in the first step of Eq. 3-10 to form adducts of
the following type:
There are many other alkylating agents which often
display widely varying reactivity and specificity toward
particular nucleic acid bases and particular nucleotide
sequences.
A striking effect of alkylation of guanine in nucleic
acids is the labilization of the N-glycosyl linkage to the
ribose or deoxyribose. This effect can be understood
in terms of the induction by resonance of a partial
positive charge on the nitrogen of the glycosyl linkage.
As is indicated by the small arrows on the right-hand
structure, the positive charge assists in an elimination
reaction that produces an oxycarbocation. The latter
can then react with a hydroxyl ion from water.
Reactions causing cleavage of the sugar-
phosphate backbone. Treatment of DNA with 16 18
M hydrazine (Eq. 5-13) leads to the destruction of the
pyrimidine rings. The reaction can be made some-
what specific for cytosine by carrying it out in the
presence of a high concentration of chloride.
551
The
remaining polymer, an apyrimidinic acid, contains
residues of ribosylurea. These undergo an amino-
catalyzed displacement and a elimination sequence
that cleaves the polynucleotide chain (Eq. 5-21).
Hydration of the aldehyde (Eq. 13-1) and several
HN
N
N
N
H
2
N
R
O
Guanosine
I
Ribose
+
+ R I

7
(5-18)
O = N N
R
R
Dialkylnitrosamines
R C C R'
O
H
R C C Y
HO R'
H
Y
H H H

+
(5-19)
R
CH
2
CH
2
N
R
CH
2
CH
2
Cl
N CH
2
CH
2
Cl CH
2
CH
2
Cl
Cl
Y
+
A nitrogen mustard gas

(5-20)
N
N
N
H
N O
O
R
H R'
N
NH
N
N
CH
3
O
NH
2
N
NH
N
N
CH
3
O
NH
2
O
OH
CH
2
+
HO
+
O
HO
CH
2
HO
255
tautomerization steps are involved in step e of this
equation. This reaction is very useful in sequence
determination (Section 6). Notice that tracts of
purine nucleotides remain intact after this treatment.
A similar base-catalyzed reaction sequence can be
used to displace N
7
-methylguanine and to cleave the
polynucleotide. Ethylnitrosourea, in its reaction with
purines, is useful as a structural probe of RNA.
4. Melting, Hybridization, and Polynucleotide
Probes
Like proteins, nucleic acids can undergo denatur-
ation. The strands of the double helix of DNA are
separated and the double-stranded regions of RNA
molecules melt. Denaturation can be accomplished
by addition of acids, bases, and alcohols or by removal
of stabilizing counter ions such as Mg
2+
. The product
is a random coil and denaturation can be described as
a helix coil transition. Denaturation of nucleic acids
by heat, like that of proteins, is cooperative (Chapter 7,
Section A,3) and can be described by a characteristic
melting temperature.
A plot of the optical absorbance at 260 nm (the
wavelength of maximum light absorption by nucleic
acids) versus temperature is known as a melting
curve (Fig. 5-45). The absorbance is lower, by up to
40%, for native than for denatured nucleic acids. This
hypochromic effect (Chapter 23) is a result of the
interaction between the closely stacked bases in the
helices of the native molecules. The melting tempera-
ture T
m
is taken as the midpoint of the increase in
absorbance (Fig. 5-45). As the percentage of G + C
increases, the nucleic acid becomes more stable toward
denaturation because of the three hydrogen bonds in
each GC pair. T
m
increases almost linearly with in-
creases in the G + C content. In the standard citrate
buffer (0.15 M NaCl + 0.015 M sodium citrate, pH 7.0)
Eq. 5-22 holds. The exact numerical relationship depends
strongly upon the ionic composition and pH of the
medium.
37,72,552,553
% (G + C) = 2.44 (T
m
69.3); T
m
in C
The curves in Fig. 5-45 appear simple, but using newer
apparatus and plotting the first derivative of the melt-
ing curve yields a complex pattern that depends on
the sequence of bases.
555
Complete denaturation of DNA leads to separation
of the two complementary strands. If a solution of
denatured DNA is cooled quickly, the denatured strands
remain separated. However, if the temperature is
held for some time just below T
m
(a process known as
annealing), the native double-stranded structure can
be reformed. An important tool for studying DNA
has been the measurement of the kinetics of reasso-
ciation of separated strands of relatively short DNA
fragments.
72,556,557
Because it depends upon the concentration of two
separated strands, reassociation obeys second-order
kinetics (Chapter 9) and Eq. 5-23, which is readily
derived by integrating Eq. 9-8 for [A] = [B] = C from
time 0 to t:
C / C
0
= 1/ (1 + k C
0
t)
H. Methods of Study
O
O H
PO
2
R'
CH
2
N O
PO
2
R
H C
NH
2
O
N
O
O H
PO
2
R'
CH
2
O
PO
2
R
H
N
H
O
PO
2
R'
CH
2
O
PO
2
R
N C
H O
H
H
H
CH
2
O
PO
2
R
N C
H OH
R' P O
O
OH

Piperidine, often used


as the displacing base

H
O C
H CH
2

R P O
O
OH

Schiff base
+

+
+ HO HN
C
O
CH
3
+
+
O
OH

a
b
c
d
e
Two
steps
H
2
O
Hydration,
tautomerization
(5-21)
(5-22)
(5-23)
256 Chapter 5. The Nucleic Acids
The initial concentration of denatured DNA, C
0
, is
related in this way to the concentration C of DNA
remaining dissociated at time t. A plot of the fraction
of molecules remaining single-stranded versus the
logarithm of C
0
t (Fig. 5-46A) is a convenient way of
displaying data. As indicated in Fig. 5-46B, the value
of C
0
t increases in direct proportion to the length of
the DNA chain in the genome, but it is very much
decreased if the sequence of bases is highly repetitive
[poly(T) and poly(A)]. The slope of the plot at the
midpoint gives an indication of the heterogeneity of
the DNA fragments in a solution.
Denatured DNA fragments can sometimes reasso-
ciate with DNA from a different source to form hybrid
duplexes. Such double helices, in which one strand
comes from one strain of an organism and the other
strand from a genetic variant of the same organism or
from a different species, are known as heteroduplex.
Some mutations consist of deletions or additions of
one or a substantial number of bases to a DNA chain.
Heteroduplexes prepared from DNA of such mutants
hybridized with that from a nonmutant strain have
normal hydrogen-bonded WatsonCrick base pairs for
the most part. However, they may have single-stranded
loops in regions where long deletions or additions
prevent complementary base pairing.
Hybridization measurements have been used in
many studies of homology of nucleic acids from differ-
ent species. A nucleic acid is cut (e.g., by sonic oscilla-
tion) into pieces of moderate length (~ 1000 nucleotides)
and is denatured. The denatured DNA fragments are
mixed with denatured DNA of another species. Nu-
cleotide sequences that are closely similar between
species tend to hybridize, whereas sequences that are
drastically different between two species do not (Fig.
5-46). One way to do such an experiment is to immo-
bilize the long-chain denatured DNA from the one
organism by embedding it in an agar gel
558
or by
absorbing it onto a nitrocellulose filter.
559,560
The DNA
fragments from the second organism are passed through
a column containing beads of the DNA-containing
agar or through the filter with adsorbed DNA. Pairing
of fragments with complementary sequences occurs
Figure 5-46 Reassociation curves for DNA from Britten
and Kohne.
556,561
(A) Time course of an ideal, second-order
reaction to illustrate the features of the log C
0
t plot. The
equation represents the fraction of DNA which remains
single-stranded at any time after the initiation of the reaction.
For this example, k is taken to be 1.0, and the fraction remain-
ing single-stranded is plotted against the product of total con-
centration and time on a logarithmic scale. (B) Reassociation
of double-stranded nucleic acids from various sources. The
genome size is indicated by the arrows near the upper nomo-
graphic scale. Over a factor of 10
9
, this value is proportional
to the C
0
t required for half-reaction. The DNA was sheared,
and the other nucleic acids are reported to have approximately
the same fragment size (about 400 nucleotides, single-stranded).
Correction has been made to give the rate that would be
observed at 0.18 M sodium-ion concentration. The tempera-
ture in each case was optimal, i.e., ~ 30C below the melting
temperature T
m
. The extent of reassociation was established
by measuring optical rotation (calf thymus DNA), ribonu-
clease resistance (MS-2), or hypochromicity.
Initial state
Half-reaction
Final state
Concentration time (C
0
t)
F
r
a
c
t
i
o
n

r
e
m
a
i
n
i
n
g
s
i
n
g
l
e
-
s
t
r
a
n
d
e
d
1.0
0.5
0
0.01 0.1 1 10 100 0.001
C
C
0
=
1
1 + kC
0
t
Nucleotide pairs
C
0
t (mol s/l)
10
6
10
4
10
2
0.1 1 10 100 10,000
1.0
0.5
0.0
F
r
a
c
t
i
o
n

r
e
m
a
i
n
i
n
g
s
i
n
g
l
e
-
s
t
r
a
n
d
e
d
1 10 10
2
10
3
10
4
10
5
10
6
10
7
10
8
10
9
10
10
Poly(U) +
poly(A)
Mouse
satellite MS-2
E. coli
Calf
(nonrepetitive
fraction)
T4
Temperature (C)
74 76 78 80 82 84 86 88 90 92 94 96 98 100
1.00
1.12
1.08
1.04
1.48
1.44
1.40
1.36
1.32
1.28
1.24
1.20
1.16
R
e
l
a
t
i
v
e

a
b
s
o
r
b
a
n
c
e

a
t

2
6
0

n
m
Serratia
Pneumococcus
Figure 5-45 A melting curve for DNA molecules from two
different sources. From Davidson.
554
A
B
257
and such paired fragments are retained while strands
that do not pair pass on through the column (or filter).
Both DNA hybrids and DNARNA hybrid du-
plexes are very important to present day genetic
research.
560,562
Molecules of mRNA that represent
transcripts of a particular gene will hybridize only
with one of the two separated strands of DNA for that
gene.
A major use of hybridization is to locate a gene or
other DNA or RNA sequence by means of a synthetic
probe.
563
This is a small piece of DNA or RNA which
is labeled in some way, e.g., with a radioisotope such
as
3
H,
32
P, or
125
I. Alternatively, the probe may carry a
highly fluorescent dye or a tag that can be recognized
by a specific antibody.
564,565
An example of the latter is
the use of the vitamin biotin and the specific binding
protein avidin (see Box 14-B).
566
Related procedures
employ labeling with digoxigenin and often employ
chemiluminescent detection.
567 569
Several methods
for preparation of probes are in use. Some are enzy-
matic but the direct chemical synthesis of oligonucleo-
tide probes is probably used the most.
One of the first methods devised for making a
highly radioactive DNA probe is called nick transla-
tion.
570
A piece of dsDNA, e.g., a restriction frag-
ment cut from a larger piece of DNA by restriction
endonucleases, is selected. A small amount of pancre-
atic DNase I is added. It creates nicks in which one
strand has been cut and some nucleotides have been
removed leaving a gap. Now the DNA is incubated
with DNA polymerase I (pol I) and a mixture of the
four mononucleotide triphosphates, the precursors of
biological synthesis of DNA (Chapter 27). Usually a
32
P or
35
S label with high radioactivity is present in
one of the nucleotide triphosphates as indicated in
the following structure. The polymerase fills the gap,
adding nucleotides to the exposed 3' end of the nicked
chain. E. coli pol I has a second enzymatic activity
which allows it to digest a polynucleotide chain from
Probes may also consist of DNA copied from
mRNA. This is known as cDNA and is also widely
used to determine indirectly the sequences of mRNA
molecules. Messenger RNA may be isolated from the
total cellular RNA by affinity chromatography on
bound poly (dT) or poly (U). These materials selec-
tively hold RNA with the poly (A) tails characteristic
of most eukaryotic mRNA (see Chapter 28). Another
source of mRNA is polyribosomes (polysomes), which
are reading mRNA and actively making proteins.
An antibody to the protein for which mRNA is desired
will often bind to the protein chains being synthesized
and precipitate the polysomes. The mRNA can be
recovered and used as a template for cDNA.
Synthesis of cDNA, usually in radiolabeled form is
accomplished with reverse transcriptase, the enzyme
from retroviruses that synthesize a DNARNA hybrid
from ssRNA.
570 572
A short oligo (dT) primer is usually
hybridized to the 3' poly (A) tail to initiate synthesis.
Reverse transcriptase also has ribonuclease (RNase H)
activity and will digest away the RNA. If desired,
synthesis of the second strand can be carried out by
a DNA polymerase to give a complete DNA duplex.
Many gene sequences have been deduced from cDNA
copies.
Often the most practical approach to obtaining a
DNA probe is synthesis of a mixture of short oligonu-
cleotides, often in radioactive form as described in the
next section. The redundancy in the genetic code,
i.e., the existence of two or more codons for most
amino acids, presents a problem in designing an oligo-
nucleotide probe based on amino acid sequence infor-
mation. Examination of Table 5-5 suggests part of the
solution. Whereas only Met and Trp have single un-
ambiguous codons nine amino acids have only two
codons each. We should try to find an amino sequence
that contains Met and Trp and as many of the nine
others as possible. We should avoid sequences that
contain Ser, Leu, or Arg because each has six codons.
We can then make a mixture of oligonucleotides, using
H. Methods of Study
P O O
HO
O
P
O
O
P
O O
O CH
2
O
H H
HO H
O

Thymine
Cytosine
Guanine or
Adenine


35
S
32
P
The four nucleotide triphosphate precursors of DNA

Nick
5 3
3 5
5 3
3 5
5 3
3 5
5
DNA polymerase I
Denaturation
Radiolabeled probe
5
(5-24)
the 5' end. Thus, as synthesis proceeds at the 3' side
the nick is translated as shown in (Eq. 5-24).
258 Chapter 5. The Nucleic Acids
a
b
c
d
e
O CH
2
O
Base 1
OH
DMT N
C
CH
2
CH
2
COOH
O
CH
2
CH
2
CH
2 Si O
O
O
Si
Si
Si
H
O CH
2
O
Base 1
O
DMT
C
O
Silica
O CH
2
O
Base 2
O
DMT
P N
C
3
H
7
C
3
H
7
O
CH
2
CH
2
NC
O CH
2
O
Base 1
O
H
C
O
Silica
O CH
2
O
Base 1
O
DMT
P
O
O CH
2
CH
2
NC
CH
2
O
Base 1
O
C
O
Silica
O CH
2
O
Base 2
O
DMT
P
O
O
CH
2
O
Base 1
O
C
O
Silica
Dicyclohexyl-
carbodiimide
Silica
support
Base 1
Second
monomer
Base 1
Base 2
I
2
, H
2
O
Conc. NH
3
Removal of all protecting
groups and release from silica
ZnBr
2
Butanol extraction
Addition of
successive
nucleotides
units
First
monomer
Base 2
Base 1
Base 2
Base 1
(5-25)
the various coding possibilities. Mixtures of as many
as 1024 (2
10
) oligonucleotides have been used. One of
these may bind tightly and specifically to the desired
DNA segment. Instead of such a complex mixture it
may be more useful to incorporate a modified base at
the most ambiguous positions. For example, inosine,
which occurs in the wobble position in anticodons
(Fig. 5-30), can pair with A, C, or T.
573
Substitution
of 2-aminoadenosine can cause a probe to bind more
tightly because a third hydrogen bond will be present
in each AT pair.
574
Another important procedure is labeling ends of
polynucleotides. Most often the 5' end is labeled with
a radioisotope or by covalent attachment of a fluores-
cent dye. For example, a polynucleotide kinase can
be used to transfer a radioactive -phospho group from
ATP to the 5' end of a polynucleotide that has a free
5'-OH group.
5. Synthesis of Oligonucleotides
and Polynucleotides
Efficient solid-phase methods of syn-
thesis analogous to those for polypeptides
(Fig. 3-15) have been devised. The pioneer-
ing work was done by H. G. Khorana, who
made the first synthetic gene
575
and later
synthesized a gene for the visual pigment
rhodopsin (Chapter 23). Several synthetic
approaches have been developed.
576
Cur-
rently the most popular method involves
the use of phosphite esters. Most nucleo-
philic groups of the monomers are deriva-
tized with removable blocking groups.
For example, N-4 of cytosine and N-6 of
adenosine may carry benzoyl groups. The
5' OH of each nucleotide is blocked by a
di-p-anisylphenylphenylmethyl (also called
dimethoxytrityl, DMTO) group. The 3'OH
is converted to one of a number of activated
derivatives such as the following N, N-
diisopropylamino phosphines.
575,577 579
Solid-phase synthesis is usually done
on a silica support with a covalently at-
tached succinamide as shown in Eq. 5-25.
The first nucleotide at the 3' end of the
chain to be synthesized is attached by an
ester linkage to the bound succinamide
(step a, Eq. 5-25). The 5'-protecting group
is removed in step b and the 5'-OH reacts
with the activated phosphine of the second
nucleotide (step c, Eq. 5-25). Steps b and c
are then repeated as often as necessary to
complete the chain. The finished polynu-
cleotide can be removed from the solid
support, the cyanoethyl groups removed
from the phosphorus atoms by elimination and all of
the other blocking groups removed by treatment with
concentrated NH
3
. The whole procedure has been
automated.
580 582
If a large piece of DNA is needed several oligonu-
cleotides can be joined end to end enzymatically (Eq.
O CH
2
O
H H
O
H
DMT
P
N O CH
2
CH
2
CN
C
C
H
3
C
H
3
C
H
3
C
H
3
C
Thyminyl
N-4-Benzoylcytosinyl
N-6-Benzyladeninyl
N-2-Isobutyrylguaninyl
H
H
Activated monomeric
nucleotides for synthesis
of polynucleotides
259 H. Methods of Study
BOX 5-D DNA FINGERPRINTING
Jeffreys et al.
a c
digested human DNA to comple-
tion with Hin f I and Sau3A restriction endonucleases.
Certain fragments, which originated from mini-
satellite bands of repetitive DNA showed a very
high degree of polymorphism among the population.
Many different fragments sharing these repeated
sequences were formed in the restriction digest. If a
suitable labeled probe was used, it hybridized with
as many as 80 different bands.
a,d f
The resulting
pattern appeared, like a fingerprint, to be different
for every individual, as is shown in the accompany-
ing photo. Unlike a fingerprint the DNA pattern
also contains information that often allows deduc-
tions about parentage.
The technique has come into widespread use in
forensic analysis, with DNA typing being possible
from a single hair.
g
In a famous early case it was
used to allow an immigrant child to be reunited
with his mother
h
and it is being used regularly to
protect innocent persons accused of rape or murder.
i
It is also widely used to provide evidence of guilt.
However, the very small chance of a close match
between unrelated persons prevents the use of DNA
typing alone as proof of guilt. Because DNA samples
are often amplified by PCR (Section H,6), there is
also a possibility of contamination and forensic use
of DNA typing is still controversial.
i m
However, DNA typing continues to be improved
f,n,o
and to be applied in a great variety of ways. For
example, the skeletal remains of a murder victim
were identified by DNA fingerprints after being
buried for eight years.
p
DNA typing is also used to
study mating habits of birds,
q
the genetic variability
of populations of whales sampled by biopsy,
r
etc.
DNA fingerprints made from one or two drops of blood
from ten different individuals. DNA was isolated, digested
to completion with restriction endonuclease Hinf I, and
subjected to electrophoresis in a 20-cm-long agarose gel
until all DNA fragments smaller than 1.5 kb in length had
passed off the gel. The DNA was then transferred to a
nitrocellulose filter by Southerns method and hybridized
with a
32
P-labeled single-stranded DNA probe prepared
from cloned human minisatellite DNA. The probe used
had the consensus composition (AGAGGTGGGCAG-
GTGG). Within the 29 tandem repeats in this 0.46-kb
probe there are various sequences close to the one shown.
Filters were then autoradiographed for four days. Two
duplicate samples (marked D) were taken from the same
individual and two others (marked S) from two sisters.
A number of bands in common are evident. From Jeffreys
et al.
c
a
Jeffreys, A. J., Wilson, V., and Thein, S. L. (1985) Nature (London)
314, 6773
b
Lewin, R. (1986) Science 233, 521522
c
Jeffreys, A. J., Wilson, V., and Thein, S. L. (1985) Nature (London)
316, 7679
d
Vassart, G., Georges, M., Monsieur, R., Brocas, H., Lequarre, A.
S., and Christophe, D. (1987) Science 235, 683684
e
Huang, L.-S., and Breslow, J. L. (1987) J. Biol. Chem. 262, 8952
8955
f
Kirby, L. T. (1990) DNA Fingerprinting, Stockton Press, New
York
g
Higuchi, R., von Beroldingen, C. H., Sensabaugh, G. F., and
Erlich, H. A. (1988) Nature (London) 332, 543546
h
Jeffreys, A. J., Brookfield, J. F. Y., and Semeonoff, R. (1985)
Nature (London) 317, 818 819
i
Balding, D. J., and Donnelly, P. (1994) Nature (London) 368,
285286
j
Neufeld, P. J., and Colman, N. (1990) Sci. Am. 262(May), 4653
k
Lewontin, R. C., and Hartl, D. L. (1991) Science 254, 17451750
l
Lander, E. S., and Budowle, B. (1994) Nature (London) 371,
735738
m
Lewontin, R. C. (1994) Nature (London) 372, 398
n
Uitterlinden, A. G., Slagboom, P. E., Knook, D. L., and Vijg, J.
(1989) Proc. Natl. Acad. Sci. U.S.A. 86, 27422746
o
Jeffreys, A. J., MacLeod, A., Tamaki, K., Neil, D. L., and
Monckton, D. G. (1991) Nature (London) 354, 204209
p
Hagelberg, E., Gray, I. C., and Jeffreys, A. J. (1991) Nature
(London) 352, 427 429
q
Burke, T., and Bruford, M. W. (1987) Nature (London) 327,
149152
r
Hoelzel, A. R., and Amos, W. (1988) Nature (London) 333, 305
260 Chapter 5. The Nucleic Acids
27-5). For example, a functional 17-bp gene for the 53-
residue human epidermal growth factor was synthe-
sized by joining ten oligonucleotides of lengths 11 59
bp.
575
DNA is often synthesized enzymatically using
methods described in Chapter 26. Cloned sequences
of synthetic DNA can also be transcribed to produce
polyribonucleotides of any desired sequence.
583
New nonenzymatic methods for RNA synthesis have
also been devised.
583 587
6. The Polymerase Chain Reaction (PCR)
This important technique was first described in
19711974 by Khorana and associates
588,589
but was not
used until it was rediscovered in 1983 by Mullis.
590 593
It was quickly developed
591,592,594 596
and has played a
major role in biochemistry ever since. It continues to
be applied in numerous ways.
589,597 599a
The PCR technique provides a way of amplifying
a small number of DNA molecules, i.e., to produce
many copies. This is often done by cloning but PCR
offers a quick and easy way to obtain millions of copies
of a desired relatively short segment of DNA. Standard
PCR can be used for up to about 5000-nucleotide
pieces. More recently modified procedures have
allowed 35-kb segments to be amplified.
600
The basic PCR procedure is initiated by hybridiz-
ing two oligonucleotide primers onto opposite strands
of denatured DNA, one at each end of the section chosen
for amplification (Fig. 5-47). A DNA polymerase is then
used to convert each of the separated strands into a
duplex. The mixture of products is then heated to
denature the two new duplexes. After cooling, the
primers, which are present in great excess, hybridize
to all four strands. In a second cycle of polymerase
action these are all converted to duplexes, etc. After 20
cycles millions of copies will be made. At first, copies
with tails extending beyond the limits specified by the
oligonucleotide primers will be formed. However, it is
easy to see that after a few cycles, most molecules will
be of just the desired length. A heat-stable polymerase
from Thermus aquaticus (Taq polymerase) is used so that
the enzyme is not denatured by the repeated cycles of
heating and cooling, which are conducted automatically
by a simple apparatus.
The polymerase chain reaction is being used to
speed up prenatal diagnosis of genetic diseases, to
detect viral infections, for tissue typing needed for
organ transplantation, in forensic procedures, and in
the study of the DNA of ancient tissues such as those
of frozen wooly mammoths.
601 603
If suitable restriction
enzyme sites are present in the primers, the amplified
DNA can be cloned readily.
604
The 3.3 x 10
9
fmol of a
DNA sequence found in a diploid chromosome pair in
a single cell can be amplified in 50 cycles to 5500 fmol,
enough to study by hybridization with radioactive
probes.
605
A large sample of a few pg of DNA can be
amplified in 20 cycles to micrograms. By placing
sequencing primers within the amplified segments,
it is possible to generate DNA that can be sequenced
directly using the dideoxy sequencing technique
(Chapter 5) without cloning.
606
The PCR technique
has also been used to amplify cDNA molecules formed
from RNA transcripts present in very low abundance.
One of the problems with the PCR is that priming may
occur by DNA fragments other than the added primers.
Contamination must be scrupulously avoided. Another
problem is that errors are introduced into DNA during
amplification by PCR. Perhaps 1 in 200 of the copies
will contain an incorrect base.
607
If such a molecule is
cloned the error will be perpetuated. Good practice
requires that more than one clone is selected and
sequenced to allow such errors to be avoided.
7. Sequence Determination
Satisfactory (but slow) methods for determining
sequences of RNA molecules have been known for
over 30 years. The procedures are somewhat parallel
to those used in sequencing proteins. However, no
similar method could be devised for DNA. Little
progress was made until rather recently when new
approaches led to extremely rapid procedures for
sequencing DNA. As a consequence, it is now much
easier to learn the sequences of genes than it is to
sequence the proteins which they encode!
Preparing the DNA. The first step is to obtain a
sample of enough identical DNA molecules to permit
sequence analysis. This in itself may be a complex
undertaking. Perhaps we want to know the sequence
of one particular gene in the 3,500,000 kilobase pairs
of DNA present in a single human cell. How can this
gene be found and the DNA be obtained for analysis?
Three techniques have been essential: cutting the DNA
with restriction endonucleases, hybridization, and
cloning. More recently PCR and related methods
608
have simplified the sample preparation. DNA can often
be amplified using primers that contain sequences that
will later serve as sequencing primers.
Restriction maps and Southern blots. Although
it doesnt require the synthesis of a primer, the Maxam
Gilbert procedure usually demands that a restriction
map of the DNA be prepared to help keep track of
the fragments being sequenced.
609
See also Chapter 26.
Figure 5-48
610 612
shows the restriction map of the mito-
chondrial DNA gene, oxi3 from yeast. This gene, which
encodes one of the subunits of cytochrome oxidase
(Chapter 18), consists of 9979 base pairs. It was cloned
in a suitable plasmid after which the restriction map
(Fig. 5-48) was prepared by cutting with 18 different
261
restriction enzymes.
611
The protein subunit contains
510 residues and therefore requires a coding capacity
in the DNA of 1530 base pairs. This is only 16% of the
total length of the gene, the majority of whose DNA is
found in the three large introns.
One way to select a desired segment of DNA from
a digest of chromosomal DNA is to sort out the restric-
tion fragments by gel electrophoresis.
613
The DNA
from the gel can be transferred to a nitrocellulose sheet
while retaining the separation pattern using a method
devised by Southern.
560,614,615
In this Southern blot
technique, solvent flows from a pool beneath the gel
up through the gel and the nitrocellulose sheet into
paper towels. The DNA is trapped on the nitrocellu-
lose in the same pattern observed in the electrophero-
gram. A suitably labeled probe such as cDNA with
incorporated
32
P is flowed repeatedly across the nitro-
cellulose sheet under conditions that favor formation
of hybrids. Only the DNA complementary to the
cDNA probe will retain the label. This DNA can then
be located with the help of a autoradiogram. It is
important that single-stranded DNA be used. If double-
stranded restriction fragments are separated on the
electropherogram they must be denatured while in
place in the gel before hybridization is attempted.
Once the desired piece of DNA has been identified
it is usually necessary to increase its amount. The
conventional approach is to incorporate the DNA
fragment into a plasmid and clone by the methods
described in Chapter 26. The selected DNA can usually
be cut cleanly from the plasmid used for cloning with
the same restriction endonuclease originally used in
H. Methods of Study
Denature (heat)
and anneal primers (cool)
a
b
Genomic
dsDNA or
cDNA
5
3
3
5
5
3
5
3
5
3
3
Primer extension with taq
polymerase and nucleotide
triphosphate mixture
5
c
3
5
+
5
3
5
3
Long templates
+
+
+
c
a
b
Repeat steps a, b, c in cycle 2
c
a
b
Repeat steps a, b, c in cycle 3
+
+
+
+
c
a
b
Cycles 4 to 25
At least 10
5
-fold increase in DNA
Intermediate-
length templates
Short templates
Cycle 1
Cycle 2
Cycle 3
Cycle 4
2X
2X
2X
Figure 5-47 Amplification of DNA
using the polymerase chain reaction
(PCR). Double-stranded DNA is dena-
tured by heating to 90 99 C (step a)
and oligonucleotide primers complemen-
tary to short 12 18 nucleotide sequences
at the two ends of the piece of DNA to
be amplified are annealed to the separated
strands by cooling to 40 75 C (step b).
The two DNA strands serve as templates
for synthesis of new complementary
strands using a heat-stable DNA polymer-
ase and a mixture of the four nucleotide
triphosphates. Nucleotide units are
added to the 3' ends of the primers, with
the new chains growing in the 5' 3'
direction (step c). Steps a, b, and c are
then repeated as many as 30 times using
a thermal cycler device that periodically
raises and lowers the temperature with
a cycle time of a few minutes. The poly-
merase is unharmed by the heating and
is reused in each cycle. An excess of the
primer and of the nucleotide triphos-
phates sufficient for all of the cycles is
present initially. In the early cycles new
long and intermediate length templates
are created. However, the number of
short templates increases exponentially
and the final product consists predomi-
nately of the short selected DNA segment
(short templates).
262 Chapter 5. The Nucleic Acids
fragmenting the DNA. An alternative procedure is to
clone a mixture of DNA fragments and then sort colo-
nies of bacteria containing the cloned fragments using
DNARNA hybridization.
616
Bacteria from the selected
colonies are then propagated to produce large amounts
of the plasmid DNA. Alternatively, PCR can be used
directly on the selected DNA fragment. This is more
often the preferred choice.
604,606
The Sanger dideoxy method. The rapid sequencing
methods all depend upon the fact that single-stranded
DNA fragments under denaturing conditions migrate
on electrophoresis in polyacrylamide gels strictly
according to their length. Thus, if a mixture contains
all lengths of radiolabeled polynucleotides from very
short oligonucleotides to fragments containing 200 or
300 bases, the polynucleotides will all appear, one above
the other, as a series of bands that can be visualized by
radioautography. The first of these methods was pub-
lished by Sanger and Coulson
617
in 1975 and was followed
in 1977 by the method which is now used.
618 621
A sample of double-stranded DNA is denatured.
One of the resulting single strands is used as a template
to direct the synthesis of a complementary strand of
radioactive DNA using a suitable DNA polymerase.
The Klenow fragment of E. coli, DNA polymerase I,
reverse transcriptase from a retrovirus, bacteriophage
T7 DNA polymerase, Taq polymerase, and specially
engineered enzymes produced from cloned genes
have all been used.
Before the sequencing begins it is necessary to
prepare a short primer that is complementary to a
sequence at one end of the DNA strand to be sequenced.
This may be prepared enzymatically,
622,623
or by non-
enzymatic synthesis. The short primer is annealed to
the end of the DNA and the resulting molecule is incu-
bated with a DNA polymerase and a mixture of the four
mononucleotide triphosphates, one of which is radio-
labeled in this position. Four reaction mixtures are
prepared. Each mixture contains all four nucleoside
triphosphates and also one of four different chain-
terminating inhibitors, the most popular of which
are the 2', 3' dideoxyribonucleoside triphosphates:
These inhibitors are added in a ratio of about 100:1
with the natural substrates. In this ratio they are
incorporated into the growing DNA chain about once
in 200 times on the average. However, in the various
growing DNA chains they are incorporated at different
points ranging from the very first nucleotide to the
last. Since the incorporated dideoxy monomer lacks
the 3-hydroxyl group needed for polymer formation,
chain growth is terminated abruptly. Synthesized
polynucleotides are denatured and subjected to electro-
phoresis in four adjacent lanes. The resulting patterns
contain bands corresponding to all of the successive
oligonucleotides but not all in the same lane. A given
lane will contain only the bands of the oligonucleotides
terminated by the particular inhibitor used. The other
bands will be found in the other three lanes. Each
band will have been terminated by the inhibitor em-
ployed in that lane. The nucleotide sequence can be
read directly from the banding pattern as is shown in
Fig. 5-49. Arabinosyl nucleotide triphosphates have
also been used as chain-terminating inhibitors. A
sequence determined by the Sanger method is usually
checked by also sequencing the complementary strand.
P O O
HO
O
P
O
O
P
O O
O CH
2
O
H H
H H
O

Thymine
Cytosine
Guanine or
Adenine
2',3'-Dideoxyribonucleotide triphosphates
Figure 5-48 A physical map of the oxi3 locus of yeast
mitochondrial DNA. The restriction fragments used for
DNA sequencing are indicated by the arrows. The extent to
which the sequences were read is represented by the lengths
of the arrows. The map units are shown in the inner circle.
The following symbols, together with the names of the
restriction enzymes (Chapter 26), are used for the restriction
sites:
From Bonitz et al.
611
Hinf I
Hpa II
Hae III
Taq I
Mbo I
Mbo II
Hha I
Rsa I
Hph I
Blg II
Bam HI
Hpa I
Alu I
Pvu II
Hinc II
Hind III
Eco RI
Eco RII
263
A major factor in the success of the dideoxy sequenc-
ing method has been the development of cloning
techniques that provide ssDNA in a form ready for
use. In any cloning procedure the DNA that is to be
sequenced has been covalently ligated to the end of a
DNA strand of the cloning vehicle, a modified plas-
mid or virus (Chapter 26). Sequencing is often done
on DNA cloned in a modified ssDNA bacterial virus
such as M13. Although dsDNA is ligated to the ds
replicating form of viral DNA, the virus particles
produced when the virus is propagated in E. coli cells
are single stranded. Cleavage of the viral DNA with
its incorporated passenger DNA with an appropriate
restriction endonuclease releases the passenger DNA
(to be sequenced) with a short piece of DNA from
the cloning vehicle attached at the 3' end. Since the
sequence of this small piece of the cloning vehicle is
known, a suitable primer of length ~1218 nucleotides
can be synthesized (or purchased) and annealed to
the DNA. This serves as the primer and allows the
sequence to be read from the 5' end to the 3' end of the
synthesized complementary strand. Double-stranded
DNA attached to vehicles such as the pUC plasmids
can also be sequenced directly if the DNA is denatured
by alkali treatment. After neutralization and precipita-
tion an appropriate primer is annealed to one or the
other of the two strands.
624
Since its introduction, many modifications and
improvements have been developed. The sequencing
gels have been improved. The use of
35
S labeling has
given sharper autoradiographs.
625
Alternatively, a
silver stain can be used with unlabeled primers.
626
GC-rich DNA sequences are often difficult to sequence,
probably because even in the denaturing polyacrylamide
gels used for sequencing they tend to form hairpin
loops, perhaps as a result of formation of Hoogsteen
base pairs (Fig. 5-7). Formation of these loops results
in uneven spacing between the adjacent bands in the
sequencing gel, so-called compression artifacts. Use
of a 7-deaza-dGTP in place of dGTP in the sequencing
reaction ameliorates this problem.
627
Sensitivity can be
improved by use of cycle sequencing in which a heat-
stable polymerase such as Taq polymerase is used, and
after heating the same template DNA is used repeatedly
to give a higher yield of labeled fragments.
628,629
About 200 400 bases can be successfully sequenced
manually in a single run. By prolonging the time of
electrophoresis in a second run, the sequence can be
extended considerably. By using the just obtained
sequence information, it is possible to select a new
start point 200 or more nucleotides further along the
template chain and to synthesize an oligonucleotide
primer to anneal to the template at this point. In this
way it is possible to walk along the template adding
additional sequences at each step. Another procedure
is to delete by mutation various segments of the cloned
DNA above the sequence that binds the primer. This
H. Methods of Study
Figure 5-49 A DNA sequencing gel obtained using a
segment of DNA from salmon sperm selected by suitable
oligonucleotide primers, amplified by PCR, and sequenced
with a
35
S label in the primer. Four samples were used, one
with each of the four dideoxy chain terminators (A, G, C, T,
A, C, G, T from left to right). After electrophoresis the shorter
fragments are at the lower end of the gel. The sequence of
the strand complementary to the template strand whose
sequence is being determined is read from the bottom of the
gel. Here it starts CTATGATAC. Reproduced by permission
of Amersham Pharmacia Biotech, Limited.
C
T
A
T
G
A
T
A
C
C A G T C A G T
264 Chapter 5. The Nucleic Acids
permits analysis of the whole cloned fragment via an
overlapping set of sequences from the deletion mu-
tants.
630,631
Many DNA sequences continue to be determined
manually by the well-developed long gel procedures
as illustrated in Fig. 5-49. However, sequencing whole
genomes has depended upon the development of high-
speed automated procedures.
632
Instead of radiolabel-
ing, fluorescent dyes may be joined to the primer to
allow detection of the chain fragments produced during
sequencing. Automatic sequencers use four dyes that
fluoresce with different colors.
633,634
A different dye is
used for each of the reaction mixtures. Then the four
samples are mixed together and the DNA fragments
are subjected to electrophoresis in a single lane. A laser
beam excites the fluorescence, scanning several lanes
with different samples as the electrophoresis progresses.
A photomultiplier tube records the fluorescence inten-
sity of each band through a series of four filters in a
rotating wheel. This allows automatic recognition of
the four different colors of fluorescence and therefore
of the nucleic acid base present in each position in the
sequence. Improved strategies for primer walking
635
and for shotgun sequencing
19,632,636
have been devised.
In the shotgun strategy, whole bacterial genomes have
been cut by restriction enzymes into large numbers of
overlapping fragments which have been separated and
sequenced.
637
A computer program is used to analyze
and assemble the sequences into a complete genomic
sequence. An example is provided by the genome of
the Methanococcus jannaschii. Its large circular chromo-
some contains 1,664,976 bp and there are two additional
pieces to the genome, one containing 58,407 bp and the
other 16,550 bp. The sequences were deduced from
36,718 individual sequencing runs on high-speed auto-
matic sequencers. For each run, on average, 481 bp
could be read.
636
To sequence the human genome faster
methods are needed.
638
Capillary electrophoresis with
a single laser beam scanning the output of 24 capillaries
has been demonstrated.
639,640
Extremely rapid sequenc-
ing of oligonucleotides up to 100 bp in length can be
accomplished by mass spectrometry.
641
This may be
an important technique for diagnosis of genetic defects
(Chapter 26).
The method of Maxam and Gilbert. The nonen-
zymatic method devised by Maxam and Gilbert
642 644
can be used to sequence either ss or dsDNA. Before
the sequencing is begun, a radioactive label is incor-
porated, usually at the 5' end. This is often done by
cleaving off any phosphate groups present on the 5'
end with alkaline phosphatase and then transferring
a new radioactive phospho group with the assistance
of the enzyme polynucleotide kinase and radioactive
-
32
p-labeled ATP. If dsDNA is used the strands are
separated so that each has a label only at one end.
The key step in sequencing by the MaxamGilbert
procedure is to cleave chemically the DNA at random
locations using reagents that have some specificity for
particular bases. The cleavage process involves three
distinct steps: (1) chemical modification, as specific as
is possible for the chosen base; (2) displacement of the
modified base from the sugar; and (3) elimination and
chain cleavage using amine catalysis (Eq. 5-21). Two
consecutive steps can often be combined. There are
several versions of the method; one involves dimethyl
sulfate as the specific reagent for guanine. It forms
N
7
-methylguanosine (see Eq. 5-18) which upon heating
with the strong base piperidine at 90C undergoes
addition of hydroxyl ion with ring opening and dis-
placement of the modified base according to Eq. 5-26.
The product, a glycosylamine of piperidine, is in
equilibrium with a Schiff base which can undergo
chain cleavage as in Eq. 5-21.
A second sample of DNA is treated with a piperidine-
formate buffer of pH 2 in the cold. This promotes the
acid depurination of both guanine and adenosine.
A third sample is treated with hydrazine, with both
cytidine and thymidine being cleaved to ribosylurea
according to Eq. 5-13. Again, this is followed by dis-
placement and elimination (Eq. 5-21) catalyzed by
piperidine. The fourth sample is also treated with
hydrazine but in the presence of a high concentration
of NaCl which inhibits the reaction with thymidine,
by lowering the pK
a
of the thymine, and allows the
cleavage to be more nearly specific for cytidine. Each
of the reactions is conducted in such a way that on the
average only one cleavage event occurs per molecule
of DNA. Since the cleavages occur at many different
points, a family of nested radioactively labeled oligo-
nucleotides, one from each original molecule, is pro-
duced. When these are sorted by polyacrylamide gel
electrophoresis, the pattern of the oligonucleotides
in the four adjacent channels allows the nucleotide
sequence to be read directly from the autoradiogram.
The MaxamGilbert method doesnt require
synthesis of a primer and it sometimes works well for
sequences that are difficult to obtain with the Sanger
Coulson procedure. The two methods may both be
used to provide additional certainty about a sequence.
The MaxamGilbert method is very convenient for
sequencing small oligonucleotides which often react
poorly with the polymerase used for the chain termi-
nation method. The method usually requires that a
restriction map be prepared.
Sequencing RNA. The first known RNA sequence,
that of an alanine tRNA, was determined by Holley
and associates in 1965. The RNA was subjected to
partial hydrolysis with pancreatic ribonuclease and
ribonuclease T
1
(Fig. 5-43). The small oligonucleotide
fragments were separated by ion exchange chromato-
graphy under denaturing conditions (7 M urea) and
were then characterized individually.
645
The availability
265
of additional enzymes such as ribonuclease U
2
, the B
cereus ribonuclease and ribonuclease Phy M of Physarum
(Fig. 5-43) and the use of radiolabeling and of two-
dimensional fingerprinting of digests have made the
procedures more versatile.
646
A valine tRNA was se-
quenced independently by Bayev
647
and Campbell.
648
Since the development of the rapid methods for
sequencing DNA, many mRNA sequences have been
determined by using reverse transcriptase to make a
cDNA strand complementary to the RNA. The cDNA
is then sequenced.
649
Rapid sequencing methods parallel
to those used for DNA have also been devised.
650 652
Nearest neighbor analysis. A technique developed
by Kornberg and associates before the availability of
sequencing methods is the nearest neighbor sequence
analysis. Using a single radioactive
32
P-containing
nucleoside triphosphate together with the three other
unlabeled nucleoside triphosphates, a primer chain
of DNA is elongated from the 3' end along a ssDNA
template chain using a DNA polymerase. The incor-
poration of
32
P from the position of the nucleotide
triphosphate occurs in the bridge phosphates that
connect the nucleotide originally carrying the
32
P to
the 3' position of the neighboring nucleotide. Cleavage
of the
32
P-containing product of the reaction with a
mixture of micrococcal DNase and spleen phospho-
diesterase, which catalyze b-type cleavage (Fig. 5-43),
gives fragments in which the
32
P will now be attached
to what was the 5 nearest neighbor to the radioactive
nucleotide in the DNA.
72,653
Measurement of the
radioactivity in each of the 3' nucleotides of thymine,
cytosine, adenine, and guanine gives the frequencies
of the adjacent pairs, TA, CA, AA, and GA. Using the
other radioactive nucleoside triphosphates one at a
time in separate experiments, all of the nearest neighbor
frequencies can be obtained. From such an experiment
it was possible to deduce that the strands in the double
helix were oriented in an antiparallel fashion, as pre-
dicted by Watson and Crick. If the strands had been
parallel, different nearest neighbor frequencies would
have been observed.
Understanding sequences. Sequences of over 20
million nucleotides from hundreds of organisms had
been determined by 1988 and the number is doubling
each 2 3 years.
654
Sequencing the human genome has
required rates of millions of bases per day. With the
massive amount of data already available it has become
of great interest to compare sequences of genes, whether
they encode similar or dissimilar proteins, to make
comparisons between species, and to search for
sequences that bind specific proteins or that encode
particular regulatory signals. Relationships of common
evolutionary origin or homology as well as other
sequence similarities are often sought.
To handle the mass of existing data, powerful
computer programs have been developed and various
graphical procedures have also been developed to
help the human mind comprehend the results.
654,655
One important problem is to define and locate what
are called consensus sequences. The problem is best
illustrated by examples.
654
The cleavage site for the
EcoRI restriction endonuclease is GAATTC. There
is no ambiguity. In a DNA of random sequence this
would be expected to occur by chance in about (1/ 4)
6
nucleotides (4 kb). On the other hand, the HinII re-
striction endonuclease cleaves within the consensus
sequence GTYRAC where Y = C or T and R = A or G.
It would be expected to occur by chance in about 1/4
5
nucleotides. Many binding sites for RNA polymerase,
the so called promoters (Chapter 28) contain the
consensus sequence TAtAaT, at position -10, ahead of
the 5' end of the sequence that is transcribed into mRNA.
The lower case t and a used here imply that other
nucleotides may often replace T or A at these positions.
There are many promoters and over 70% of those
described have this consensus sequence. All have the
less restricted sequence TAxxxT, where x may be any
nucleotide. Our definition of consensus sequence is some-
what arbitrary. Now consider the problem of locating
a 35 site whose consensus sequence is TTGACA
but which may, for different genes, be shifted back-
ward or forward by a nucleotide or two. This is a
consensus sequence. Therefore, in many cases one
or more substitutions in the sequence will have been
made. The result is that the sequence of nucleotides in
N
N
N
N
H
NH
2
O
CH
2
O
O
P
CH
3
O
N
N
N
H
NH
2
O
CH
2
O
O
P
O
N
C
O
H
CH
3
N
O
CH
2
O
O
P
+
HO

N
Piperidine
a
b
Base
displacement
(5-26)
H. Methods of Study
266 Chapter 5. The Nucleic Acids
which the consensus sequence is to be found is likely
to appear entirely random. Sophisticated computer
programs are helpful in locating it.
654
8. ProteinDNA Interactions
The most detailed information about interactions
of proteins with DNA is coming from X-ray crystallo-
graphic studies. Examples are seen in Figs. 5-35 to 5-40.
Several other methods have also been very useful.
Much has been learned from the effects of mutations
in DNA-binding proteins or in regions of DNA to which
a protein binds. Binding of proteins to DNA can also
be recognized by its effects on the mobility of DNA
during gel electrophoresis.
656,657
Chemical
658
or laser-
induced crosslinking can show that within a complex
a specific residue in a protein is adjacent to a certain
sequence in the DNA.
The technique of protection mapping or foot-
printing is widely used to determine which nucleo-
tides in a sequence are covered by a bound protein.
659
A reagent which attacks and cleaves DNA nearly
randomly is used. DNase I was first introduced for
this purpose
659
and has been used widely. An impor-
tant finding is that certain sites that are readily cleaved
(hypersensitive sites) are frequently located in chro-
matin undergoing transcription. Footprinting has also
been accomplished with other nucleases, with dimeth-
ylsulfate (which acts on A and C), with carbodiimides
(which act on U and G), and with the MaxamGilbert
guanine-specific cleavage (Eq. 5-26). One of the most
popular methods employs cleavage by hydroxyl radi-
cals.
660 663
Photofootprinting depends upon decreased
or increased sensitivity to ultraviolet light at sites
bound by proteins.
214,662,664
In footprinting experiments
the DNA to be studied is radioactively labeled at one
end of one strand. In the absence of the protecting
protein, denaturation and electrophoresis of the cleaved
fragments yields a nearly random ladder of DNA
fragments. In the presence of the binding protein some
cleavage products will be missing from the ladder. The
bound protein leaves a footprint (Fig. 5-50A,C).
Related methods are being applied to the determi-
nation of the secondary structure of RNA molecules
665,666
and to the study of interactions with proteins. For
example, treatment with dimethyl sulfate under appro-
priate conditions methylates bases that are not paired,
giving largely 1-methyladenosine and 3-methyl-
cytidine.
667
9. Nuclear Magnetic Resonance
Much of the initial effort to study polynucleotides
by NMR spectroscopy was directed toward transfer
RNAs, only a few of which have been crystallized in a
form suitable for X-ray diffraction. Study of the other
tRNAs by NMR techniques has established that all
of the tRNAs have a similar architecture and that the
structures observed in the crystals are preserved in
solution.
668
Figure 5-51 shows the low-field end of the
NMR spectrum of a valine-specific tRNA from E. coli.
The spectrum is run in H
2
O rather than D
2
O so that
exchangeable hydrogens in the hydrogen bonds of
the WatsonCrick base pairs can be observed.
669
The
protons giving rise to the downfield resonances are
primarily those attached to nitrogen atoms of the rings
and in hydrogen-bonded positions. These protons are
shielded by adjacent electron-donating groups and by
their attachment to the semiaromatic rings of the bases.
The NMR signals are further shifted downfield to
varying degrees depending upon whether or not the
proton being observed is attached to a base that is
stacked with other bases. The size of the shift also
depends upon which neighboring bases are present.
The proton on N-3 of AU base pairs is deshielded
more than the proton on the N-1 of GC base pairs.
Therefore, the AU protons appear further downfield
than the GC protons. The stronger ring current in A
than in C enhances this separation.
All of the resonances in Fig. 5-51 have been
assigned to particular bases. This was done in part by
varying the temperature, changing the magnesium
ion concentration, and predicting shifts caused by
ring currents in adjacent bases making use of the X-ray
crystal structures. NMR spectra of hairpin helical frag-
ments also provided essential information. From inte-
gration of the areas under the peaks it was concluded
that the 20 resonances seen below 11 ppm represent
27 protons. Twenty of these are in Watson-Crick base
pairs and correspond to those expected from the X-ray
structure. Six more belong to protons involved in
tertiary interactions, such as base pair triplets or non-
WatsonCrick pairs. One of these, labeled G in the
figure, is in the dihydrouridine stem and involves the
ring proton of N-1 of m
7
G46 which is hydrogen bond-
ed to N-7 of G22 in the major groove of the RNA. G22
is located at the beginning of the extra loop.
Measurements of the NOE of nearby protons in
both small RNA molecules
668,669
and DNA oligonu-
cleotides
670,671
provided much additional information.
Figures 5-51B and C show NOESY spectra of the
tRNA
Val
and the way in which weak cross-peaks
between the H-bonded imino protons in adjacent base
pairs (Fig. 5-51B) can be used to establish connec-
tivities.
669
Beginning with resonance B, it is possible
to establish the sequence of the NH groups giving rise
to these resonances as OBUGJNT. Using other data as
well, it was concluded that these represent the seven
base pairs of the acceptor stem (see Fig. 5-30), with
resonance C representing the first GC pair, resonance
B the second, etc. Resonance A was identified as
coming from the base triplet containing a Hoogsteen
267
base pair of 4-thiouracil at position 8 with A14 (see Fig.
5-7). Its connectivity to the sequence KCEO is also
outlined in Fig. 5-51B. However, it could not be estab-
lished without additional data which also helped to
identify the sequence O-K as residues 10 13 of the
dihydrouracil stem. Peak O is a multiproton peak
representing not only GC 10 but also UA 7. In general,
the GC protons are at the higher field side of the spec-
trum and the AU protons at the lower side. However,
AU 7 is shifted to an anomolously high position.
Cross-peaks between imino protons of uracil and the
nearby C2 protons of adenine in WatsonCrick AU base
pairs or C8 protons of Hoogsteen AU pairs can be ob-
served in the 6.5 9 ppm region as shown in Fig. 5-35B.
This region also contains information about other
protons bound to the nuclei acid bases.
Figure 5-50 (A) Hydroxyl radical footprints of phage
repressor on both strands of the 120-bp restriction
fragment of E. coli DNA carrying the O
R
1 operator
sequence. Bottom and top refer to the DNA
strands as drawn in (C), and as diagrammed in (B).
Labeling of the 5' end of the Bgl II restriction fragment
afforded data for the bottom strand and labeling of
the 3' Bgl II end afforded data for the top strand. Lanes
1 and 12, untreated DNA, labeled on the bottom and
top strands, respectively; lanes 2 and 10, products of
DNase I digestion of DNA labeled on the bottom and
top strands, respectively, with no repressor present;
lanes 3 and 11, products of DNase I digestion of DNA
labeled on the bottom and top strands, respectively,
complexed with repressor (675 nM); lanes 4 and 9,
products of MaxamGilbert guanine-specific sequenc-
ing reactions performed on DNA labeled on the bottom
and top strands, respectively; lanes 5 and 6, products
of hydroxyl radical cutting of DNA labeled on the
bottom strand, complexed with repressor. Lane 5,
90-nM repressor and lane 6, 675-nM repressor.
Lanes 7 and 8, products of hydroxyl radical cutting
of DNA labeled on the top strand, complexed with
repressor. Lane 7, 675 nM repressor. Lane 8, 90 nM
repressor. The labels a, b, c, a, b, and c mark the
hydroxyl radical footprints. Courtesy of Thomas
Tullius.
661
H. Methods of Study
Similar techniques are being used for the study of
DNA.
672
The presence of a second hydrogen in the 2'
position of the deoxyribose rings of DNA adds several
H-H distances (Fig. 5-52) that can be measured in addi-
tion to those seen in RNAs. Characteristic differences
are seen in the NOESY plots of A, B, and Z forms of
DNA.
670,671,673,674
Although detailed structural infor-
mation has been obtained for short segments of DNA,
spectra of larger oligonucleotides are impossible to
analyze with two-dimensional methods because of
extensive overlap of resonances.
665
The difficulty is
already apparent in the 17 base pair DNA segment for
which a one-dimensional spectrum as well as COSY
and NOESY spectra are shown in Fig. 5-53.
Some help with the complexity can be obtained
by incorporation of
13
C-enriched methyl groups into
A
C
B
268 Chapter 5. The Nucleic Acids
a
b
c
d
g
o
15.0 14.0 13.0 12.0 11.0 10.0 9.0 8.0 7.0
15.0
14.0
13.0
12.0
11.0
10.0
9.0
8.0
7.0
A
B
C
D
E
F
G
H
I
J L
K
M
N
O
P
Q
R
S
T
U
V
W
X
15 14 13 12 11 10 ppm
10
11
12
13
14
15
E
C
O
K
R
U
O
G
N
J
T
K
P
L
V
F
Q
D
H2
AH8
H2
H2
TH5
H2
H2
TMe
AH8
H
H
H
H
H
H
H
H
H
A
T
A
Figure 5-52 Segment of a DNA chain in the B conformation illustrating
some intrachain NOEs that may be observed. The close juxtaposition of
proton pairs is provided by the H2'-endo (
2
E) conformation of the sugar
rings with anti base conformation (Fig. 5-11). After Cohen.
675
Figure 5-51 (A) The low-field region of the one-dimensional
1
H NMR spectrum of E. coli tRNA
1
Val
at 27C in H
2
O. Resonances
are identified by letters A X. (B) NOESY spectrum of the same tRNA under similar conditions showing the imino-imino NOEs.
In the lower right sector the connectivity traces of the acceptor helix and dihydrouridine helix are shown as solid and dotted
lines, respectively. In the NOESY sample the two protons in peak EF are partially resolved whereas the two protons in peak
T have coalesced. (C) NOESY spectrum of E. coli tRNA
1
Val
at 32C showing the imino and aromatic proton regions. AU-type
imino protons have been connected horizontally by a dotted line to the cross-peak of their proximal C2-H or C8-H in the 7 to 9
ppm region, which has been labeled with the corresponding lower-case letter. From Hare et al.
669
Courtesy of Brian Reid.
B
C
A
269
5 6 7 8 4 3 2 1 ppm
C8H
C2H
C6H
C1H
C5H
C3H
C4H
C5,5H
C2,2H
CH
3
A T G C A T G G G T T C C C T A T
T A C G T A C C C A A G G G A T A
1 5
34 30
10
25
15 17
20 18
OR3
1.0
ppm
2.0 3.0 4.0 5.0 6.0 7.0 8.0
1.0
2.0
3.0
4.0
5.0
6.0
7.0
8.0
ppm
OR3
a
b
c
d
e
f
g
H. Methods of Study
Figure 5-53 (A)
1
H NMR spectrum of a 17 base-pair DNA segment from the operator sequence OR3 from bacteriophage in
D
2
O at 37C. (B) Combined COSY above the diagonal and NOESY (below the diagonal) spectra. C5H and C6H J coupling is
established from cross-peaks in box d for cytosines and in box a for thymines. Two unresolved cross-peaks give rise to the more
intense spots marked by arrows. Box b contains cross-peaks from scalar coupling of the two H2' protons to the H1' protons
of the deoxyribose rings. Most of the aromatic proton resonances could be assigned using the NOE cross-peaks in box f. For
further details see Wemmer et al.
676
See also Bax and Lerner.
672
Courtesy of B. Reid.
B
A
270 Chapter 5. The Nucleic Acids
1. Portugal, F. H., and Cohen, J. S. (1977) A
Century of DNA: A History of the Discovery of
the Structure and Function of the Genetic Sub-
stance, MIT Press, Cambridge, Massachusetts
2. Levene, P. A., and Tipson, R. S. (1935) J. Biol.
Chem. 109, 623630
3. Schlenk, F. (1988) Trends Biochem. Sci. 13, 6769
4. Watson, J. D., and Crick, F. H. C. (1953) Nature
(London) 171, 737738
5. Watson, J. D. (1968) The Double Helix, Atheneum,
New York
6. Crick, F. (1988) What a Mad Pursuit: A Personal
View of Scientific Discovery, Basic Books, New
York
7. Olby, R. (1974) The Path to the Double Helix,
Macmillan, London
8. Wilson, H. R. (1988) Trends Biochem. Sci. 13,
275278
9. Chambers, D. A., Reid, K. B. M., and Cohen,
R. L. (1994) FASEB J. 8, 12191226
9a. Piper, A. (1998) Trends Biochem. Sci. 23, 151154
10. Arnott, S., and Hukins, D. W. L. (1973) J. Mol.
Biol. 81, 93105
11. Proudfoot, N. J., Shander, M. H. M., Manley, J.
L., Gefter, M. L., and Maniatis, T. (1980) Science
209, 13291336
12. Tucker, P. W., Liu, C.-P., Mushinski, J. F., and
Blattner, F. R. (1980) Science 209, 13531360
13. Yamada, Y., Mudryj, M., Sullivan, M., and de
Crombrugghe, B. (1983) J. Biol. Chem. 258,
27582761
14. Tsuzuki, T., Obaru, K., Setoyama, C., and
Shimada, K. (1987) J. Mol. Biol. 198, 2131
15. Ratner, L., and 18 other authors (1985) Nature
(London) 313, 277284
16. Dunn, J. J., and Studier, F. W. (1983) J. Mol.
Biol. 166, 477 535
17. Peden, K. W. C. (1983) Gene 22, 277280
Figure 5-54 (A) An
19
F NMR spectrum of the 76-residue
E. coli tRNA
Val
containing 5-fluorouracil in 14 positions.
Recorded at 47C. The numbers above the resonances
indicate the position in the sequence. (The sequence is not
identical to that for the yeast tRNA shown in Fig. 5-30.)
Modified from Chu et al.
694
Courtesy of Jack Horowitz.
(B) A similar spectrum for a 35-residue minihelix that contains
the acceptor stem of the tRNA
Val
and seven fluorouracils.
The broad peaks B, D, and E are shifted far upfield by reac-
tion with bisulfite (Eq. 5-11) suggesting that they are not
hydrogen bonded and are present in the loop of the stem
loop structure. Peaks A, E, F, and G correspond to resonances
64, 7, 67, and 4, respectively, in (A) and represent fluorouracil
in the stem structure. From Chu et al.
694
Courtesy of Jack
Horowitz. (C) A
31
P NMR spectrum of a synthetic 14 base-
pair DNA segment related to the E. coli lac operator. The
palindromic sequence is TCTGAGCGCTCAGA. The numbers
refer to the positions from the 5' end. From Schroeder et al.
688
polynucleotides. One way to do this is to grow yeast
on a medium containing methionine with
13
C in its
methyl group. The most intense peaks in
13
C NMR
spectra of tRNA molecules from this yeast will represent
methyl groups in modified bases in the tRNA. Incor-
porated
13
C also permits study of internal motion with-
in tRNA or other oligonucleotides by NMR methods.
677
However, as with protein NMR spectroscopy the major
recent advances have come from systematic incorpora-
tion of both
13
C and
15
N into nucleic acids and the
development of three- and four-dimensional NMR
methods.
678684b
Also important are methods for replac-
ing some hydrogen atoms with deuterium to simplify
spectra.
685,686
The very sensitive
19
F nucleus can be introduced
into tRNAs by incorporation of 5-fluorouracil in place
of uracil
687
(Fig. 5-54A,B). Phosphorus 31 NMR spectra
(Fig. 5-54C) can provide information about conforma-
tions of the chain.
688
A few recent NMR investigations of polynucle-
otides include studies of triple-helical DNA,
689
Holli-
day junctions,
290
double-stranded oligonucleotides
containing adducts of carcinogens,
690,691
of hairpin
loops with sheared AA and GG pairs,
692
and of
proton exchange in both imino and amino groups.
693
55
64
59
33
47
17
34
7
54
67
8
4
12
29
E F
G
B/C/D
A
ppm 8 7 6 5 4 3 2 1
3.4 3.6 3.8 4.0 4.2 4.4 4.6 4.8 5.0 ppm
C11
G4
T3.T13
G6.G8.T1
C7
A5
T10.A12
C9
A2
A
B
C
References
271
References
18. Anderson, S., Bankier, A. T., Barrell, B. G.,
deBruijn, M. H. L., Coulson, A. R., Drouin, J.,
Eperon, I. C., Nierlich, D. P., Roe, B. A.,
Sanger, F., Schreier, P. H., Smith, A. J. H.,
Staden, R., and Young, I. G. (1981) Nature
(London) 290, 457470
19. Fraser, C. M., and 28 other authors (1995)
Science 270, 397403
20. Fleischmann, R. D., and 39 other authors
(1995) Science 269, 496512
20a. Doolittle, R. F. (1998) Nature (London) 392,
339342
21. Nowak, R. (1995) Science 267, 172174
22. Dujon, B., and 107 other authors (1994) Nature
(London) 369, 371378
23. Johnston, M., and 34 other authors (1994)
Science 265, 20772082
24. Goffeau, A., Barrell, B. G., Bussey, H., Davis,
R. W., Dujon, B., Feldmann, H., Galibert, F.,
Hoheisel, J. D., Jacq, C., Johnston, M., Louis,
E. J., Mewes, H. W., Murakami, Y., Philippsen,
P., Tettelin, H., and Oliver, S. G. (1996) Science
274, 546557
24a. Macilwain, C. (2000) Nature (London) 405,
983 984
25. Libecq, C., ed. (1992) Biochemical Nomencla-
ture, Portland Press, London and Chapel Hill,
North Carolina (for the International Union of
Biochemistry and Molecular Biology)
26. Shabarova, Z., and Bogdanov, A. (1994)
Advanced Organic Chemistry of Nucleic Acids,
VCH Publ., Weinheim
27. Saenger, W. (1984) Principles of Nucleic Acid
Structure, Springer, New York
28. Hurst, D. T. (1980) An Introduction to the
Chemistry and Biochemistry of Pyrimidines,
Purines and Pteridines, Wiley, New York
29. Markowski, V., Sullivan, G. R., and Roberts, J.
D. (1977) J. Am. Chem. Soc. 99, 714718
30. Katritzky, A. R., and Karelson, M. (1991) J.
Am. Chem. Soc. 113, 15611566
31. Szczesniak, M., Leszczynski, J., and Person,
W. B. (1992) J. Am. Chem. Soc. 114, 27312733
32. Scanlan, M. J., and Hillier, I. H. (1984) J. Am.
Chem. Soc. 106, 37373745
33. Brown, D. M. (1974) in Basic Principles in
Nucleic Acid Chemistry, Vol. II (Tso, P. O. P.,
ed), pp. 290, Academic Press, New York
34. Hoogsteen, K. (1963) Acta Crystallogr. 16, 907
916
35. Frank-Kamenetskii, M. D., and Mirkin, S. M.
(1995) Ann. Rev. Biochem. 64, 6595
36. Portugal, J. (1989) Trends Biochem. Sci. 14, 127
130
37. Adams, R. L. P., Knowler, J. T., and Leader, D.
P. (1992) The Biochemistry of the Nucleic Acids,
11th ed., Chapman & Hall, London
37a. Saenger, W. (1973) Angew. Chem. Int. Ed. Engl.
12, 591601
38. Fang, G., and Cech, T. R. (1993) Biochemistry
32, 1164611657
39. Gellert, M., Lipsett, M. N., and Davies, D. R.
(1962) Proc. Natl. Acad. Sci. U.S.A. 48, 2013
2018
40. Kennell, D. E. (1971) Prog. Nucleic Acid Res.
Mol. Biol. 11, 259301
40a. Stofer, E., Chipot, C., and Lavery, R. (1999)
J. Am. Chem. Soc. 121, 95039508
40b. Peyret, N., Seneviratne, P. A., Allawi, H. T.,
and SantaLucia, J., Jr. (1999) Biochemistry 38,
34683477
41. Pranata, J., Wierschke, S. G., and Jorgensen,
W. L. (1991) J. Am. Chem. Soc. 113, 28102819
42. Dang, L. X., and Kollman, P. A. (1990) J. Am.
Chem. Soc. 112, 503507
43. Newcomb, L. F., and Gellman, S. H. (1994)
J. Am. Chem. Soc. 116, 49934994
44. Borer, P. N., Dengler, B., Tinoco, I., Jr., and
Uhlenbeck, O. C. (1974) J. Mol. Biol. 86, 843
853
45. Freier, S. M., Kierzek, R., Jaeger, J. A.,
Sugimoto, N., Caruthers, M. H., Neilson, T.,
and Turner, D. H. (1986) Proc. Natl. Acad. Sci.
U.S.A. 83, 93739377
46. Sarai, A., Mazur, J., Nussinov, R., and Jernigan,
R. L. (1988) Biochemistry 27, 84988502
47. Hunter, C. A. (1993) J. Mol. Biol. 230, 10251054
48. Friedman, R. A., and Honig, B. (1995) Biophys.
J. 69, 15281535
49. DePrisco Albergo, D., Marky, L. A., Breslauer,
K. J., and Turner, D. H. (1981) Biochemistry 20,
14091413
50. Privalov, P. L., and Filimonov, V. V. (1978)
J. Mol. Biol. 122, 447464
51. Privalov, P. L., and Filimonov, V. V. (1978)
J. Mol. Biol. 122, 465470
52. Petersen, S. B., and Led, J. J. (1981) J. Am.
Chem. Soc. 103, 53085313
53. Petruska, J., and Goodman, M. F. (1995) J. Biol.
Chem. 270, 746750
54. SantaLucia, J., Jr., Allawi, H. T., and Seneviratne,
P. A. (1996) Biochemistry 35, 35553562
55. Egli, M., Usman, N., and Rich, A. (1993)
Biochemistry 32, 32213237
56. Watson, J. D., and Crick, F. H. C. (1953) Nature
(London) 171, 964967
57. Topal, M. D., and Fresco, J. R. (1976) Nature
(London) 263, 285289
58. Tinoco, I., Jr., Borer, P. N., Dengler, B., Levine,
M. D., Uhlenbeck, O. C., Crothers, D. M., and
Gralla, J. (1973) Nature New Biol. 246, 4041
59. Fazakerley, G. V., Gdaniec, Z., and Sowers, L.
C. (1993) J. Mol. Biol. 230, 610
60. Florin, J., Hrouda, V., and Hobza, P. (1994)
J. Am. Chem. Soc. 116, 14571460
61. Douhal, A., Kim, S. K., and Zewail, A. H.
(1995) Nature (London) 378, 260263
62. Gabb, H. A., and Harvey, S. C. (1993) J. Am.
Chem. Soc. 115, 42184227
63. Ellervik, U., and Magnusson, G. (1994) J. Am.
Chem. Soc. 116, 23402347
64. Thibaudeau, C., Plavec, J., and
Chattopadhyaya, J. (1994) J. Am. Chem. Soc.
116, 80338037
65. Santos, R. A., Tang, P., and Harbison, G. S.
(1989) Biochemistry 28, 93729378
66. Sundaralingam, M. (1969) Biopolymers 7, 821
860
67. Vologodskii, A. (1992) Topology and Physics of
Circular DNA, CRC Press, Boca Raton, Florida
68. Baikalov, I., Grzeskowiak, K., Yanagi, K.,
Quintana, J., and Dickerson, R. E. (1993) J. Mol.
Biol. 231, 768784
69. Lipanov, A., Kopka, M. L., Kaczor-Grzeskowiak,
M., Quintana, J., and Dickerson, R. E. (1993)
Biochemistry 32, 13731389
70. Schlick, T., Hingerty, B. E., Peskin, C. S., Overton,
M. L., and Broyde, S. (1991) Theoretical
Biochemistry and Molecular Physics, Vol. 1
(Beveridge, D. L. and Lavery, R., eds), pp.
3958, Adenine Press, Guilderland, New York
71. Wang, A. H.-J., Quigley, G. J., Kolpak, F. J.,
Crawford, J. L., van Boom, J. H., van der
Marel, G., and Rich, A. (1979) Nature (London)
282, 680686
72. Kornberg, A., and Baker, T. A. (1992) DNA
Replication, 2nd ed., Freeman, New York
73. Kaufmann, W. K., and Paules, R. S. (1996)
FASEB J. 10, 238247
74. Lindahl, T. (1993) Nature (London) 362, 709715
75. Rodley, G. A., and Bates, R. H. T. (1980) Trends
Biochem. Sci. 5, 231233
76. Kennard, O., and Salisbury, S. A. (1993) J. Biol.
Chem. 268, 1070110704
77. Drew, H. R., and Dickerson, R. E. (1981) J. Mol.
Biol. 151, 535556
78. Dickerson, R. E. (1983) Sci. Am. 249(Dec), 94
111
78a. Dickerson, R. E. (1997) in International Tables
for Macromolecular Crystallography (Rossmann,
M. G., and Arnold, E., eds), (Chapter 23, Part c)
79. Shui, X., McFail-Isom, L., Hu, G. G., and
Williams, L. D. (1998) Biochemistry 37, 8341
8355
80. Gorin, A. A., Zhurkin, V. B., and Olson, W. K.
(1995) J. Mol. Biol. 247, 3448
81. Tereshko, V., Minasov, G., and Egli, M. (1999)
J. Am. Chem. Soc. 121, 470471
82. Yuan, H., Quintana, J., and Dickerson, R. E.
(1992) Biochemistry 31, 80098021
83. Ban, C., Ramakrishnan, B., and Sundara-
lingam, M. (1994) J. Mol. Biol. 236, 275285
83a. Kielkopf, C. L., Ding, S., Kuhn, P., and Rees,
D. C. (2000) J. Mol. Biol. 296, 787801
84. Johansson, E., Parkinson, G., and Neidle, S.
(2000) J. Mol. Biol. 300, 551561
85. Shen, L. X., Cai, Z., and Tinoco, I., Jr. (1995)
FASEB J. 9, 10231033
86. Rosenberg, J. M., Seeman, N. C., Day, R. O.,
and Rich, A. (1976) J. Mol. Biol. 104, 145167
87. Marino, J. P., Gregorian, R. S., Jr.,
Csankovszki, G., and Crothers, D. M. (1995)
Science 268, 14481454
88. Iwamoto, S., and Hsu, M.-T. (1983) Nature
(London) 305, 7072
89. Morel, G., ed. (1995) Visualization of Nucleic
Acids, CRC Press, Boca Raton, Florida
90. Arscott, P. G., Lee, G., Bloomfield, V. A., and
Evans, D. F. (1989) Nature (London) 339,
484486
91. Jares-Erijman, E. A., and Jovin, T. M. (1996)
J. Mol. Biol. 257, 597617
92. Duan, Y., Wilkosz, P., Crowley, M., and
Rosenberg, J. M. (1997) J. Mol. Biol. 272, 553
572
93. Auffinger, P., and Westhof, E. (2000) J. Mol.
Biol. 300, 11131131
93a. Soler-Lpez, M., Malinina, L., Liu, J., Huynh-
Dinh, T., and Subirana, J. A. (1999) J. Biol.
Chem. 274, 2368323686
94. Schneider, B., and Berman, H. M. (1995)
Biophys. J. 69, 26612669
95. Tereshko, V., Minasov, G., and Egli, M. (1999)
J. Am. Chem. Soc. 121, 35903595
96. Franklin, R. E., and Gosling, R. G. (1953)
Nature (London) 172, 156157
97. Ramakrishnan, B., and Sundaralingam, M.
(1995) Biophys. J. 69, 553558
98. Ramakrishnan, B., and Sundaralingam, M.
(1993) J. Mol. Biol. 231, 431444
99. Eisenstein, M., and Shakked, Z. (1995) J. Mol.
Biol. 248, 662678
99a. Ng, H.-L., Kopka, M. L., and Dickerson, R. E.
(2000) Proc. Natl. Acad. Sci. U.S.A. 97, 2035
2039
100. Selsing, E., Wells, R. D., Alden, C. J., and
Arnott, S. (1979) J. Biol. Chem. 254, 54175422
101. Mahendrasingam, A., Forsyth, V. T., Hussain,
R., Greenall, R. J., Pigram, W. J., and Fuller, W.
(1986) Science 233, 195197
102. Hopkins, R. C. (1981) Science 211, 289291
103. Gupta, G., Bansal, M., and Sasisekharan, V.
(1980) Proc. Natl. Acad. Sci. U.S.A. 77, 64866490
104. Arnott, S., Chandrasekaran, R., Birdsall, D. L.,
Leslie, A. G. W., and Ratliff, R. L. (1980)
Nature (London) 283, 743745
105. Herbert, A., and Rich, A. (1996) J. Biol. Chem.
271, 1159511598
106. Gessner, R. V., Frederick, C. A., Quigley, G. J.,
Rich, A., and Wang, A. H.-J. (1989) J. Biol.
Chem. 264, 79217935
107. Zhou, G., and Ho, P. S. (1990) Biochemistry 29,
72297236
272 Chapter 5. The Nucleic Acids
176. Mascotti, D. P., and Lohman, T. M. (1990)
Proc. Natl. Acad. Sci. U.S.A. 87, 31423146
177. Li, A. Z., Huang, H., Re, X., Qi, L. J., and
Marx, K. A. (1998) Biophys. J. 74, 964973
178. Frydman, L., Rossomando, P. C., Frydman, V.,
Fernandez, C. O., Frydman, B., and Samejima,
K. (1992) Proc. Natl. Acad. Sci. U.S.A. 89,
91869190
179. Jain, S., Zon, G., and Sundaralingam, M.
(1989) Biochemistry 28, 23602364
180. Pelta, J., Durand, D., Doucet, J., and Livolant,
F. (1996) Biophys. J. 71, 4863
181. Rajski, S. R., Kumar, S., Roberts, R. J., and
Barton, J. K. (1999) J. Am. Chem. Soc. 121,
56155616
181a. Aich, P., Labiuk, S. L., Tari, L. W., Dalbaere, L.
J. T., Roesler, W. J., Falk, K. J., Steer, R. P., and
Lee, J. S. (1999) J. Mol. Biol. 294, 477485
182. Fink, H.-W., and Schnenberger, C. (1999)
Nature (London) 398, 407410
183. Marini, J. C., Miller, K. G., and Englund, P. T.
(1980) J. Biol. Chem. 255, 49764979
184. Wasserman, S. A., Dungan, J. M., and
Cozzarelli, N. R. (1985) Science 229, 171174
185. Du, S. M., Wang, H., Tse-Dinh, Y.-C., and
Seeman, N. C. (1995) Biochemistry 34, 673682
186. Du, S. M., Stollar, B. D., and Seeman, N. C.
(1995) J. Am. Chem. Soc. 117, 11941200
187. Wasserman, S. A., and Cozzarelli, N. R. (1986)
Science 232, 951960
188. White, J. H., and Cozzarelli, N. R. (1984) Proc.
Natl. Acad. Sci. U.S.A. 81, 33223326
189. Cipra, B. (1992) Science 255, 403
190. Adams, C. C. (1994) The Knot Book, Freeman,
New York
191. Krasnow, M. A., Stasiak, A., Spengler, S. J.,
Dean, F., Koller, T., and Cozzarelli, N. R.
(1983) Nature (London) 304, 559560
192. Du, S. M., and Seeman, N. C. (1992) J. Am.
Chem. Soc. 114, 96529655
193. Wang, J. C. (1980) Trends Biochem. Sci. 5,
219221
194. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Part 3, Freeman, San
Francisco, California (pp. 12651290)
195. Lebowitz, J. (1990) Trends Biochem. Sci. 15,
202207
196. Bates, A. D., and Maxwell, A. (1993) DNA
Topology, Oxford Univ. Press, London
197. Yang, Y., Westcott, T. P., Pedersen, S. C.,
Tobias, I., and Olson, W. K. (1995) Trends
Biochem. Sci. 20, 313319
198. Vinograd, J., Lebowitz, J., and Watson, R.
(1968) J. Mol. Biol. 33, 173197
199. White, J. H., Cozzarelli, N. R., and Bauer, W.
R. (1988) Science 241, 323327
199a. Vologodskii, A. V., and Cozzarelli, N. R.
(1994) Annu. Rev. Biophys. Biomol. Struct. 23,
609643
199b. Cozzarelli, N. R., Boles, T. C., and White, J. H.
(1990) in DNA Topology and its Biological
Effects, pp. 139184, Cold Spring Harbor Lab.
Press, Cold Spring Harbor, New York
199c. Podtelezhnikov, A. A., Cozzarelli, N. R., and
Vologodskii, A. V. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 1297412979
200. Reich, Z., Levin-Zaidman, S., Gutman, S. B.,
Arad, T., and Minsky, A. (1994) Biochemistry
33, 1417714184
201. Eriksson, M. A. L., Hrd, T., and Nilsson, L.
(1995) Biophys. J. 68, 402426
202. Keller, W. (1975) Proc. Natl. Acad. Sci. U.S.A.
72, 25502554
203. Kornberg, R. D., and Klug, A. (1981) Sci. Am.
244(Feb), 5264
204. Wang, B.-C., Rose, J., Arents, G., and
Moudrianakis, E. N. (1994) J. Mol. Biol. 236,
179188
References
108. Parkinson, G. N., Arvanitis, G. M., Lessinger,
L., Ginell, S. L., Jones, R., Gaffney, B., and
Berman, H. M. (1995) Biochemistry 34, 15487
15495
109. Sadasivan, C., and Gautham, N. (1995) J. Mol.
Biol. 248, 918 930
110. Bancroft, D., Williams, L. D., Rich, A., and
Egli, M. (1994) Biochemistry 33, 10731086
111. Rich, A., Nordheim, A., and Wang, A. H.-J.
(1984) Ann. Rev. Biochem. 53, 791846
112. Moore, M. H., Van Meervelt, L., Salisbury, S.
A., Lin, P. K. T., and Brown, D. M. (1995) J.
Mol. Biol. 251, 665673
113. Wang, Y., Thomas, G. A., and Peticolas, W. L.
(1987) Biochemistry 26, 51785186
114. Saenger, W., Hunter, W. N., and Kennard, O.
(1986) Nature (London) 324, 385388
115. Frank-Kamenetskii, M. (1986) Nature (London)
324, 305
116. Basham, B., Schroth, G. P., and Ho, P. S. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 64646468
117. Misra, V. K., and Honig, B. (1996) Biochemistry
35, 11151124
118. McLean, M. J., Blaho, J. A., Kilpatrick, M. W.,
and Wells, R. D. (1986) Proc. Natl. Acad. Sci.
U.S.A. 83, 58845888
119. Kim, J.-m, and DasSarma, S. (1996) J. Biol.
Chem. 271, 1972419731
120. Nordheim, A., Pardue, M. L., Lafer, E. M.,
Mller, A., Stollar, B. D., and Rich, A. (1981)
Nature (London) 294, 417422
121. Jaworski, A., Hsieh, W.-T., Blaho, J. A.,
Larson, J. E., and Wells, R. D. (1987) Science
238, 773777
122. Zacharias, W., OConner, T. R., and Larson, J.
E. (1988) Biochemistry 27, 29702978
123. Nordheim, A., and Rich, A. (1983) Nature
(London) 303, 674679
124. Wells, R. D. (1988) J. Biol. Chem. 263, 10951098
125. Rentzeperis, D., Kupke, D. W., and Marky, L.
A. (1994) Biochemistry 33, 95889591
126. Liu, K., Miles, H. T., Frazier, J., and Sasisekha-
ran, V. (1993) Biochemistry 32, 1180211809
127. Zhou, N., Germann, M. W., van de Sande, J.
H., Pattabiraman, N., and Vogel, H. J. (1993)
Biochemistry 32, 646656
128. Otto, C., Thomas, G. A., Rippe, K., Jovin, T.
M., and Peticolas, W. L. (1991) Biochemistry 30,
30623069
129. Trifonov, E. N. (1991) Trends Biochem. Sci. 16,
467470
130. Rich, A. (1998) Proc. Natl. Acad. Sci. U.S.A. 95,
1399914000
131. Hageman, P. J. (1988) Ann. Rev. Biophys.
Biophys. Chem. 17, 265286
132. Poncin, M., Hartmann, B., and Lavery, R.
(1992) J. Mol. Biol. 226, 775794
133. MacKerell, A. D., Jr., Wirkiewicz-Kuczera, J.,
and Karplus, M. (1995) J. Am. Chem. Soc. 117,
1194611975
134. Kosikov, K. M., Gorin, A. A., Zhurkin, V. B.,
and Olson, W. K. (1999) J. Mol. Biol. 289,
13011326
135. Peticolas, W. L., Wang, Y., and Thomas, G. A.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 2579
2583
136. Dickerson, R. E., and Drew, H. R. (1981) Proc.
Natl. Acad. Sci. U.S.A. 78, 73187322
136a. Packer, M. J., and Hunter, C. A. (1998) J. Mol.
Biol. 280, 407420
137. Hogan, M., Dattagupta, N., and Crothers, D.
M. (1979) Nature (London) 278, 521524
138. Mitra, C. K., Sarma, M. H., and Sarma, R. H.
(1981) J. Am. Chem. Soc. 103, 67276737
139. Cluzel, P., Lebrun, A., Heller, C., Lavery, R.,
Viovy, J.-L., Chatenay, D., and Caron, F. (1996)
Science 271, 792794
140. Smith, S. B., Cui, Y., and Bustamante, C.
(1996) Science 271, 795799
141. Strick, T. R., Allemand, J. F., Bensimon, D.,
Bensimon, A., and Croquette, V. (1996) Science
271, 18351837
141a. Allemand, J. F., Bensimon, D., Lavery, R., and
Croquette, V. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 1415214157
142. Dickerson, R. E. (1983) J. Mol. Biol. 166, 419
441
143. Tung, C.-S., and Harvey, S. C. (1986) J. Biol.
Chem. 261, 37003709
144. Ulanovsky, L. E., and Trifonov, E. N. (1987)
Nature (London) 326, 720722
145. Diekmann, S. (1989) EMBO J. 8, 14
146. El Hassan, M. A., and Calladine, C. R. (1996)
J. Mol. Biol. 259, 95103
147. Calladine, C. R. (1982) J. Mol. Biol. 161, 343352
148. Hunter, C. A., and Lu, X.-J. (1997) J. Mol. Biol.
265, 603619
149. Babcock, M. S., Pednault, E. P. D., and Olson,
W. K. (1994) J. Mol. Biol. 273, 125156
150. Lu, X.-J., El Hassan, M. A., and Hunter, C. A.
(1997) J. Mol. Biol. 273, 668680
151. Kitchin, P. A., Klein, V. A., Ryan, K. A., Gann,
K. L., Rauch, C. A., Kang, D. S., Wells, R. D.,
and England, P. T. (1986) J. Biol. Chem. 261,
1130211309
152. Koo, H.-S., Wu, H.-M., and Crothers, D. M.
(1986) Nature (London) 320, 501506
153. Zahn, K., and Blattner, F. B. (1987) Science 236,
416422
154. Snyder, M., Buchman, A. R., and Davis, R. W.
(1986) Nature (London) 324, 8789
155. Travers, A. A. (1987) Trends Biochem. Sci. 12,
108112
156. Grzeskowiak, K., Goodsell, D. S., Kaczor-
Grzeskowiak, M., Cascio, D., and Dickerson,
R. E. (1993) Biochemistry 32, 89238931
157. Allewell, N. (1988) Trends Biochem. Sci. 13,
193195
158. Haran, T. E., Kahn, J. D., and Crothers, D. M.
(1994) J. Mol. Biol. 244, 135143
159. Strauss, J. K., and Maher, L. J., III. (1994)
Science 266, 18291834
160. Olson, W. K., Marky, N. L., Jernigan, R. L.,
and Zhurkin, V. B. (1993) J. Mol. Biol. 232,
530554
161. Price, M. A., and Tullius, T. D. (1993)
Biochemistry 32, 127136
162. DiGabriele, A. D., and Steitz, T. A. (1993) J.
Mol. Biol. 231, 10241039
163. Chuprina, V. P., Fedoroff, O. Y., and Reid, B.
R. (1991) Biochemistry 30, 561568
164. Crothers, D. M. (1994) Science 266, 18191820
165. Husain, I., Griffith, J., and Sancar, A. (1988)
Proc. Natl. Acad. Sci. U.S.A. 85, 25582562
166. Srinivasan, A. R., Torres, R., Clark, W., and
Olson, W. K. (1987) J. Biomol. Struct. Dyn. 5,
459496
167. Lutter, L. C., Halvorson, H. R., and Calladine,
C. R. (1996) J. Mol. Biol. 261, 620633
168. Priv, G. G., Heinemann, V., Chandrasegaran,
S., Kan, L.-S., Kopka, M. L., and Dickerson, R.
E. (1987) Science 238, 498504
169. Lilley, D. M. J. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 71407142
170. Woodson, S. A., and Crothers, D. M. (1988)
Biochemistry 27, 31303141
171. Miller, M., Harrison, R. W., Wlodawer, A.,
Appella, E., and Sussman, J. L. (1988) Nature
(London) 334, 8586
172. Hunter, W. N., Brown, T., Kneale, G., Anand,
N. N., Rabinovich, D., and Kennard, O. (1987)
J. Biol. Chem. 262, 99629970
173. Lane, A., Martin, S. R., Ebel, S., and Brown, T.
(1992) Biochemistry 31, 1208712095
174. Li, Y., and Agrawal, S. (1995) Biochemistry 34,
1005610062
175. Manning, G. S. (1978) Q. Rev. Biophys. 11,
179246
273
References
205. Malik, M., Bensaid, A., Rouviere-Yaniv, J., and
Drlica, K. (1996) J. Mol. Biol. 256, 6676
206. Serrano, M., Salas, M., and Hermoso, J. M.
(1993) Trends Biochem. Sci. 18, 202206
207. Seidl, A., and Hinz, H.-J. (1984) Proc. Natl.
Acad. Sci. U.S.A. 81, 13121316
208. Rahmouni, A. R., and Wells, R. D. (1989)
Science 246, 358363
209. Spolar, R. S., and Record, M. T., Jr. (1994)
Science 263, 777784
210. White, J. H., Gallo, R. M., and Bauer, W. R.
(1992) Trends Biochem. Sci. 17, 712
211. van Holde, K., and Zlatanova, J. (1995) J. Biol.
Chem. 270, 83738376
212. Keller, W. (1975) Proc. Natl. Acad. Sci. U.S.A.
72, 48764880
213. Hamiche, A., Schultz, P., Ramakrishnan, V.,
Oudet, P., and Prunell, A. (1996) J. Mol. Biol.
257, 3042
214. Alfonso, P. J., Crippa, M. P., Hayes, J. J., and
Bustin, M. (1994) J. Mol. Biol. 236, 189198
215. Bednar, J., Furrer, P., Stasiak, A., Dubochet, J.,
Egelman, E. H., and Bates, A. D. (1994) J. Mol.
Biol. 235, 825847
216. Kumar, A. (1995) Biochemistry 34, 1292112925
217. Stigter, D. (1995) Biophys. J. 69, 380388
218. Baeza, I., Gariglio, P., Rangel, L. M., Chavez,
P., Cervantes, L., Arguello, C., Wong, C., and
Montaez, C. (1987) Biochemistry 26, 6387
6392
219. Schmid, M. B. (1988) Trends Biochem. Sci. 13,
131135
220. Quigley, G. J., Wang, A. H.-J., Ughetto, G., van
der Marel, G., van Boom, J. H., and Rich, A.
(1980) Proc. Natl. Acad. Sci. U.S.A. 77, 7204
7208
221. Campbell, A. M., and Jolly, D. J. (1973)
Biochem. J. 133, 209226
222. Depew, R. E., and Wang, J. C. (1975) Proc.
Natl. Acad. Sci. U.S.A. 72, 42754279
223. Westof, E., and Sundaralingam, M. (1980)
Proc. Natl. Acad. Sci. U.S.A. 77, 18521856
224. Sobell, H. M. (1974) Sci. Am. 231(Aug), 8291
225. Chen, H., and Patel, D. J. (1995) J. Am. Chem.
Soc. 117, 59015913
226. Chaires, J. B., Satyanarayana, S., Suh, D., Fokt,
I., Przewloka, T., and Priebe, W. (1996)
Biochemistry 35, 20472053
227. Rajeswari, M. R., Bose, H. S., Kukreti, S.,
Gupta, A., Chauhan, V. S., and Roy, K. B.
(1992) Biochemistry 31, 62376241
227a. Leitner, D., Schrder, W., and Weisz, K. (2000)
Biochemistry 39, 5886 5892
228. Felsenfeld, G., Davies, D., and Rich, A. (1957)
J. Am. Chem. Soc. 79, 20232024
229. Felsenfeld, G., and Miles, H. T. (1967) Ann.
Rev. Biochem. 36, 407448
230. Arnott, S., and Selsing, E. (1974) J. Mol. Biol.
88, 509521
231. Wang, E., Koshlap, K. M., Gillespie, P.,
Dervan, P. B., and Feigon, J. (1996) J. Mol. Biol.
257, 10521069
232. Radhakrishnan, I., and Patel, D. J. (1994)
Biochemistry 33, 1140511416
233. Radhakrishnan, I., and Patel, D. J. (1994) J.
Mol. Biol. 241, 600619
234. Rhee, S., Han, Z.-j, Liu, K., Miles, H. T., and
Davies, D. R. (1999) Biochemistry 38, 16810
16815
235. Roberts, R. W., and Crothers, D. M. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 43204325
236. van Dongen, M. J. P., Heus, H. A., Wymenga,
S. S., van der Marel, G. A., van Boom, J. H.,
and Hilbers, C. W. (1996) Biochemistry 35,
17331739
237. Klysik, J. (1995) J. Mol. Biol. 245, 499507
238. Srinivasan, A. R., and Olson, W. K. (1998)
J. Am. Chem. Soc. 120, 484491
239. Hanvey, J. C., Shimizu, M., and Wells, R. D.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 6292
6296
240. Htun, H., and Dahlberg, J. E. (1989) Science
243, 15711576
241. Mirkin, S. M., and Frank-Kamenetskii, M. D.
(1994) Annu. Rev. Biophys. Biomol. Struct. 23,
541576
242. Singh, S., Patel, P. K., and Hosur, R. V. (1997)
Biochemistry 36, 1321413222
243. Van Meervelt, L., Vlieghe, D., Dautant, A.,
Gallois, B., Prcigoux, G., and Kennard, O.
(1995) Nature (London) 374, 742744
244. Reddy, G., Jwang, B., Rao, B. J., and Radding,
C. M. (1994) Biochemistry 33, 1148611492
245. Kim, M. G., Zhurkin, V. B., Jernigan, R. L.,
and Camerini-Otero, R. D. (1995) J. Mol. Biol.
247, 874889
246. Cox, M. M. (1995) J. Biol. Chem. 270, 26021
26024
247. Jain, S. K., Cox, M. M., and Inman, R. B. (1995)
J. Biol. Chem. 270, 49434949
248. Best, G. C., and Dervan, P. B. (1995) J. Am.
Chem. Soc. 117, 11871193
249. Greenberg, W. A., and Dervan, P. B. (1995)
J. Am. Chem. Soc. 117, 50165022
250. Hashem, G. M., Pham, L. P., Vaughan, M. R.,
and Gray, D. M. (1998) Biochemistry 37, 6172
251. Gryaznov, S. M., Lloyd, D. H., Chen, J.-K.,
Schultz, R. G., DeDionisio, L. A., Ratmeyer, L.,
and Wilson, W. D. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 57985802
252. Betts, L., Josey, J. A., Veal, J. M., and Jordan, S.
R. (1995) Science 270, 18381841
253. Tomac, S., Sarkar, M., Ratilainen, T., Wittung,
P., Nielsen, P. E., Nordn, B., and Grslund, A.
(1996) J. Am. Chem. Soc. 118, 55445552
253a. Ray, A., and Nordn, B. (2000) FASEB J. 14,
1041 1060
253b. Izvolsky, K. I., Demidov, V. V., Nielsen, P. E.,
and Frank-Kamenetskii, M. D. (2000)
Biochemistry 39, 10908 10913
254. White, S., Szewczyk, J. W., Turner, J. M.,
Baird, E. E., and Dervan, P. B. (1998) Nature
(London) 391, 468471
255. Marsh, T. C., and Henderson, E. (1994)
Biochemistry 33, 1071810724
256. Blackburn, E. H. (1991) Nature (London) 350,
569573
257. Blackburn, E. H. (1991) Trends Biochem. Sci. 16,
378381
258. Moyzis, R. K. (1991) Sci. Am. 265(Aug), 4855
259. Henderson, E., Hardin, C. C., Wolk, S. K.,
Tinoco, I., Jr., and Blackburn, E. (1987) Cell 51,
899908
260. Laughlan, G., Murchie, A. I. H., Norman, D.
G., Moore, M. H., Moody, P. C. E., Lilley, D.
M. J., and Luisi, B. (1994) Science 265, 520527
261. Kang, C. H., Zhang, X., Ratliff, R., Moyzis, R.,
and Rich, A. (1992) Nature (London) 356, 126
131
262. Smith, F. W., and Feigon, J. (1993) Biochemistry
32, 86828692
263. Balagurumoorthy, P., and Brahmachari, S. K.
(1994) J. Biol. Chem. 269, 2185821869
264. Williamson, J. R. (1994) Annu. Rev. Biophys.
Biomol. Struct. 23, 703730
265. Frank-Kamenetskii, M. (1992) Nature (London)
356, 105
266. Wang, K. Y., Swaminathan, S., and Bolton, P.
H. (1994) Biochemistry 33, 75177527
267. Wang, Y., and Patel, D. J. (1995) J. Mol. Biol.
251, 7694
268. Laporte, L., and Thomas, G. J., Jr. (1998)
Biochemistry 37, 13271335
269. Ross, W. S., and Hardin, C. C. (1994) J. Am.
Chem. Soc. 116, 60706080
270. Sen, D., and Gilbert, W. (1990) Nature (London)
344, 410414
271. Miura, T., Benevides, J. M., and Thomas, G. J.,
Jr. (1995) J. Mol. Biol. 248, 233238
272. Schierer, T., and Henderson, E. (1994)
Biochemistry 33, 22402246
273. Hardin, C. C., Corregan, M., Brown, B. A., II,
and Frederick, L. N. (1993) Biochemistry 32,
58705880
274. Cheong, C., and Moore, P. B. (1992)
Biochemistry 31, 84068414
275. Chen, F.-M. (1995) J. Biol. Chem. 270, 23090
23096
276. Kettani, A., Kumar, R. A., and Patel, D. J.
(1995) J. Mol. Biol. 254, 638656
277. Kang, C. H., Berger, I., Lockshin, C., Ratliff,
R., Moyzis, R., and Rich, A. (1994) Proc. Natl.
Acad. Sci. U.S.A. 91, 1163611640
278. Gehring, K., Leroy, J.-L., and Guron, M.
(1993) Nature (London) 363, 561565
279. Lacroix, L., Mergny, J.-L., Leroy, J.-L., and
Hlne, C. (1996) Biochemistry 35, 87158722
280. Chen, L., Cai, L., Zhang, X., and Rich, A.
(1994) Biochemistry 33, 1354013546
281. Kang, C. H., Berger, I., Lockshin, C., Ratliff,
R., Moyzis, R., and Rich, A. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 38743878
282. Benevides, J. M., Kang, C., and Thomas, G. J.,
Jr. (1996) Biochemistry 35, 57475755
283. Gallego, J., Chou, S.-H., and Reid, B. R. (1997)
J. Mol. Biol. 273, 840856
284. Selsing, E., Wells, R. D., Alden, C. J., and
Arnott, S. (1979) J. Biol. Chem. 254, 54175422
285. Nishizaki, T., Iwai, S., Ohkubo, T., Kojima, C.,
Nakamura, H., Kyogoku, Y., and Ohtsuka, E.
(1996) Biochemistry 35, 40164025
286. Salazar, M., Champoux, J. J., and Reid, B. R.
(1993) Biochemistry 32, 739744
287. Seeman, N. C., and Kallenbach, N. R. (1994)
Annu. Rev. Biophys. Biomol. Struct. 23, 5386
288. Zhang, S., Fu, T.-J., and Seeman, N. C. (1993)
Biochemistry 32, 80628067
289. Pikkemaat, J. A., van den Elst, H., van Boom,
J. H., and Altona, C. (1994) Biochemistry 33,
1489614907
290. Carlstrm, G., and Chazin, W. J. (1996)
Biochemistry 35, 35343544
291. Holliday, R. (1964) Genet. Res. Camb. 5, 282304
292. Bennett, R. J., and West, S. C. (1995) J. Mol.
Biol. 252, 213226
293. Grainger, R. J., Murchie, A. I. H., and Lilley, D.
M. J. (1998) Biochemistry 37, 2332
294. Lilley, D. M. J. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 95139515
295. Panyutin, I. G., Biswas, I., and Hsieh, P. (1995)
EMBO J. 14, 18191826
296. Chen, J., and Seeman, N. C. (1991) Nature
(London) 350, 631633
297. Zhang, Y., and Seeman, N. C. (1994) J. Am.
Chem. Soc. 116, 16611669
297a. Liu, F., Sha, R., and Seeman, N. C. (1999) J. Am.
Chem. Soc. 121, 917922
297b. Fahlman, R. P., and Sen, D. (1999) J. Am. Chem.
Soc. 121, 1107911085
298. Feagin, J. E. (1990) J. Biol. Chem. 265, 19373
19376
299. Stuart, K. (1991) Trends Biochem. Sci. 16, 6872
300. Simons, R. W., and Grunberg-Manago, M.,
eds. (1997) RNA Structure and Function, Cold
Spring Harbor Lab. Press, Cold Spring
Harbor, New York
301. Nicoloso, M., Qu, L.-H., Michot, B., and
Bachellerie, J.-P. (1996) J. Mol. Biol. 260, 178195
302. Gold, L., Polisky, B., Uhlenbeck, O., and Yarus,
M. (1995) Ann. Rev. Biochem. 64, 763797
303. Draper, D. E. (1996) Trends Biochem. Sci. 21,
145149
304. Sich, C., Ohlenschlger, O., Ramachandran,
R., Grlach, M., and Brown, L. R. (1997)
Biochemistry 36, 1398914002
274 Chapter 5. The Nucleic Acids
370. Patel, D. J., Suri, A. K., Jiang, F., Jiang, L., Fan,
P., Kumar, R. A., and Nonin, S. (1997) J. Mol.
Biol. 272, 645664
371. Crick, F. H. C. (1966) Cold Spring Harbor Symp.
Quant. Biol. 31, 39
372. Cornish-Bowden, A. (1996) Trends Biochem.
Sci. 21, 155
373. Hall, B. D. (1979) Nature (London) 282, 129130
374. Barrell, B. G., Bankier, A. T., and Drouin, J.
(1979) Nature (London) 282, 189194
375. Horowitz, S., and Gorovsky, M. A. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 24522455
376. Chambers, I., and Harrison, P. R. (1987) Trends
Biochem. Sci. 12, 255256
377. Gardner, M. (1970) Sci. Am. 223(Aug), 110112
378. Mirkin, S. M., Lyamichev, V. I., Drushlyak, K.
N., Dobrynin, V. N., Filippov, S. A., and
Frank-Kamenetskii, M. D. (1987) Nature
(London) 330, 495497
379. Guieysse, A.-L., Praseuth, D., and Hlne, C.
(1997) J. Mol. Biol. 267, 289298
380. Panayotatos, N., and Fontaine, A. (1987) J.
Biol. Chem. 262, 1136411368
381. Pabo, C. O., and Sauer, R. T. (1984) Ann. Rev.
Biochem. 53, 293321
382. Berg, O. G., and von Hippel, P. H. (1988)
Trends Biochem. Sci. 13, 207211
383. Marmorstein, R. Q., Joachimiak, A., Sprinzl,
M., and Sigler, P. B. (1987) J. Biol. Chem. 262,
49224927
384. Muto, A., and Osawa, S. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 166169
385. Singer, C. E., and Ames, B. N. (1979) Science
170, 822826
386. Singer, C. E., and Ames, B. N. (1972) Science
175, 1391
387. Harrison, S. C. (1991) Nature (London) 353,
715719
388. Churchill, M. E. A., and Travers, A. A. (1991)
Trends Biochem. Sci. 16, 9297
389. Travers, A. (1993) DNA-Protein Interactions,
Chapman & Hall, New York
390. Jones, S., van Heyningen, P., Berman, H. M.,
and Thornton, J. M. (1999) J. Mol. Biol. 287,
877896
391. Arents, G., and Moudrianakis, E. N. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 1117011174
392. Starich, M. R., Sandman, K., Reeve, J. N., and
Summers, M. F. (1996) J. Mol. Biol. 255, 187203
393. Hud, N. V., Milanovich, F. P., and Balhorn, R.
(1994) Biochemistry 33, 75287535
394. Lohman, T. M., and Ferrari, M. E. (1994) Ann.
Rev. Biochem. 63, 527570
395. Overman, L. B., and Lohman, T. M. (1994) J.
Mol. Biol. 236, 165178
396. Folmer, R. H. A., Nilges, M., Folkers, P. J. M.,
Konings, R. N. H., and Hilbers, C. W. (1994) J.
Mol. Biol. 240, 341357
397. Olah, G. A., Gray, D. M., Gray, C. W., Kergil,
D. L., Sosnick, T. R., Mark, B. L., Vaughan, M.
R., and Trewhella, J. (1995) J. Mol. Biol. 249,
576594
398. Kanellopoulos, P. N., Tsernoglou, D., van der
Vliet, P. C., and Tucker, P. A. (1996) J. Mol. Biol.
257, 18
399. Bogdarina, I., Fox, D. G., and Kneale, G. G.
(1998) J. Mol. Biol. 275, 443452
400. Pluta, A. F., Mackay, A. M., Ainsztein, A. M.,
Goldberg, I. G., and Earnshaw, W. C. (1995)
Science 270, 15911594
401. Schevitz, R. W., Otwinowski, Z., Joachimiak,
A., Lawson, C. L., and Sigler, P. B. (1985)
Nature (London) 317, 782786
402. Zhang, R.-G, Joachimiak, A., Lawson, C. L.,
Schevitz, R. W., Otwinowski, Z., and Sigler, P.
B. (1987) Nature (London) 327, 591597
References
305. Egli, M., Portmann, S., and Usman, N. (1996)
Biochemistry 35, 84898494
306. Auffinger, P., and Westhof, E. (1997) J. Mol.
Biol. 274, 5463
307. Salazar, M., Fedoroff, O. Y., Miller, J. M.,
Ribeiro, N. S., and Reid, B. R. (1993)
Biochemistry 32, 42074215
308. Fedoroff, O. Y., Salazar, M., and Reid, B. R.
(1993) J. Mol. Biol. 233, 509523
309. Ratmeyer, L., Vinayak, R., Zhong, Y. Y., Zon,
G., and Wilson, W. D. (1994) Biochemistry 33,
52985304
310. Varani, G., Cheong, C., and Tinoco, I., Jr.
(1991) Biochemistry 30, 32803289
311. Butcher, S. E., Dieckmann, T., and Feigon, J.
(1997) J. Mol. Biol. 268, 348358
312. van Dongen, M. J. P., Wijmenga, S. S., van der
Marel, G. A., van Boom, J. H., and Hilbers, C.
W. (1996) J. Mol. Biol. 263, 715729
313. SantaLucia, J., Jr., Kierzek, R., and Turner, D.
H. (1992) Science 256, 217219
314. Murphy, F. L., and Cech, T. R. (1994) J. Mol.
Biol. 236, 4963
315. Jucker, F. M., Heus, H. A., Yip, P. F., Moors, E.
H. M., and Pardi, A. (1996) J. Mol. Biol. 264,
968980
315a. Gutell, R. R., Cannone, J. J., Konings, D., and
Gautheret, D. (2000) J. Mol. Biol. 300, 791803
316. Fountain, M. A., Serra, M. J., Krugh, T. R.,
and Turner, D. H. (1996) Biochemistry 35,
65396548
317. Huang, S., Wang, Y.-X., and Draper, D. E.
(1996) J. Mol. Biol. 258, 308321
318. Nowakowski, J., and Tinoco, I., Jr. (1996)
Biochemistry 35, 25772585
319. Chang, K.-Y., and Tinoco, I., Jr. (1997) J. Mol.
Biol. 269, 5266
320. Rosendahl, G., Hansen, L. H., and
Douthwaite, S. (1995) J. Mol. Biol. 249, 5968
321. Pleij, C. W. A. (1990) Trends Biochem. Sci. 15,
143147
322. ten Dam, E., Pleij, K., and Draper, D. (1992)
Biochemistry 31, 1166511676
323. Shen, L. X., and Tinoco, I., Jr. (1995) J. Mol.
Biol. 247, 963978
324. Pinard, R., Payant, C., and Brakier-Gingras, L.
(1995) Biochemistry 34, 96119616
325. Puglisi, J. D., Wyatt, J. R., and Tinoco, I., Jr.
(1988) Nature (London) 331, 283286
326. Qiu, H., Kaluarachchi, K., Du, Z., Hoffman,
D. W., and Giedroc, D. P. (1996) Biochemistry
35, 41764186
326a. Kolk, M. H., van der Graaf, M., Wijmenga, S.
S., Pleij, C. W. A., Heus, H. A., and Hilbers, C.
W. (1998) Science 280, 434438
327. Shi, P.-Y., Brinton, M. A., Veal, J. M., Zhong, Y.
Y., and Wilson, W. D. (1996) Biochemistry 35,
42224230
328. Hubbard, J. M., and Hearst, J. E. (1991)
Biochemistry 30, 54585465
329. Soll, D., and Abelson, J., eds. (1979) Transfer
RNA, Cold Spring Harbor Lab. Press, Cold
Spring Harbor, New York
330. Zimmermann, R. A. (1996) Science 271, 1240
1241
331. Steinberg, S., Gautheret, D., and Cedergren,
R. (1994) J. Mol. Biol. 236, 982989
332. Holbrook, S. R., Sussman, J. L., Warrant, R.
W., and Kim, S.-H. (1978) J. Mol. Biol. 123,
631660
333. Rich, A., and Kim, S. H. (1978) Sci. Am. 238
(Jan), 5262
334. Rich, A. (1978) Trends Biochem. Sci. 3, 3437
335. Moras, D., Comarmond, M. B., Fischer, J.,
Weiss, R., Thierry, J. C., Ebel, J. P., and Gieg,
R. (1980) Nature (London) 288, 669674
336. Woo, N. H., Roe, B. A., and Rich, A. (1980)
Nature (London) 286, 346351
337. Wakao, H., Romby, P., Westhof, E., Laalami,
S., Grunberg-Manago, M., Ebel, J.-P.,
Ehresmann, C., and Ehresmann, B. (1989) J.
Biol. Chem. 264, 2036320371
338. Hou, Y.-M., and Schimmel, P. (1992)
Biochemistry 31, 41574160
339. Leehey, M. A., Squassoni, C. A., Friederich, M.
W., Mills, J. B., and Hagerman, P. J. (1995)
Biochemistry 34, 1623516239
340. Quigley, G. J., and Rich, A. (1976) Science 194,
796806
341. Brimacombe, R., Atmadja, J., Stiege, W., and
Schler, D. (1988) J. Mol. Biol. 199, 115136
342. Mueller, F., and Brimacombe, R. (1997) J. Mol.
Biol. 271, 524544
343. Turner, D. H., and Sugimoto, N. (1988) Ann.
Rev. Biophys. Biophys. Chem. 17, 167192
344. Brimacombe, R. (1988) Biochemistry 27, 4207
4213
345. Schnare, M. N., Damberger, S. H., Gray, M.
W., and Gutell, R. R. (1996) J. Mol. Biol. 256,
701719
346. Yonath, A. (1984) Trends Biochem. Sci. 9, 227
230
347. Pennisi, E. (1999) Science 285, 20482051
348. Svergun, D. I., Koch, M. H. J., Pedersen, J. S.,
and Serdyuk, I. N. (1994) J. Mol. Biol. 240,
7886
349. Vladimirov, S. N., Druzina, Z., Wang, R., and
Cooperman, B. S. (2000) Biochemistry 39, 183
193
350. Gregory, S. T., and Dahlberg, A. E. (1999) J.
Mol. Biol. 285, 14751483
351. Ban, N., Nissen, P., Hansen, J., Moore, P. B.,
and Steitz, T. A. (2000) Science 289, 905920
352. Cate, J. H., Yusupov, M. M., Yusupova, G. Z.,
Earnest, T. N., and Noller, H. F. (1999) Science
285, 20952104
353. Culver, G. M., Cate, J. H., Yusupova, G. Z.,
Yusupov, M. M., and Noller, H. F. (1999)
Science 285, 21332135
354. Carter, A. P., Clemons, W. M., Brodersen, D.
E., Morgan-Warren, R. J., Wimberly, B. T., and
Ramakrishnan, V. (2000) Nature (London) 407,
340348
355. Nissen, P., Hansen, J., Ban, N., Moore, P. B.,
and Steitz, T. A. (2000) Science 289, 920930
356. Muth, G. W., Ortoleva-Donnelly, L., and
Strobel, S. A. (2000) Science 289, 947950
357. Cech, T. R. (2000) Science 289, 878879
358. Pennisi, E. (1996) Science 273, 574575
359. OGara, M., Klimasauskas, S., Roberts, R. J.,
and Cheng, X. (1996) J. Mol. Biol. 261, 634645
360. Gommers-Ampt, J. H., and Borst, P. (1995)
FASEB J. 9, 10341042
361. Maltman, K. L., Neuhard, J., and Warren, R.
A. J. (1981) Biochemistry 20, 35863591
362. Lipscomb, L. A., Peek, M. E., Morningstar, M.
L., Verghis, S. M., Miller, E. M., Rich, A.,
Essigmann, J. M., and Williams, L. D. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 719723
363. Leonard, G. A., McAuley-Hecht, K. E., Gibson,
N. J., Brown, T., Watson, W. P., and Hunter, W.
N. (1994) Biochemistry 33, 47554761
364. Wrzesinski, J., Bakin, A., Nurse, K., Lane, B.
G., and Ofengand, J. (1995) Biochemistry 34,
89048913
365. Ellington, A. D., and Szostak, J. W. (1990)
Nature (London) 346, 818822
366. Fan, P., Suri, A. K., Fiala, R., Live, D., and
Patel, D. J. (1996) J. Mol. Biol. 258, 480500
367. Jenison, R. D., Gill, S. C., Pardi, A., and
Polisky, B. (1994) Science 263, 14251429
368. Hamasaki, K., Killian, J., Cho, J., and Rando,
R. R. (1998) Biochemistry 37, 656663
369. Connell, G. J., Illangesekare, M., and Yarus,
M. (1993) Biochemistry 32, 54975502
275
References
403. Otwinowski, Z., Schevitz, R. W., Zhang, R.-G.,
Lawson, C. L., Joachimiak, A., Marmorstein,
R. Q., Luisi, B. F., and Sigler, P. B. (1988)
Nature (London) 335, 321329
404. Shakked, Z., Guzikevich Guerstein, G.,
Frolow, F., Rabinovich, D., Joachimiak, A.,
and Sigler, P. B. (1994) Nature (London) 368,
469473
405. Lawson, C. L., and Carey, J. (1993) Nature
(London) 366, 178182
406. Lawson, C. L., and Sigler, P. B. (1988) Nature
(London) 333, 869871
407. Guenot, J., Fletterick, R. J., and Kollman, P. A.
(1994) Protein Sci. 3, 12761285
408. Rastinejad, F., Artz, P., and Lu, P. (1993) J. Mol.
Biol. 233, 389399
409. Markiewicz, P., Kleina, L. G., Cruz, C., Ehret,
S., and Miller, J. H. (1994) J. Mol. Biol. 240,
421433
410. Lewis, M., Chang, G., Horton, N. C., Kercher,
M. A., Pace, H. C., Schumacher, M. A.,
Brennan, R. G., and Lu, P. (1996) Science 271,
12471254
411. Brennan, R. G., and Matthews, B. W. (1989) J.
Biol. Chem. 264, 19031906
412. Jordan, S. R., and Pabo, C. O. (1988) Science
242, 893899
413. Wolberger, C., Dong, Y., Ptashne, M., and
Harrison, S. C. (1988) Nature (London) 335,
789795
414. Aggarwal, A. K., Rodgers, D. W., Drottar, M.,
Ptashne, M., and Harrison, S. C. (1988) Science
242, 899907
415. Bell, A. C., and Koudelka, G. B. (1993) J. Mol.
Biol. 234, 542553
416. Mandel-Gutfreund, Y., Schueler, O., and
Margalit, H. (1995) J. Mol. Biol. 253, 370382
417. Ferr-DAmar, A. R., Prendergast, G. C., Ziff,
E. B., and Burley, S. K. (1993) Nature (London)
363, 3844
418. Junius, F. K., ODonoghue, S. I., Nilges, M.,
Weiss, A. S., and King, G. F. (1996) J. Biol.
Chem. 271, 1366313667
419. Keller, W., Knig, P., and Richmond, T. J.
(1995) J. Mol. Biol. 254, 657667
420. Harbury, P. B., Kim, P. S., and Alber, T. (1994)
Nature (London) 371, 8083
421. Klug, A., and Schwabe, J. W. R. (1995) FASEB J.
9, 597604
422. Rhodes, D., and Klug, A. (1993) Sci. Am. 268
(Feb), 5665
423. Pavletich, N. P., and Pabo, C. O. (1991) Science
252, 809817
424. Mackay, J. P., and Crossley, M. (1998) Trends
Biochem. Sci. 23, 14
425. Miller, J., McLachlan, A. D., and Klug, A.
(1985) EMBO J. 4, 16091614
426. Pieler, T., and Theunissen, O. (1993) Trends
Biochem. Sci. 18, 226230
427. Pavletich, N. P., and Pabo, C. O. (1993) Science
261, 17011707
428. Krizek, B. A., Amann, B. T., Kilfoil, V. J.,
Merkle, D. L., and Berg, J. M. (1991) J. Am.
Chem. Soc. 113, 45184523
429. Somers, W. S., and Phillips, S. E. V. (1992)
Nature (London) 359, 387393
430. Phillips, S. E. V. (1994) Annu. Rev. Biophys.
Biomol. Struct. 23, 671701
431. Breg, J. N., van Opheusden, H. J., Burgering,
M. J. M., Boelens, R., and Kaptein, R. (1990)
Nature (London) 346, 586589
432. Bonnefoy, E., Takahashi, M., and Yaniv, J. R.
(1994) J. Mol. Biol. 242, 116129
433. Mller, C. W., Rey, F. A., Sodeoka, M.,
Verdine, G. L., and Harrison, S. C. (1995)
Nature (London) 373, 311317
434. Ghosh, G., Van Duyne, G., Ghosh, S., and
Sigler, P. B. (1995) Nature (London) 373, 303310
435. Baltimore, D., and Beg, A. A. (1995) Nature
(London) 373, 287288
436. Kaufmann, E., Mller, D., and Knchel, W.
(1995) J. Mol. Biol. 248, 239254
437. Burd, C. G., and Dreyfuss, G. (1994) Science
265, 615621
438. Nagai, K., and Mattaj, I. W., eds. (1994) RNA-
Protein Interactions, IRL Press, Oxford
439. Draper, D. E. (1995) Ann. Rev. Biochem. 64,
593620
440. Hentze, M. W. (1994) Trends Biochem. Sci. 19,
101103
441. Allain, F. H.-T., Gubser, C. C., Howe, P. W. A.,
Nagai, K., Neuhaus, D., and Varani, G. (1996)
Nature (London) 380, 646650
442. Kenan, D. J., Query, C. C., and Keene, J. D.
(1991) Trends Biochem. Sci. 16, 214220
443. Fierro-Monti, I., and Mathews, M. B. (2000)
Trends Biochem. Sci. 25, 241246
444. Voyles, B. A. (1993) The Biology of Viruses,
Mosby, St. Louis, Missouri
445. Fields, B. N., and Knipe, D. M., eds. (1990)
Fields Virology, 2nd ed., Raven Press, New York
446. Cann, A. (1993) Principles of Molecular Virology,
Academic Press, San Diego, California
447. Endemann, H., and Model, P. (1995) J. Mol.
Biol. 250, 496506
448. Williams, K. A., Glibowicka, M., Li, Z., Li, H.,
Khan, A. R., Chen, Y. M. Y., Wang, J., Marvin,
D. A., and Deber, C. M. (1995) J. Mol. Biol. 252,
614
449. Wen, Z. Q., Overman, S. A., and Thomas, G.
J., Jr. (1997) Biochemistry 36, 78107820
450. Sanger, F., Air, G. M., Barrell, B. G., Brown, N.
L., Coulson, A. R., Fiddes, J. C., Hutchison, C.
A., III, Slocombe, P. M., and Smith, M. (1977)
Nature (London) 265, 687695
451. McKenna, R., Ilag, L. L., and Rossmann, M. G.
(1994) J. Mol. Biol. 237, 517543
452. McKenna, R., Xia, D., Willingmann, P., Ilag, L.
L., Krishnaswamy, S., Rossmann, M. G.,
Olson, N. H., Baker, T. S., and Incardona, N.
L. (1992) Nature (London) 355, 137143
453. Godson, G. N. (1978) Trends Biochem. Sci. 3,
249253
454. McKenna, R., Bowman, B. R., Ilag, L. L.,
Rossmann, M. G., and Fane, B. A. (1996) J.
Mol. Biol. 256, 736750
455. Wu, H., and Rossmann, M. G. (1993) J. Mol.
Biol. 233, 231244
456. Tsao, J., Chapman, M. S., Agbandje, M.,
Keller, W., Smith, K., Wu, H., Luo, M., Smith,
T. J., Rossmann, M. G., Compans, R. W., and
Parrish, C. R. (1991) Science 251, 14561464
457. Chipman, P. R., Agbandje-McKenna, M.,
Kajigaya, S., Brown, K. E., Young, N. S., Baker,
T. S., and Rossmann, M. G. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 75027506
458. Howarth, A. J., and Goodman, R. M. (1982)
Trends Biochem. Sci. 7, 180182
459. Tiollais, P., Pourcel, C., and Dejean, A. (1985)
Nature (London) 317, 489495
460. Tiollais, P., and Buendia, M.-A. (1991) Sci. Am.
264(Apr), 116123
461. Bttcher, B., Wynne, S. A., and Crowther, R.
A. (1997) Nature (London) 386, 8891
462. Reddy, V. B., Thimmappaya, B., Dhar, R.,
Subramanian, K. N., Zain, B. S., Pan, J.,
Ghosh, P. K., Celma, M. L., and Weissman, S.
M. (1978) Science 200, 494502
463. Liddington, R. C., Yan, Y., Moulai, J., Sahli, R.,
Benjamin, T. L., and Harrison, S. C. (1991)
Nature (London) 354, 278284
464. Griffith, J. P., Griffith, D. L., Rayment, I.,
Murakami, W. T., and Caspar, D. L. D. (1992)
Nature (London) 355, 652654
465. Yang, R. C. A., and Wu, R. (1979) Science 206,
456462
466. Howley, P. M. (1986) N. Engl. J. Med. 315,
10891090
467. Hess, M., Cuzange, A., Ruigrok, R. W. H.,
Chroboczek, J., and Jacrot, B. (1995) J. Mol.
Biol. 252, 379385
468. Athappilly, F. K., Murali, R., Rux, J. J., Cai, Z.,
and Burnett, R. M. (1994) J. Mol. Biol. 242,
430455
469. Witkowski, J. A. (1988) Trends Biochem. Sci. 13,
110113
470. Sugden, B. (1991) Trends Biochem. Sci. 16, 4546
471. Zhou, Z. H., Prasad, B. V. V., Jakana, J., Rixon,
F. J., and Chiu, W. (1994) J. Mol. Biol. 242, 456
469
472. Trus, B. L., Booy, F. P., Newcomb, W. W.,
Brown, J. C., Homa, F. L., Thomsen, D. R., and
Steven, A. C. (1996) J. Mol. Biol. 263, 447462
473. Henle, W., Henle, G., and Lennette, E. T.
(1974) Sci. Am. 241(Jul), 4859
474. Berardelli, P. (1997) Science 278, 1710
475. Senkevich, T. G., Bugert, J. J., Sisler, J. R.,
Koonin, E. V., Darai, G., and Moss, B. (1996)
Science 273, 813816
476. Hohn, T., Hohn, B., and Pfeiffer, P. (1985)
Trends Biochem. Sci. 10, 205209
477. Rosa, P., Mantovani, S., Rosboch, R., and
Huttner, W. B. (1992) J. Biol. Chem. 267, 12227
12232
478. Diener, T. O. (1991) FASEB J. 5, 28082813
479. Diener, T. V. (1984) Trends Biochem. Sci. 9,
133136
480. Diener, T. O. (1979) Viroids and Viroid Diseases,
Wiley, New York
481. Martnez-Soriano, J. P., Galindo-Alonso, J.,
Maroon, C. J. M., Yucel, I., Smith, D. R., and
Diener, T. O. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 93979401
482. Polson, A. G., Bass, B. L., and Casey, J. L.
(1996) Nature (London) 380, 454456
483. Wang, K.-S., Choo, Q.-L., Weiner, A. J., Ou, J.-
H., Najarian, R. C., Thayer, R. M., Mullenbach,
G. T., Denniston, K. J., Gerin, J. L., and
Houghton, M. (1986) Nature (London) 323,
508514
484. Lai, M. M. C. (1995) Ann. Rev. Biochem. 64,
259286
485. Unge, T., Liljas, L., Strandberg, B., Vaara, I.,
Kannan, K. K., Fridborg, K., Nordman, C. E.,
and Lentz, P. J., Jr. (1980) Nature (London) 285,
373377
486. Larson, S. B., Koszelak, S., Day, J., Green-
wood, A., Dodds, J. A., and McPherson, A.
(1993) Nature (London) 361, 179182
487. Larson, S. B., Koszelak, S., Day, J., Green-
wood, A., Dodds, J. A., and McPherson, A.
(1993) J. Mol. Biol. 231, 375391
488. Golmohammadi, R., Valegrd, K., Fridborg, K.,
and Liljas, L. (1993) J. Mol. Biol. 234, 620639
489. LeCuyer, K. A., Behlen, L. S., and Uhlenbeck,
O. C. (1995) Biochemistry 34, 1060010606
490. Stonehouse, N. J., Valegrd, K.,
Golmohammadi, R., van den Worm, S.,
Walton, C., Stockley, P. G., and Liljas, L. (1996)
J. Mol. Biol. 256, 330339
491. Beremarand, M. N., and Blumenthal, T. (1979)
Cell 18, 257266
492. Hogle, J. M., Chow, M., and Filman, D. J.
(1985) Science 229, 13581365
493. Hogle, J. M., Chow, M., and Filman, D. J.
(1987) Sci. Am. 256(Mar), 4249
494. Najaran, R., Caput, D., Gee, W., Potter, S. J.,
Renard, A., Merryweather, J., Van Nest, G.,
and Dina, D. (1985) Proc. Natl. Acad. Sci.
U.S.A. 82, 26272631
495. Rossmann, M. G., Arnold, E., Griffith, J. P.,
Kamer, G., Luo, M., Smith, T. J., Vriend, G.,
Rueckert, R. R., Sherry, B., McKinlay, M. A.,
Diana, G., and Otto, M. (1987) Trends Biochem.
Sci. 12, 313318
276 Chapter 5. The Nucleic Acids
559. Gillespie, D. (1968) in Methods in Enzymology,
Vol. 12B (Grossman, L., and Moldave, K.,
eds), pp. 641668, Academic Press, New York
560. Hall, B. D., Haarr, L., and Kleppe, K. (1980)
Trends Biochem. Sci. 5, 254256
561. Britten, R. J., and Kohne, D. E. (1967) Carnegie
Instit. Washington Yearbook, 65th ed., Carnegie
Instit., Washington, D. C. (pp. 78106)
562. Marmur, J. (1994) Trends Biochem. Sci. 19, 343
346
563. Szabo, P., and Ward, D. C. (1982) Trends
Biochem. Sci. 7, 425427
564. Kessler, C., ed. (1992) Nonradioactive Labeling
and Detection of Biomolecules, Springer-Verlag,
New York
565. Leitch, A. R., Chwarzacher, T., Jackson, D.,
and Leitch, I. J. (1994) In Situ Hybridization:
a Practical Guide, Bios, Oxford
566. Kumar, A., Tchen, P., Roullet, F., and Cohen, J.
(1988) Anal. Biochem. 169, 376
567. Isaac, P. G., ed. (1994) Protocols for Nucleic Acid
Analysis by Nonradioactive Probes, Humana
Press, Totowa, New Jersey
568. Gillevet, P. M. (1990) Nature (London) 348,
657658
569. Negro, F., Pacchioni, D., Shimizu, Y., Miller,
R. H., Bussolati, G., Purcell, R. H., and
Bonino, F. (1992) Proc. Natl. Acad. Sci. U.S.A.
89, 22472251
570. Maniatis, T., Fritsch, E. F., and Sambrook, J.
(1982) Molecular Cloning. A Laboratory Manual,
Cold Spring Harbor Lab. Press, Cold Spring
Harbor, New York
571. Gubler, U., and Hoffman, B. J. (1983) Gene 25,
263269
572. Watson, C. J., and Jackson, J. F. (1985) in DNA
Cloning Vol. I: A Practical Approach (Glover, D.
M., ed), pp. 79100, IRL Press, Washington, DC
573. Ohtsuka, E., Matsuki, S., Ikehara, M.,
Takahashi, Y., and Matsubara, K. (1985) J. Biol.
Chem. 260, 26052608
574. Warner, B. D., Warner, M. E., Karus, G. A., Ku,
L., Brown-Shimer, S., and Urdea, M. S. (1983)
DNA 4, 401411
575. Sekiya, T., Takeya, T., Brown, E. L., Belagaje,
R., Contreras, R., Fritz, H.-J., Gait, M. J., Lees,
R. G., Ryan, M. J., Khorana, H. G., and Norris,
K. E. (1979) J. Biol. Chem. 254, 57875801
576. Wada, T., Sato, Y., Honda, F., Kawahara, S.-i,
and Sekine, M. (1997) J. Am. Chem. Soc. 119,
1271012721
577. Matteucci, M. D., and Caruthers, M. H. (1981)
J. Am. Chem. Soc. 103, 31853191
578. Beaucage, S. L., and Caruthers, M. H. (1981)
Tetrahedron Letters 22, 18591862
579. Air, G. M. (1979) CRC Crit. Revs. Biochem. 6,
133
580. Alvarado-Urbina, G., Sathe, G. M., Liu, W.-C.,
Gillen, M. F., Duck, P. D., Bender, R., and
Ogilvie, K. K. (1981) Science 214, 270274
581. Caruthers, M. H. (1985) Science 230, 281285
582. Lashkari, D. A., Hunicke-Smith, S. P., Norgren,
R. M., Davis, R. W., and Brennan, T. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 79127915
583. Usman, N., Ogilvie, K. K., Jiang, M.-Y., and
Cedergren, R. J. (1987) J. Am. Chem. Soc. 109,
78457854
584. Ebe, K., Sched, M., Rossi, J. J., and Wallace,
R. B. (1987) DNA 6, 497504
585. Usman, N., and Cedergren, R. (1992) Trends
Biochem. Sci. 17, 334339
586. Eaton, B. E., and Pieken, W. A. (1995) Ann.
Rev. Biochem. 64, 837863
587. Rohatgi, R., Bartel, D. P., and Szostak, J. W.
(1996) J. Am. Chem. Soc. 118, 33403344
588. Kleppe, K., Ohtsuka, E., Kleppe, R., and
Khorana, H. G. (1971) J. Mol. Biol. 56, 341361
589. Panet, A., and Khorana, H. G. (1974) J. Biol.
Chem. 249, 52135221
References
496. Olson, N. H., Kolatkar, P. R., Oliveira, M. A.,
Cheng, R. H., Greve, J. M., McClelland, A.,
Baker, T. S., and Rossmann, M. G. (1993) Proc.
Natl. Acad. Sci. U.S.A. 90, 507511
497. Hadfield, A. T., Oliveira, M. A., Kim, K. H.,
Minor, I., Kremer, M. J., Heinz, B. A., Shepard,
D., Pevear, D. C., Rueckert, R. R., and Rossmann,
M. G. (1995) J. Mol. Biol. 253, 6173
498. Kpper, H., Keller, W., Kurz, C., Forss, S.,
Schaller, H., Franze, R., Stohmaier, K.,
Marquardt, O., Zaslavsky, V. G., and
Hofschneider, P. H. (1981) Nature (London)
289, 555559
499. Brown, F. (1981) Trends Biochem. Sci. 6, 325327
500. Acharya, R., Fry, E., Stuart, D., Fox, G.,
Rowlands, D., and Brown, F. (1989) Nature
(London) 337, 709716
501. Luo, M., Vriend, G., Kamer, G., Minor, I.,
Arnold, E., Rossmann, M. G., Boege, U.,
Scraba, D. G., Duke, G. M., and Palmenberg,
A. C. (1987) Science 235, 182191
502. Choi, H.-K., Tong, L., Minor, W., Dumas, P.,
Boege, U., Rossmann, M. G., and Wengler, G.
(1991) Nature (London) 354, 3743
503. Simons, K., Garoff, H., and Helenius, A.
(1982) Sci. Am. 246(Feb), 5866
504. Harrison, S. C., Olson, A. J., Schutt, C. E.,
Winkler, F. K., and Bricogne, G. (1978) Nature
(London) 276, 368373
505. Erickson, J. W., Silva, A. M., Murthy, M. R. N.,
Fita, I., and Rossmann, M. G. (1985) Science
229, 625629
506. Bloomer, A. C., Champness, J. N., Bricogne,
G., Staden, R., and Klug, A. (1978) Nature
(London) 276, 362368
507. Holmes, K. C. (1980) Trends Biochem. Sci. 5, 47
507a. Lauffer, M. A. (1984) Trends Biochem. Sci. 9,
369371
508. Wang, H., and Stubbs, G. (1994) J. Mol. Biol.
239, 371384
509. Kaplan, M. M., and Webster, R. G. (1977) Sci.
Am. 237(Dec), 88106
510. von Itzstein, M., and 17 other authors. (1993)
Nature (London) 363, 418423
511. Clarke, D. K., Duarte, E. A., Elena, S. F., Moya,
A., Domingo, E., and Holland, J. (1994) Proc.
Natl. Acad. Sci. U.S.A. 91, 48214824
512. Varmus, H. (1988) Science 240, 14271434
513. Darlix, J.-L., Lapadat-Tapolsky, M., de
Rocquigny, H., and Roques, B. P. (1995) J. Mol.
Biol. 254, 523537
514. Lwer, R., Lwer, J., and Kurth, R. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 51775184
515. Stephenson, J. R., ed. (1980) Molecular Biology
of RNA Tumor Viruses, Academic Press, New
York
516. Compans, R. W., and Bishop, D. H. L., eds.
(1983) Double-Stranded RNA Viruses, Elsevier,
Biomedical, New York
517. Thornton, A., and Zuckerman, A. J. (1975)
Nature (London) 254, 557558
518. Baldwin, M. A., Cohen, F. E., and Prusiner, S.
B. (1995) J. Biol. Chem. 270, 1919719200
519. Manuelidis, L., Sklaviadis, T., Akowitz, A.,
and Fritch, W. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 51245128
520. Prusiner, S. B. (1996) Trends Biochem. Sci. 21,
482487
521. Prusiner, S. B. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 1336313383
522. Weissmann, C. (1999) J. Biol. Chem. 274, 36
523. Chesebro, B. (1998) Science 286, 660662
524. Balter, M. (1999) Science 286, 660662
525. Temin, H. M. (1978) Trends Biochem. Sci. 3,
N80N83
525a. Sambrook, J., and Russell, D. (2000) Molecular
Cloning: A Laboratory Manual, 3rd ed., Cold
Spring Harbor Lab. Press, Plainview, New
York
526. Grossman, L., and Moldave, K., eds. (1967)
Methods in Enzymology, Vol. 12A, Academic
Press, New York (pp. 531708)
527. Robyt, J. F., and White, B. J. (1996) Biochemical
Techniques, Theory and Practice, 2nd ed.,
Waveland Press, Prospect Heights, Illinois
528. Blackburn, G. M., and Gait, M. J., eds. (1996)
Nucleic Acids in Chemistry and Biology, 2nd ed.,
Oxford Univ. Press, Oxford
529. Rajeswari, M. R., Montenay-Garestier, T., and
Helene, C. (1987) Biochemistry 26, 68256831
530. Hecker, R., Colpan, M., and Riesner, D. (1985)
Journal of Chromatography 326, 251261
531. Edwardson, P. A. D., Atkinson, T., Lowe, C.
R., and Small, D. A. P. (1986) Anal. Biochem.
152, 215220
532. Lawrance, S. K., Smith, C. L., Srivastava, R.,
Cantor, C. R., and Weissman, S. M. (1987)
Science 235, 13871390
533. Cantor, C. R., Smith, C. L., and Matthew, M.
K. (1988) Ann. Rev. Biophys. Biophys. Chem. 17,
287304
534. Smith, C. L., and Cantor, C. R. (1987) Trends
Biochem. Sci. 12, 284287
535. Kochetkov, N. K., and Budovskii, E. I. (1972)
Organic Chemistry of Nucleic Acids, Plenum,
New York (Part A, pp. 137147)
536. Bozic, D., Grazulis, S., Siksnys, V., and Huber,
R. (1996) J. Mol. Biol. 255, 176186
537. Smith, H. O. (1979) Science 205, 455462
538. Air, G. M. (1979) CRC Crit. Revs. Biochem. 6,
133
539. Roberts, R. J. (1980) Methods Enzymol. 65, 115
540. Koob, M., and Szybalski, W. (1990) Science
250, 271273
541. Veselkov, A. G., Demidov, V. V., Frank-
Kamenetskii, M. D., and Nielsen, P. E. (1996)
Nature (London) 379, 214
542. Zaug, A. J., Been, M. D., and Cech, T. R. (1986)
Nature (London) 324, 429433
543. Uhlenbeck, O. C. (1987) Nature (London) 328,
596600
544. Landgraf, R., Chen, C.-hB., and Sigman, D. S.
(1994) Biochemistry 33, 1060710615
545. Adams, R. L. P., Burdon, R. H., Campbell, A.
M., and Smellie, R. M. S. (1976) Davidsons The
Biochemistry of the Nucleic Acids, 8th ed.,
Academic Press, New York (pp. 5082)
546. Rayford, R., Anthony, D. D., Jr., ONeill, R. E.,
Jr., and Merrick, W. C. (1985) J. Biol. Chem.
260, 1570815713
547. Bochner, B. R., and Ames, B. N. (1982) J. Biol.
Chem. 257, 97599769
548. Chen, H., and Shaw, B. R. (1994) Biochemistry
33, 41214129
549. Hayatsu, H., and Shiragami, M. (1979)
Biochemistry 18, 632647
550. Spiro, T. G., ed. (1980) Nucleic Acid-Metal Ion
Interactions, Wiley, New York
551. Dische, Z. (1955) Nucleic Acids 1, 755
552. Marmur, J., and Doty, P. (1962) J. Mol. Biol. 5,
109118
553. Korolev, N., Lyubartsev, A. P., and Norden-
skild, L. (1998) Biophys. J. 75, 30413056
554. Davidson, J. N. (1972) The Biochemistry of
Nucleic Acids, 7th ed., Academic Press, New
York (p. 148)
555. Wada, A., Yabuki, S., and Husimi, Y. (1980)
Crit. Revs. Biochem. 9, 87144
556. Britten, R. J., and Kohne, D. E. (1968) Science
161, 529540
557. Wilson, D. A., and Thomas, C. A., Jr. (1974)
J. Mol. Biol. 84, 115144
558. Bendich, A. J., and Bolton, E. T. (1968) in
Methods in Enzymology, Vol. 12B (Grossman,
L., and Moldave, K., eds), pp. 635640,
Academic Press, New York
277
References
590. Saiki, R. K., Scharf, S., Faloona, F., Mullis, K.
B., Horn, G. T., Erlich, H. A., and Arnheim, N.
(1985) Science 230, 13501354
591. Mullis, K. B. (1990) Sci. Am. 262(Apr), 5665
592. Rabinow, P. (1996) Making PCR: A Story of
Biotechnology, Univ. Chicago Press, Chicago,
Illinois
593. Mullis, K., Ferr, F., and Gibbs, R., eds. (1994)
The Polymerase Chain Reaction, Birkhuser,
Boston, Massachusetts
594. Saiki, R. K., Gelfand, D. H., Stoffel, S., Scharf,
S. J., Higuchi, R., Horn, G. T., Mullis, K. B.,
and Erlich, H. A. (1988) Science 239, 487491
595. Erlich, H. A., Gelfand, D. H., and Saiki, R. K.
(1988) Nature (London) 331, 461462
596. Appenzeller, T. (1990) Science 247, 10301032
597. Newton, C., and Graham, A. (1997) PCR, 2nd
ed., Springer-Verlag, New York
598. Bloch, W. (1991) Biochemistry 30, 27352747
599. White, B. A., ed. (1993) PCR Protocols,
Humana Press, Totowa, New Jersey
599a. Innis, M. A., Gelfand, D. H., and Sninsky, J. J.,
eds. (1999) PCR Applications, Academic Press,
San Diego, California
600. Cohen, J. (1994) Science 263, 15641565
601. Pbo, S. (1993) Sci. Am. 269(Nov), 8692
602. Pbo, S., Higuchi, R. G., and Wilson, A. C.
(1989) J. Biol. Chem. 264, 97099712
603. Hss, M., Pbo, S., and Vereshchagin, N. K.
(1994) Nature (London) 370, 333
604. Scharf, S. J., Horn, G. T., and Erlich, H. A.
(1986) Science 233, 10761078
605. Li, H., Gyllensten, U. B., Cui, X., Saiki, R. K.,
Erlich, H. A., and Arnheim, N. (1988) Nature
(London) 335, 414417
606. Engelke, D. R., Hoener, P. A., and Collins, F. S.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 544548
607. Karlovsky, P. (1990) Trends Biochem. Sci. 15, 419
608. Hengen, P. N. (1995) Trends Biochem. Sci. 20,
372373
609. Wong, G. K.-S., Yu, J., Thayer, E. C., and
Olson, M. V. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 52255230
610. Bernardi, G. (1978) Nature (London) 276, 558
559
611. Bonitz, S. G., Coruzzi, G., Thalenfeld, B. E.,
Tzagoloff, A., and Macino, G. (1980) J. Biol.
Chem. 255, 1192711941
612. Martin, N. C., Miller, D. L., and Donelson, J.
E. (1979) J. Biol. Chem. 254, 1172911734
613. Fischer, S. G., and Lerman, L. S. (1979)
Methods Enzymol. 68, 183191
614. Southern, E. M. (1975) J. Mol. Biol. 98, 503517
615. Southern, E. (1979) Methods Enzymol. 68, 152
176
616. Grunstein, M., and Wallis, J. (1979) Methods
Enzymol. 68, 379 389
617. Sanger, F., and Coulson, A. R. (1975) J. Mol.
Biol. 94, 441448
618. Sanger, F., Nicklen, S., and Coulson, A. R. (1977)
Proc. Natl. Acad. Sci. U.S.A. 74, 54635467
619. Alphey, L. (1997) DNA Sequencing, Springer-
Verlag, New York
620. Ansorge, W., Voss, H., and Zimmerman, J.,
eds. (1997) DNA Sequencing Strategies -
Automated and Advanced Approaches, Wiley,
New York
621. Griffin, H. G., and Griffin, A. M., eds. (1993)
DNA Sequencing Protocols, Humana Press,
Totowa, New Jersey
622. Smith, A. J. H. (1980) Methods Enzymol. 65,
560580
623. Gillam, S., and Smith, M. (1980) Methods
Enzymol. 65, 687701
624. Chen, E. Y., and Seeburg, P. H. (1985) DNA 4,
165170
625. Ornstein, D., Moen, P. T., and Kashdan, M. A.
(1985) DNA 4, 94
626. Bassam, B. J., Caetano-Anolles, G., and
Gresshoff, P. M. (1991) Anal. Biochem. 196,
8083
627. Mizusawa, S., Nishimura, S., and Seela, F.
(1986) Nucleic Acids Res. 14, 13191324
628. Gerken, T. A., Gupta, R., and Jentoft, N. (1992)
Biochemistry 31, 639648
629. Hengen, P. N. (1996) Trends Biochem. Sci. 21,
3334
630. Poncz, M., Solowiejczyk, D., Ballantine, M.,
Schwartz, E., and Surrey, S. (1982) Proc. Natl.
Acad. Sci. U.S.A. 79, 42984302
631. Henikoff, S. (1984) Gene 28, 351359
632. Adams, M. D., Kerlavage, A. R., Kelley, J. M.,
Gocayne, J. D., Fields, C., Fraser, C. M., and
Venter, J. C. (1994) Nature (London) 368, 474475
633. Smith, L. M., Sanders, J. Z., Kaiser, R. J.,
Hughes, P., Dodd, C., Connell, C. R., Heiner,
C., Kent, S. B. H., and Hood, L. E. (1986)
Nature (London) 321, 674679
634. Ju, J., Ruan, C., Fuller, C. W., Glazer, A. N.,
and Mathies, R. A. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 43474351
635. Fu, D.-J., Broude, N. E., Kster, H., Smith, C.
L., and Cantor, C. R. (1995) Proc. Natl. Acad.
Sci. U.S.A. 92, 1016210166
636. Bult, C. J., and 39 other authors. (1996) Science
273, 10581073
637. Sutcliffe, J. G. (1995) Trends Biochem. Sci. 20, 8790
638. Rowen, L., Mahairas, G., and Hood, L. (1997)
Science 278, 605607
639. Mathies, R. A., Huang, X. C., and Quesada,
M. A. (1992) Anal. Chem. 64, 21492154
640. Yeung, E. S., and Li, Q. (1998) in High
Performance Capillary Electrophoresis (Khaldi,
M. G., ed), Wiley, New York
641. Little, D. P., Aaserud, D. J., Valaskovic, G. A.,
and McLafferty, F. W. (1996) J. Am. Chem. Soc.
118, 93529359
642. Maxam, A. M., and Gilbert, W. (1977) Proc.
Natl. Acad. Sci. U.S.A. 74, 560564
643. Gilbert, W. (1981) Science 214, 13051312
644. Maxam, A. M., and Gilbert, W. (1980) Methods
Enzymol. 65, 499560
645. Brownlee, G. G. (1972) in Determination of
Sequences in RNA (Work, T. S., and Work, E.,
eds), North-Holland Publ., Amsterdam
646. Pilly, D., Niemeyer, A., Schmidt, M., and
Bargetzi, J. P. (1978) J. Biol. Chem. 253, 437445
647. Bayev, A. A. (1995) Comprehensive Biochemistry
38, 439479
648. Campbell, P. N. (1995) Trends Biochem. Sci. 20,
259260
649. Myers, T. W., and Gelfand, D. H. (1991)
Biochemistry 30, 76617666
650. Simoncsits, A., Brownlee, G. G., Brown, R. S.,
Rubin, J. R., and Guilley, H. (1977) Nature
(London) 269, 833836
651. Stanley, J., and Vassilenko, S. (1978) Nature
(London) 274, 8789
652. Kitamura, N., and Wimmer, E. (1980) Proc.
Natl. Acad. Sci. U.S.A. 77, 31963200
653. Josse, J., Kaiser, A. D., and Kornberg, A. (1961)
J. Biol. Chem. 236, 864875
654. Stormo, G. D. (1988) Ann. Rev. Biophys.
Biophys. Chem. 17, 241263
655. Goad, W. B. (1986) Ann. Rev. Biophys. Biophys.
Chem. 15, 7995
656. Garner, M. M., and Revzin, A. (1986) Trends
Biochem. Sci. 11, 395396
657. Berger, R., Duncan, M. R., and Berman, B.
(1993) BioTechniques 15, 650652
658. Welsh, J., and Cantor, C. R. (1984) Trends
Biochem. Sci. 9, 505508
659. Galas, D., and Schmitz, A. (1978) Nucleic Acids
Res. 5, 31573170
660. Tullius, T. D. (1987) Trends Biochem. Sci. 12,
297300
661. Tullius, T. D., and Dombroski, B. A. (1986)
Proc. Natl. Acad. Sci. U.S.A. 83, 54695473
662. Prigodich, R. V., and Martin, C. T. (1990)
Biochemistry 29, 80178019
663. Flaus, A., Luger, K., Tan, S., and Richmond,
T. J. (1996) Proc. Natl. Acad. Sci. U.S.A. 93,
13701375
664. Tornaletti, S., and Pfeifer, G. P. (1995) J. Mol.
Biol. 249, 714728
664a. Chaulk, S. G., Pezacki, J. P., and MacMillan,
A. M. (2000) Biochemistry 39, 10448 10453
665. Jaeger, J. A., SantaLucia, J., Jr., and Tinoco, I.,
Jr. (1993) Ann. Rev. Biochem. 62, 255287
666. Httenhofer, A., and Noller, H. F. (1994)
EMBO J. 13, 38923901
667. Laughrea, M., and Tam, J. (1992) Biochemistry
31, 1203512041
668. Reid, B. R. (1981) Ann. Rev. Biochem. 50, 969996
669. Hare, D. R., Ribeiro, N. S., Wemmer, D. E., and
Reid, B. R. (1985) Biochemistry 24, 43004306
670. Ikuta, S., Chattopadhyaya, R., Ito, H.,
Dickerson, R. E., and Kearns, D. R. (1986)
Biochemistry 25, 48404849
671. Lefvre, J.-F., Lane, A. N., and Jardetzky, O.
(1987) Biochemistry 26, 50765090
672. Bax, A., and Lerner, L. (1986) Science 232,
960967
673. Patel, D. J., Shapiro, L., and Hare, D. (1987)
Ann. Rev. Biophys. Biophys. Chem. 16, 423 454
674. Flynn, P. F., Kintanar, A., Reid, B. R., and
Drobny, G. (1988) Biochemistry 27, 11911197
675. Cohen, J. S. (1987) Trends Biochem. Sci. 12,
133135
676. Wemmer, D. E., Chou, S.-H., and Reid, B. R.
(1984) J. Mol. Biol. 180, 4160
677. Schmidt, P. G., Sierzputowska-Gracz, H., and
Agris, P. F. (1987) Biochemistry 26, 85298534
678. Farmer, B. T., Jr., and Mller, L. (1993) J. Am.
Chem. Soc. 115, 1104011041
679. Nikonowicz, E. P., and Pardi, A. (1992) Nature
(London) 355, 184186
680. Tate, S.-i, Ono, A., and Kainosho, M. (1994)
J. Am. Chem. Soc. 116, 59775978
681. Marino, J. P., Prestegard, J. H., and Crothers,
D. M. (1994) J. Am. Chem. Soc. 116, 22052206
682. Moore, P. B. (1995) Acc. Chem. Res. 28, 251256
683. Pardi, A. (1995) Nature Struct. Biol. 1, 846849
684. Mer, G., and Chazin, W. J. (1998) J. Am. Chem.
Soc. 120, 607608
684a. Nikonowicz, E. P., Michnicka, M., and
DeJong, E. (1998) J. Am. Chem. Soc. 120,
3813 3814
684b. Liu, A., Majumdar, A., Hu, W., Kettani, A.,
Skripkin, E., and Patel, D. J. (2000) J. Am.
Chem. Soc. 122, 3206 3210
685. Tolbert, T. J., and Williamson, J. R. (1996) J.
Am. Chem. Soc. 118, 79297940
686. Xu, J., Lapham, J., and Crothers, D. M. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 4448
687. Horowitz, J., Ofengand, J., Daniel, W. E. Jr.,
and Cohn, M. (1977) J. Biol. Chem. 252, 4418
4420
688. Schroeder, S. A., Roongta, V., Fu, J. M., Jones,
C. R., and Gorenstein, D. G. (1989) Biochemistry
28, 82928303
689. Radhakrishnan, I., Patel, D. J., and Gao, X.
(1992) Biochemistry 31, 25142523
690. Schurter, E. J., Sayer, J. M., Oh-hara, T., Yeh,
H. J. C., Yagi, H., Luxon, B. A., Jerina, D. M.,
and Gorenstein, D. G. (1995) Biochemistry 34,
90099020
691. Mao, H., Deng, Z., Wang, F., Harris, T. M., and
Stone, M. P. (1998) Biochemistry 37, 43744387
692. Chou, S.-H., Zhu, L., Gao, Z., Cheng, J.-W., and
Reid, B. R. (1996) J. Mol. Biol. 264, 9811001
693. Kettani, A., Guron, M., and Leroy, J.-L. (1997)
J. Am. Chem. Soc. 119, 11081115
694. Chu, Wen-C., Liu, J. C.-H., and Horowitz, J.
(1997) Nucleic Acids Res. 25, 39443949
278 Chapter 5. The Nucleic Acids
1. Describe the typical distribution pattern of RNA
and DNA in bacterial cells and in eukaryotic cells.
2. Draw the structures of the WatsonCrick base
pairs guanine cytosine (GC) and adenine thym-
ine (AT). Also draw the GU pair, which is not a
Watson Crick pair.
3. Draw the tautomeric structures possible for the
cation formed by protonation of 9-methyladenine.
4. What unusual base pairs could arise from a minor
tautomer of cytosine or from a minor tautomer of
guanine?
5. The minor imino tautomeric form of adenosine
occurs infrequently in DNA. Can this cause muta-
tions? Explain; draw structures to illustrate your
answer.
6. Will the substitution of hypoxanthine for adenine
in DNA result in mutation? Explain.
7. Why is the methylation of DNA to form O
6
-methyl
guanine mutagenic?
8. Draw the structure of a dinucleotide that might be
obtained by the partial hydrolysis of RNA. Indicate
the following:
a) The 5' end
b) The 3' end
c) The torsion angle
d) The point of cleavage by pancreatic ribonuclease
e) The point of cleavage by periodic acid
f) Two points at which the structure might be
methylated by modifying enzymes
acting on a polynucleotide
9. Draw the structure of guanosine-5'-phosphate in
such a way that the configurations of the sugar
ring and of the glycosidic linkage are clearly indi-
cated. State whether you have drawn a syn or an
anti conformer. Circle the most acidic proton in
the guanine ring and indicate its approximate pK
a
.
Which is the most basic center? What is the ap-
proximate pK
a
of the conjugate acid?
10. What are the chemical functional groups in DNA?
In RNA?
Study Questions
11. Electrophoresis of a mixture of the dinucleotides
ApC and ApU at pH 3.5 separates two compo-
nents. Identify these and explain the order of
migration. Be as quantitative as possible.
12. Draw the structure of the predominant form of
pGpC as it occurs at pH 3.5.
13. Why is DNA denatured at pH 11?
14. Draw a schematic representation of the polynucle-
otide portion of a DNA molecule and of an RNA
molecule and indicate positions of cleavage by the
following treatments:
a) Mild HCl
b) More vigorous HCl
c) Mild NaOH
d) More vigorous NaOH
e) Pancreatic RNase
f) Pancreatic DNase
g) Splenic DNase
h) Splenic phosphodiesterase
i) Snake venom phophodiesterase
j) Dnase from Micrococcus
15. A sample of DNA from a virus was hydrolyzed by
acid and was found to have the following base
composition (in mol%): adenine, 30; thymine, 39;
guanine, 18; cytosine, 13. This differs from that of
most DNA preparations. Offer a possible explana-
tion. Sketch the expected temperature-absorbance
profile of this DNA. Do you expect much hyper-
chromicity? Explain your answer.
16. Adenine is found to constitute 16.3% of the nucleic
acid bases in a sample of bacterial DNA. What are
the percentages of the other three bases?
17. For the following DNA sequence
3'-CGATACGGCTATGCCATAGGC-5'
write a) the sequence of the complementary
DNA strand;
b) the sequence of the corresponding
segment of mRNA formed using the
DNA segment above as the template;
c) the amino acid sequence encoded by
this segment.
18. What is the molecular mass of a segment of B-DNA
that encodes a 386-residue protein? What is the
length in nm? in ? Do not make allowance for
introns.
279
Study Questions
19. Complete the following table:
Protein
Polysaccharide
Nucleic acid
Teichoic acid
Poly--hydroxybutyrate
20. What is meant by the T
m
of a DNA sample? How
does T
m
vary with base composition and what is
the explanation of this?
21. Isolated naked bacterial DNA, from which
proteins have been removed, is supercoiled. DNA
in the bacterial chromosome is also supercoiled.
When naked DNA is nicked, its supercoiling is
abolished. In contrast nicking the chromosomal
DNA does not abolish its supercoiling. Explain.
22. A closed circular duplex DNA has a 90 base-pair
segment of alternating G and C residues. Upon
transfer to a solution containing a high salt concen-
tration, this segment undergoes a transition from
the B conformation to the Z conformation.
a) Explain why the high salt concentration
induces a B Z transformation.
b) What changes would you expect in (1) the
linking number Lk, (2) the writhe Wr, and (3)
the twist Tw of the DNA as a result of this
transition.
23. Name two or more characteristics of a DNA se-
quence or of its environment that will favor con-
version of B-DNA into Z-DNA.
24. Suppose one double helical turn of a superhelical
DNA molecule changes from a B conformation to
the Z conformation. Calculate the approximate
changes in (1) the linking Lk, (2) the writhe Wr,
and (3) the twist Tw of the DNA as a result of this
transition. Show your calculations and explain
your answers. For this problem assume that the B
form of DNA has 10.4 bp per turn. Why is the B
Z transition favored in naturally occurring super-
coiled DNA?
25. A circular DNA plasmid of length 1144 bp is super-
coiled with a twist (Tw) of 110. Assume that the
DNA has 10.4 bp per turn in its relaxed state.
a) What is the linking number Lk and the writhe
Wr in the plasmid?
b) Is the plasmid negatively or positively super-
coiled?
c) Ethidium bromide is an intercalating agent that
inserts between the stacked base pairs, separat-
ing the stacks and causing local unwinding
that decreases the value of Tw. What effect
would ethidium bromide have on the migra-
tion rate of the plasmid during electrophoresis?
d) If part of the plasmid were to undergo a transi-
tion from B-DNA to Z-DNA, what would be
the effect on Lk, Tw, and Wr?
26. You have been given a sample of nucleic acid,
describe two ways you could determine whether
it is RNA or DNA and two ways to determine
whether it is single- or double-stranded.
27. What conclusions can you draw about the nature
of the protein binding site on DNA from the obser-
vation that methylation of cytosine residues in the
protein-recognition sequence inhibits protein
binding?
28. The anticodon loop of one of the tRNA Gly mole-
cules from E. coli is as follows. Identify the anti-
codon, reading from 3' to 5'. This tRNA recognizes
two different Gly codons. What are they? Write
them from 5' to 3'.
The complete tRNA contains 75 nucleotides.
Sketch the rest of the molecule in the cloverleaf
representation. Label the 5' and 3' ends and the
dihydrouridine and TC loops. What are the last
three nucleotides at the 3' end?
29. Are viruses alive? Explain your answer.
Name
Range of
molecular
masses Linkage Monomer
C
G
A
U
C
C
U
G
C
C
A
A
G
G
U
C
G
35
30
tRNA
Gly
3
280 Chapter 6. Thermodynamics and Biochemical Equilibria
281
282
282
282
282
284
284
285
285
286
286
287
287
288
289
289
292
292
292
292
293
297
297
297
300
302
302
303
303
303
304
305
305
307
307
311
311
312
319
321
A. Thermodynamics
1. The First Law of Thermodynamics
2. Enthalpy Changes and Thermochemistry
Processes at constant pressure
Enthalpies of combustion and physiological fuel values
3. The Second Law of Thermodynamics
The thermodynamic temperature
Entropies from measurement of heat capacities
4. A Criterion of Spontaneity: The Gibbs Energy
5. Practical Thermochemistry
Summing changes in Gibbs energy
Reactions in solution
G and the equilibrium constant
Activity coefficients and concentration
equilibrium constants
Changes in equilibria with temperature
6. Thermodynamics and Life Processes
B. Tables of G Values for Biochemical Compounds
1. Gibbs Energies of Formation
2. Gibbs Energies of Dissociation of Protons
3. Group Transfer Potentials
4. Constants That Vary with pH and Magnesium
Ion Concentrations
5. A New Standard for Biochemical Thermodynamic?
6. Bond Energies and Approximate Methods for
Estimation of Thermodynamic Data
7. Gibbs Energies of Combustion by O
2
and by NAD
+
C. Electrode Potentials and Gibbs Energy Changes
for OxidationReduction Reactions
D. The Adenylate System
1. Storage and Utilization of Energy
2. Synthesis of ATP
3. Creatine Phosphate, an Energy Buffer
4. Phosphorus-31 NMR
E. Complex Biochemical Equilibria
1. Effects of pH on Equilbria
2. Microscopic Dissociation Constants and
Tautomerization
3. The Binding of Metal Ions
Factors affecting the strength of binding of a
metal in a complex
How properties of the metal ion affect chelation
Metal binding sites in cells
Calcium-binding proteins
References
Study Questions
295
299
308
314
317
283
283
285
290
293
294
298
301
310
311
Boxes
Box 6-A Measurement of Intracellular pH
Box 6-B Magnesium
Box 6-C Linear Gibbs Energy Relationships
Box 6-D Calcium
Box 6-E Metallothioneins
Tables
Table 6-1 Units of Energy and Work and the Values
of Some Physical Constants
Table 6-2 Caloric Values of Food Components
Table 6-3 Entropies of Selected Substances
Table 6-4 Gibbs Energies of Formation and of
Oxidation at 25C for Compounds of
Biochemical Interest
Table 6-5 Values of pKa, G, and H for
Ionization of Acids at 25C
Table 6-6 Gibbs Energies of Hydrolysis at 25C
(in kJ mol
-1
)
Table 6-7 Empirical Bond Energies and Resonance
Energies
Table 6-8 Reduction Potentials of Some Biologically
Important Systems
Table 6-9 Logarithms of Binding Constants for
Some 1:1 Metal Complexes at 25C
Table 6-10 Ionic Radii in Nanometers for Some
Metallic and Nonmetallic Ions
Contents
Ice and water are in equilibrium at 0C and atmospheric pressure. When ice melts under these conditions
the heat Q absorbed from the surroundings is the enthalpy change, H, which equals 6.008 kJ mol
1
. For
a reversible reaction the entropy change, S, equals H/T and the entropy increases when the ice melts
by 6.008 x 10
3
J / 273.16 = 22.0 JK
1
. Along the frozen edges of the river the ice and water are not in a true
equilibrium but in a steady state. It is an open system in which water flows and energy is exchanged
with the surroundings. Similarly, the cells of Vorticella represent open systems through which water and
nutrients flow, and energy is exchanged with the surrounding water. Many chemical reactions within the
cells are near equilibrium while the organism maintains its own structure. Photos: Frosty River, Banff
Natl. Park, Alberta Stephen J. Kraseman; Vorticella courtesy of Ralph Buchsbaum.
281
Thermodynamics and Biochemical Equilibria
6
We all know from experience the importance of
energy to life. We know that we must eat and that
hard work not only tires us but also makes us hungry.
Our bodies generate heat, an observation that led
Lavoisier around 1780 to the conclusion that respira-
tion represented a slow combustion of foods within
the body. It soon became clear that respiration must
provide the energy for both the mechanical work done
by muscles and the chemical synthesis of body constit-
uents. All organisms require energy and the ways in
which living things obtain and utilize energy is a major
theme of biology.
The discovery of the first and second laws of
thermodynamics permitted the development of precise,
quantitative relationships between heat, energy, and
work. It also allowed chemical equilibria to be
understood. Modern biochemical literature abounds
with references to the thermodynamic quantities
energy E, enthalpy H, entropy S, and Gibbs energy
(also called free energy) G. The purposes of this chapter
are: (1) to provide a short review of thermodynamic
equations, (2) to provide tables of thermodynamic
quantities for biochemical substances and to explain
the use of these data in the consideration of equi-
libria in biochemical systems, and (3) to introduce
the adenylate system, which consists of adenosine
triphosphate (ATP), adenosine diphosphate (ADP),
adenosine mono-phosphate (AMP), and inorganic
phosphate (P
i
). This system plays a central role in
energy metabolism, and (4) to provide a quantitative
understanding of the effects of pH and of metal ions
on biochemical equilibria. Many readers will want to go
directly to Section D, which deals with the adenylate system
and its significance for life.
A. Thermodynamics
Thermodynamics is concerned with the quantita-
tive description of heat and energy changes and of
chemical equilibria.
1 10
Knowledge of changes in
thermodynamic quantities, such as H and S, enables
us to predict the equilibrium positions in reactions and
whether or not under given circumstances a reaction
will or will not take place. Furthermore, the consider-
ation of thermodynamic quantities provides insight into
the nature of forces responsible for bonding between
molecules, enzymatic catalysis, functioning of DNA
and RNA, and many other phenomena.
It is important to realize that while thermodynamic
information will tell us whether or not a reaction can
take place it says nothing about the rate of the reaction.
It will not even say whether a reaction will proceed at
all within a given period of time. This has led to the
occasional assertion that thermodynamics is not rele-
vant to biochemistry. This is certainly not true; it is
important to understand energy relationships in bio-
chemical reactions. At the same time, one should avoid
the trap of assuming that thermodynamic calculations
appropriate for equilibriumsituations can always be
applied directly to the steady state found in a living cell.
Thermodynamics is an exact science and its laws
deal with measurable quantities whose values are
determined only by the state of the system under
consideration. For example, the system might be the
solution in a flask resting in a thermostated bath. To
specify its state we would have to say whether it is
pure solid, liquid, or gas, or a solution of specified
composition and give the temperature and pressure.
The flask, the bath, and everything else would be
282 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-3)
(6-4)
(6-1)
called surroundings or environment. The system plus
surroundings is sometimes referred to as the universe.
1. The First Law of Thermodynamics
The first law of thermodynamics asserts the con-
servation of energy and also the equivalence of work
and heat. Work and heat are both regarded as energy
in transit. Heat may be absorbed by a system from the
surroundings or evolved by a system and absorbed in
the surroundings. Work can be done by a system on
the surroundings or it can be done on a system. The
first law postulates that there is an internal energy E
(also designated U), which is dependent only on the
present state of the system and in no way is dependent
upon the history of the system. The first law states
that E can be changed only by the flow of energy as heat or
by work. In other words, energy can neither be created
nor destroyed.
In mathematical form, the first law is given as
follows:
E E E Q W = = (products) (reactants)
Here Q is the heat absorbed by the system from the
surroundings and W is the work done by the system
on the surroundings. Energy, heat, and work are all
measured in the same units. Chemists have tradition-
ally used the calorie (cal) or kilocalorie (kcal) but
are switching to the SI unit, the joule (Table 6-1).
10a
Work done by the system may be mechanical (e.g., by
changing the volume of the surroundings), electrical
(e.g., by charging of a battery), or chemical (e.g., by
effecting the synthesis of a polypeptide from amino
acids).
2. Enthalpy Changes and Thermochemistry
We are most often interested in the changes in the
thermodynamic functions when a chemical reaction
takes place; for example, the heat absorbed by the
system within a bomb calorimeter where the volume
stays constant (Q
V
) is a direct measure of the change in E:
Q E
V
=
To measure E for combustion of a biochemical com-
pound, the substance may be placed in a bomb together
with gaseous oxygen and the mixture ignited within
the calorimeter by an electric spark. In this case, heat
will be evolved from the bomb and will pass into the
surroundings. Q
V
and E will be negative. The bomb
calorimeter is designed to measure Q
V
and thereby to
give us a way of determining E for reactions.
Processes at constant pressure. Chemical and
biochemical reactions are much more likely to be con-
ducted at constant pressure (usually 1 atm) than they
are at constant volume. For this reason, chemists tend
to use the enthalpy H more often than the internal
energy E.
H = E + PV
It follows from Eq. 6-3 that if the pressure is constant, H
P
is equal to E
P
+ P V . Since in a process at constant
pressure, P V is exactly the pressurevolume work
done on the surroundings, the heat absorbed at constant
pressure (Q
P
) is a measure of H
P
.
Since enthalpy changes can be obtained directly
from measurement of heat absorption at constant
pressure, even small values of H for chemical and
biochemical reactions can be measured using a micro-
calorimeter.
11,12
Using the technique of pulsed acoustic
calorimetry, changes during biochemical processes can
be followed on a timescale of fractions of a millisecond.
An example is the laser-induced dissociation of a
carbon monoxidemyoglobin complex.
13
The term enthalpy was coined to distinguish H from
E, but we sometimes tend to be careless about language
and many discussions of energy in the literature are
in fact about enthalpy. The difference is often not
significant because if the pressurevolume work is
negligible, E and H are the same.
Enthalpies of combustion and physiological
fuel values. The heat of combustion (H
c
) of an
organic substance is usually determined from E
c
,
which is measured in a bomb calorimeter. Since E
V
and E
P
are nearly identical, it follows that H
P
= E
V
+ P V. Here V is the volume change which would
have occurred if the reaction were carried out
at constant pressure P; thus, H
P
can be estimated by
calculation. Since H is desired for combustion to
carbon dioxide, water, elemental nitrogen (N
2
), and
sulfur, correction must be made for the amounts of the
latter elements converted into oxides. By these proce-
dures, it has been possible to obtain highly accurate
values of H
c
both for biochemical compounds and
for mixed foodstuffs. In nutrition, H
c
is sometimes
referred to as the gross energy. Values are usually
expressed in kilocalories (kcal) by chemists but often
as Cal (with a capital C) in the nutritional literature.
Caloric values of foods (physiological fuel values)
are enthalpies of combustion but with an opposite
sign, ( H
c
), and corrected for energy lost in urine
(e.g., as urea) and feces. While enthalpies of combustion
of foods are all negative, the caloric values are given
as positive numbers. Caloric values for proteins are
(6-2)
Q
P
= E
P
+ P V = H
P
283
calculated for the conversion of the nitrogen to urea,
the major nitrogenous excretion product in mammals,
rather than to elemental nitrogen. Typical values are
shown in Table 6-2.
From a thermochemical viewpoint, can a human
or animal be regarded as just a catalyst for the com-
bustion of foodstuffs? To answer this question, large
calorimeters were constructed into which an animal or
a human being was placed. If, while in the calorimeter,
the subject neither gained nor lost weight, the heat
evolved should have been just equal to H for com-
bustion of the food consumed to CO
2
, water, and urea.
That this prediction was verifed experimentally does
not seem surprising, but at the time that the experi-
ments were first done in the early years of the century
there may have been those who doubted that the first
law of thermodynamics applied to mammals.
In practice, animal calorimetry is quite complicated
because of the inherent difficulty of accurate heat
measurements, uncertainties about the amount of food
stored, and the necessity of corrections for H
c
of the
waste products. However, the measurement of energy
metabolism has been of considerable importance in
nutrition and medicine. Indirect methods of calorime-
tery have been developed for use in measuring the basal
metabolic rate of humans. For a good discussion see
White et al.
14
The basal metabolic rate is the rate of heat evolution
in the resting, postabsorptive state, in which the subject
has not eaten recently. In this condition, stored foods
provide the energy and are oxidized at a relatively
constant rate. The basal metabolic rate tends to be
proportional to the surface area; which can be approxi-
mated (in units of m
2
) as 1/60 [height (cm) mass
(kg)].
15
For a young adult female, the basal metabolic
rate is typically 154 kJ h
1
m
2
and for a young adult
male 172 kJ h
1
m
2
. This is 320 360 kJ h
1
for a 70-kg
person. Note that 360 kJ h
1
is the same as the power
output of a 100-watt lightbulb. While there is consid-
erable variation among individuals, basal metabolic
rates far below or above normal may indicate a patho-
logical condition such as an insufficiency or oversupply
of the thyroid hormone thyroxine. Metabolic rates fall
somewhat below the basal value during sleep and are
much higher than basal during hard exercise. A human
may attain rates as high as 2500 kJ (600 kcal) per hour.
At a basal rate of 320 kJ (76 kcal) per hour, a person
requires 7680 kJ (1835 kcal) each 24 h to supply his or
her basal needs, plus additional energy during periods
of muscular exercise. Routine light exercise as in the
office or during housework increases metabolism to
about double the basal rate. Although the caloric values
in Table 6-2 are reliable for prediction of metabolic
energy needs, they must be adjusted to predict the
efficiency of utilization for growth. In one study
16
a
group of rats deposited 28% of the available energy
from sucrose as body protein and fats, but fats were
deposited with an efficiency of 36%.
The joule, SI unit of energy
1 J = 1 kg m
2
s
2
= 1 N m (newton meter)
= 1 W s (watt second)
= 1 C V (coulomb volt)
Thermochemical calorie
1 cal = 4.184 J
Large calorie
1 Cal = 1 kcal = 4.184 kJ
Work required to raise 1 kg 1 m on earth
(at sea level) = 9.807 J
Gibbs energy of hydrolysis of 1 mole of ATP
at pH 7, millimolar concentrations =
12.48 kcal = 52.2 kJ
Work required to concentrate 1 mole of a substance
1000-fold, e.g., from 10
6
to 10
3
M
= 4.09 kcal = 17.1 kJ
Avogadros number, the number of particles in a mole
N = 6.0220 x 10
23
Faraday 1 F = 96,485 C mol
1
(coulombs per mole)
Coulomb 1 C = 1 A s (ampere second)
= 6.241 x 10
18
electronic charges
The Boltzmann constant
k
B
= 1.3807 x 10
23
J deg
1
The gas constant, R = N k
B
R = 8.3144 J deg
1
mol
1
= 1.9872 cal deg
1
mol
1
= 0.08206 1 atm deg
1
mol
1
and at 25C RT = 2.479 kJ mol
1
The unit of temperature is the kelvin (K); 0C =
273.16 K
ln x = 2.3026 log x
One atmosphere (atm) = 101.325 kilopascals (kPa)
TABLE 6-1
Units of Energy and Work and the Values of Some
Physical Constants
TABLE 6-2
Caloric Values of Food Components
Component Caloric values per gram
Carbohydrates 4.1 kcal 17 kJ
Pure glucose 3.75 kcal 15.7 kJ
Lipids 9.3 kcal 39 kJ
Proteins
a
4.1 kcal 17 kJ
a
Nitrogen excreted as urea.
A. Thermodynamics
284 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-5)
(6-6)
3. The Second Law of Thermodynamics
Why does heat flow from a warm body into a cold
one? Why doesnt it ever flow in the reverse direction?
We can see that differences in temperature control the
direction of flow of heat, but this observation raises
still another question: What is temperature? Reflec-
tion on these questions, and on the interconversion of
heat and work, led to the discovery of the second law
of thermodynamics and to the definition of a new
thermodynamic function, the entropy S.
Consider the melting of ice. This is a phase transi-
tion that usually takes place at constant temperature
and pressure. At a temperature just above 0C ice
melts completely, but at a temperature just below 0C
it does not melt at all. At 0C we have an equilibrium.
In the language of thermodynamics, the melting of ice
at 0C is a reversible reaction. What criterion could
be used to predict this behavior for water? For many
familiar phenomena, e.g., combustion, a spontaneous
reaction is accompanied by the evolution of a large
amount of heat, i.e., H is negative. However, when
ice melts it absorbs heat. The H of fusion amounts to
6.008 kJ mol
1
at 0C and is nearly the same just below
0C, where the ice does not melt, and just above 0C,
where the ice melts completely. In the latter case, the
melting of ice is a spontaneous reaction for which H
is positive. It is clear from such facts that the sign of the
enthalpy change does not serve as a criterion of spontaneity.
A correct understanding of the icewater transi-
tion came when it was recognized that when ice melts
not only does H increase by 6.008 kJ mol
1
, as the mole-
cules acquire additional internal energy of translation,
vibration, and rotation, but also the molecules become
more disordered. Although historically entropy was
introduced in a different context, it is now recognized
to be a measure of microscopic disorder. When ice
melts, the entropy S increases because the structure
becomes less ordered.
The second law of thermodynamics is stated in
many different ways, but the usual mathematical
formulation asserts that for the universe (or for an
isolated system)
S (system + surroundings) = 0
for reversible processes
S > 0
for real (nonreversible) processes
The second law is sometimes stated in another way:
The entropy of the universe always increases.
The second law also defines both S and the thermo-
dynamic temperature scale as follows:
dS q T
reversible
=
Here q is an infinitesimal quantity of heat absorbed
from the surroundings by the system and T is measured
in kelvins (K). For a reversible phase transition such as
the melting of ice at constant pressure and temperature,
the change in entropy of the H
2
O is just H/T.
S Q T H T
P T
)
, , reversible
= =
Entropy is measured in units of joules per kelvin (or C)
or calories per K, the latter sometimes being abbreviated
as e. u. (entropy units). Since the melting of ice at 0C
is a reversible process, the second law asserts that the
entropy of the surroundings decreases by the same
amount that the entropy of the water increases. The
value of T S is numerically equal to the heat of fusion,
6.008 kJ mol
1
in the case of water at 0C. Thus, the
entropy increase in the ice as it melts at 0C is 6.008 x
10
3
J/273.16 K = 22.0 J K
1
.
The thermodynamic temperature. The definition
of thermodynamic temperature in kelvins (Eq. 6-8) also
follows from Eq. 6-6. See textbooks of thermodynamics
for further treatment.
T E S H S
V P
= = ( ) ( )
The entropy of a substance can be given a precise
mathematical formulation involving the degree of
molecular disorder (Eq. 6-9).
S k =
B
ln
Here k
B
is the Boltzmann constant (see Table 6-1)
and is given precisely as the number of microscopic
states (different arrangements of the particles) of the
system corresponding to a given macroscopic state,
i.e., to a given temperature, pressure, and quantity. It
increases as volume or temperature is increased and in
going from solid to liquid to gaseous states. Equation
6-9 is not part of classical thermodynamics (which
deals only with macroscopic systems, i.e., with large
collections of molecules). However, using the methods
of statistical thermodynamics,
17
this equation can be
used to predict the entropies of gases.
The racemization of an amino acid provides a bio-
chemical example that can be related directly to Eq. 6-9.
A solution of an L-amino acid will be efficiently changed
into the racemic mixture of 50% D and 50% L by the
action of an enzyme (a racemase) with no uptake or
evolution of heat. Thus, H = 0 and the only change
is an entropy change. Let us designate for the pure
isomer as . Since there are just two choices of con-
figuration for each of the N molecules in 1 mole of the
racemate we see that for the racemate
= 2
N
(6-7)
(6-8)
(6-9)
(6-10)
285
Applying Eq. 6-9 we calculate S as follows:
S k k
Nk R
N
= +
= = =

B B
B
1
J K mol
(ln ln ) ln
ln ln .
2
2 2 5 76
1
Entropies from measurement of heat capacities.
It follows from Eq. 6-9 that S = 0 when T = 0 for a
perfect crystalline substance in which no molecular
disorder exists. The third law of thermodynamics asserts
that as the thermodynamic temperature T approaches
0 K the entropy S also approaches zero for perfect
crystalline substances. From this it follows that at any
temperature above 0 K, the entropy is given by Eq. 6-12.
S C d T
p
T
=

ln
0
In this equation C
P
is the heat capacity at constant
pressure:
C H T
P P
= ( )
If C
P
is measured at a series of low temperatures down
to near zero K, Eq. 6-12 can be used to evaluate the
absolute entropy S. If phase transitions occur as the
temperature is raised, entropy increments given by Eq.
6-7 must be added to the value of S given by Eq. 6-12.
For a few compounds, such as water (Chapter 2),
molecular disorder is present in the crystalline state
even at 0 K. For these substances a term representing
the entropy at 0 K must be added to Eq. 6-12.
The entropies of a few substances are given in
Table 6-3. Notice how the entropy increases with
increasing complexity of structure, with transitions
from solid to liquid to gas, and with decreasing hard-
ness of solid substances.
Measurements of C
p
versus temperature for solu-
tions of macromolecules or for biological membranes
(Fig. 8-9) over a narrower temperature range are also
of interest. These can be obtained with a differential
scanning calorimeter
18,19
or by an indirect procedure.
20
Denaturation of polymers or phase changes in mem-
branes may be observed. Larger values of C
p
are ob-
served for open, denatured, or random-coil structures
that are usually present at higher temperatures than
for tightly folded molecules.
4. A Criterion of Spontaneity: The Gibbs
Energy
We have seen that while many spontaneous pro-
cesses, e.g., combustion of organic compounds, are
accompanied by liberation of heat (negative H), others
are accompanied by absorption of heat from the sur-
roundings (positive H). An example of the latter is
the melting of ice at a temperature just above 0C,
during which there is a large increase in the entropy of
the water. As we have seen, at 0C at equilibrium T S
is just equal to H (Eq. 6-7).
The recognition that H T S = 0 for a system at
equilibrium led J. W. Gibbs to realize that the proper
thermodynamic function for determining the sponta-
neity of a reaction is what is now known as the Gibbs
energy or Gibbs function G (Eq. 6-14).
G H TS =
In the older literature the Gibbs energy was usually
called the free energy or Gibbs free energy and was
often given the symbol F. For a process at constant
temperature and pressure the change in G is given by
Eq. 6-15 in which all quantities refer to the system.
G H T S
T P ,
=
For a reversible (equilibrium) process doing only
pressurevolume work:
G H T S
T
)
, reversible
= = 0
It can also be shown readily that G is negative for any
spontaneous (irreversible) process. Such a process is
called exergonic. Likewise, if G is positive, a given
(6-11)
(6-12)
(6-13)
C (diamond) s 0.55 2.3
C (graphite) s 1.36 5.7
Cu s 8.0 33
Na s 12.2 51
H
2
O (ice) s 9.8 41
H
2
O l 16.7 70
H
2
O g (1 atm) 45.1 189
He g 30.1 126
H
2
g 31.2 131
N
2
g 45.8 192
CO
2
g 51.1 214
Benzene g 64.3 269
Cyclohexane g 71.3 298
Entropy S
Substance State
b
cal K
1
mol
1
J K
1
mol
1
a
All values are given in entropy units (e.u.) of calories per Kelvin
per mole and in joules per Kelvin per mole at 25C (298.16 K).
b
Here s stands for solid, l for liquid, and g for gaseous.
A. Thermodynamics
(6-14)
(6-15)
(6-16)
TABLE 6-3
Entropies of Selected Substances
a
286 Chapter 6. Thermodynamics and Biochemical Equilibria
reaction will not proceed spontaneously and is called
endergonic. The magnitude of the decrease in the
Gibbs energy (G) is a direct measure of the maximum
work which could be obtained from a given chemical
reaction if that reaction could be coupled in some
fashion reversibly to a system able to do work. It
represents the maximum amount of electrical work
that could be extracted or the maximum amount of
muscular work or osmotic work obtainable from a
reaction in a biological system. In any real system,
the amount of work obtainable is necessarily less than
G because real processes are irreversible, i.e., entropy
is created.
Returning to the older assumption that the magni-
tude of H might be an index of work obtainable, we
note that T S amounts to only a few kilojoules for
most reactions. Therefore, if H is large, as in the
combustion of foodstuffs, it is not greatly different from
G for the same process. Therefore, we can justify use
of the caloric value of a food as an approximate measure
of the work obtainable from its metabolism in the
body.
5. Practical Thermochemistry
For thermodynamic data to be useful in chemical
calculations, we must agree upon standard states for
elements and compounds. If we wish to talk about the
change in the Gibbs energy that occurs when one or
more pure compounds are converted to other pure
substances, we must agree upon a state (crystalline,
liquid, gaseous, or in solution) and upon a pressure
(especially when gases are involved) at which the data
apply. The standard pressure is usually 1 atm. Stan-
dard states of the elements are the pure crystalline, solid,
or gaseous materials, e.g., C (graphite), S (crystalline,
rhombic), P (crystalline, white), and N
2
, O
2
, and H
2
(gaseous). It is also essential to specify the temperature.
Thermodynamic data are most often given for 25C,
but there is a standard state for each substance at each
temperature.
It is usually impractical to measure the values of
G or H, but G and H for a chemical reaction can be
evaluated. Changes in Gibbs energy can be calculated
from tables of G for formation of compounds from
the elements (Eq. 6-17). These values of G
f
can be
obtained experimentally by measuring H of combus-
tion for the compound of interest and for H
2
, elemental
carbon, and other elements present in the compound
and also by obtaining entropies from heat capacity
measurements. Many other tabulated G
f
values have
been obtained indirectly utilizing data from equilibrium
constants. The resulting standard Gibbs energies
of formation are given the symbol G
f
. The values of
G
f
for the elements in their standard states are all exactly
zero.
Summing changes in Gibbs energy. A conve-
nient feature of thermodynamic calculations is that if
two or more chemical equations are summed, G for
the resulting overall equation is just the sum of the
Gs for the individual equations as illustrated in Eqs.
6-17 to 6-20. The same applies for H and S.
CH COOH C H O
for acetic acid kJ mol
O C CO
for CO kJ mol
O H H O
for H O kJ mol
CH COOH l O CO H O (l)
f
f
f
c
3 2 2
1
2 2
2
1
2 2 2
2
1
3 2 2 2
1 2 2
396 4
2 2 2
2 788 8
2 2
2 474 4
2 2 2
( )
.
.
.
( )
+ +
= +
+
=
+
=
+ +

G
G
G
G =

for acetic acid kJ mol 866 8
1
.
In this example an equation for the decomposition of
acetic acid into its elements (Eq. 6-17) has been summed
with Eqs. 6-18 and 6-19, which represent the formation
of the proper number of molecules of CO
2
and H
2
O
from the elements. The sum of the three equations
gives the equation for the combustion of acetic acid to
CO
2
and water, and the sum of the G values for the
three equations gives G for combustion of acetic acid.
The resulting value of G is for combustion of pure
liquid acetic acid by oxygen at 1 atm to give CO
2
at 1
atm and pure liquid water, all reactants and products
being in their standard states.
The process described in the preceding paragraph
is represented by Eq. 6-21, which is a general equation
for calculation of G for any reaction from G
f
of
products and reactants.
G G G =
f f
products reactants ( ) ( )
How does the change in Gibbs energy vary if we go
from the standard state of a compound to some other
state? Consider a change of pressure in a gas. It is
easy to show (see any thermodynamics text) that
( ) G P V
T
=
Using Eq. 6-22 together with the perfect gas law, we
obtain the relationship (Eq. 6-23) between the Gibbs
energy G of one mole of a substance at pressure P and
the standard Gibbs energy G at pressure P.
G G RT
P
P
RT P =

= ln ln
(6-17)
(6-18)
(6-19)
(6-20)
(6-21)
(6-22)
(6-23)
287
(6-28)
(6-27)
Here the bar over the symbol G indicates that the
Gibbs energy is for one mole of substance. Since P is
by definition 1 atm, the Gibbs energy change per mole
upon changing the pressure from P to P is just RT ln P.
Reactions in solution. It is customary in books
on thermodynamics to develop most of the important
thermodynamic equations as applied to a perfect gas,
but we will move at this point to a consideration of
biochemical substances in solution. Biochemists are
usually interested in the behavior of substances dis-
solved in relatively dilute aqueous solutions but also
in cytoplasm, in which some concentrations may be
very high. Sometimes the interest may be in nonaque-
ous solutions. In any case, it is necessary to establish
a standard state for the solute. The standard state of a
substance in aqueous solution is customarily taken as
a strictly hypothetical one molal solution (one mole of
solute per kilogram of water) whose properties are those
of a solute at infinite dilution. An equation exactly anal-
ogous to Eq. 6-23 can be written relating the Gibbs
energy of one mole of dissolved solute G
i
to the Gibbs
energy G
i
in the hypothetical standard state of unit
activity and to the activity a
i
of the solute (Eq. 6-24).
G G RT a
i i i
= + ln
Here the subscript i designates a particular component
in a solution which also contains solvent and, perhaps,
other components. To be precise, G
i
is a partial molar
Gibbs energy, i.e., the changes in total Gibbs energy of
a very large volume of solution when one mole of the
component is added.
From Eq. 6-24 it follows that the Gibbs energy change
for dilution from one activity a
1
to another a
2
is:
G a a RT a a ( ) ln ( ) dilution from to
1 2 2 1
=
Equation 6-24 and the equations that follow from it
apply to molal activities. However, the concentration
can be substituted for activity in very dilute solution
where the behavior of the dissolved molecules approx-
imates that of the hypothetical ideal solution for which
the standard state is defined. For any real solution, the
activity can be expressed as the product of an activity
coefficient and the concentration (Eq. 6-26).
a c =
where a = activity, = activity coefficient, and c = molal
concentration. Thus, to use the tabulations of thermo-
dynamic functions for substances in solution to predict
behavior in other than very dilute solution, we must
multiply the concentration of every component by the
appropriate activity coefficient. For the approximate
calculations which are often of interest to biochemists,
it is customary to equate concentration with activity.
Furthermore, in dilute solutions, the more usual molar
concentrations (moles per liter) are nearly equal to molal
concentrations. The same equations can be used with
mole fractions rather than molal concentrations.
G and the equilibrium constant. Consider
the following generalized chemical equation (Eq. 6-27)
for reaction of a moles of A with b moles of B to give
products C and D, etc.
a A + b B +
. . .
= c C + d D
. . .
The standard Gibbs energy change G for the process
is given by Eq. 6-28:
G = c G(C) + d G(D) +
. . .
a G(A) b G(B)
. . .
The symbol G (A) designates the Gibbs energy of
A, etc. The value of G for any desired concentrations
of reactants or products can be related to this G by
applying to each component Eq. 6-24 with the follow-
ing result:
Here a
C
represents the activity of component C, etc.
This useful equation permits us to calculate G for
the low concentrations usually found in biochemical
systems. These are more often in the millimolar range
or less rather than approaching the hypothetical 1 M of
the standard state. Often concentrations are substituted
in Eq. 6-29 for activities:
G G RT
c d
a b
+ ln
[ ] [ ]
[ ] [ ]
C D
A B
Equation 6-29 is used in another way by noting that
G = 0 when a system is at equilibrium and that at
equilibrium the product a
C
c
a
D
d
. . ./a
A
a
a
B
b
. . . is just the
equilibrium constant K. It follows that
G RT K RT K
T K
K
K
= =
=
=
=

ln . log
. log
. log
. log
2 303
19 145
5 708 25
1 364 25
1
1
1
J mol
kJ mol at C
kcal mol at C
Although the units of G are kJ mol
1
, the Gibbs energy
change in Eq. 6-31 is that for the reactions of a moles of
A. Thermodynamics
(6-24)
(6-25)
(6-29)
(6-30)
(6-31)
(6-26)
G = G + RT ln
a
C
c
a
D
d
. . .
a
A
a
a
B
b
. . .
288 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-35)
(6-33)
(6-34)
A, b moles of B, etc., as in the equation used to define
K (Eq. 6-27 in this instance). It is also important to
realize that the log term in Eq. 6-31 must be unitless.
Although we usually write ln K or log K, K here repre-
sents K/Q, where Q = 1 because it has the same form
as K but with all components in their standard states.
Since the units of K and Q are the same, log K is unit-
less. Similar considerations apply to expressions of K
in exponential form.
Activity coefficients and concentration
equilibrium constants. Strictly speaking, Eq. 6-31
applies only to thermodynamic equilibrium constants
that is, to constants that employ activities rather than
concentrations. The experimental determination of
such constants requires measurements of the apparent
equilibrium constant or concentration equilibrium
constant
21
K
c
at a series of different concentrations
and extrapolation to infinite dilution (Eq. 6-32).
K
c
c d
a b
=
=
concentration equilibrium constant
C D
A B
at equilibrium
[ ] [ ]
[ ] [ ]
Extrapolation of K
c
to infinite dilution to give K is
usually easy because the activity coefficients of most
ionic substances vary in a regular manner with ionic
strength and follow the DebyeHckel equation
(Eq. 6-33) in very dilute solutions (ionic strength < 0.01).
The integers z
1
and z
2
are the numbers of charges
(valences) for the cation and anion of the salt. The
ionic strength (, or I) is evaluated as follows:
Here c
i
are the molar concentrations of the ions. The
summation is carried out over all the ions present. The
activity coefficient (Eq. 6-33) is the mean activity
coefficient for both the cation and anion.
Equation 6-33 suggests that extrapolation of equi-
librium constants to infinite dilution is done appropri-
ately by plotting log K
c
vs . For example, Fig. 6-1
shows plots of pK
a
for dissociation of H
2
PO
4

, AMP

,
and ADP
2
, and ATP
3
vs . The variation of pK
a
with at low concentrations (Eq. 6-35) is derived by
application of the Debye-Hckel equation (Eq. 6-33):
Straight lines of slope 0.509 (z
A
2
z
HA
2
) are expected.
The observed (negative) slopes (Fig. 6-1) are ~ 1.5 for
H
2
PO
4

and AMP

, ~ 2.5 for ADP


2
, and ~ 3.5 for ATP
3
.
The data over the entire range of ionic strength are
fitted by empirical relationships of the type of Eq. 6-35a:
in which a and b are empirically determined constants.
For example, for H
2
PO
4

a = 1.52 and b = 1.96. The


value of pK
a
found was 7.18, about 0.22 greater than
the value at = 0.2, an ionic strength more commonly
used in the laboratory and close to that found in tissues.
Note that the difference between the extrapolated pK
a
for ATP
3
of 7.68 and the observed value of ~ 7.04 at =
0.2 is even greater. Serious errors can be introduced
into calculations by using extrapolated values for K for
solutions of appreciable ionic strength. The errors will
be maximal for ions of high charge type such as ATP
3
and ATP
4
.
(6-32)
(6-35a)
Another problem with equilibrium constants for
reactions that use or produce hydrogen ions is that
there is no rigorous relationship between pH and a
H
+
or [H
+
]. Indeed, the concept of an activity of a single
ion has little meaning in thermodynamics. Neverthe-
less, in the pH range of interest to biochemists, results
that are very close to those obtained by more rigorous
methods are achieved by assuming that the pH meter
Figure 6-1 The apparent pK
a
values for secondary ionizations
of AMP, ADP, ATP, and H
3
PO
4
(abbreviated ortho) plotted
against . Temperature: 25C. From R. C. Phillips et al.
22
7.8
7.6
7.4
7.2
7.0
6.8
6.6
6.4
6.2
0 10 20 30 40
25C
ATP
ortho
ADP
AMP
p
K
c
10
2

i
c
i
z
i
2 1
2
=
log = 0.509z
1
z
2

pK
a
= pK
a
0.509 (z
A
2
z
HA
2
)
pK
a
= pK
a
a b + for < 0.2

289
responds to hydrogen ion activity. The almost univer-
sal practice of biochemists is to assume the pH meter
reading obtained with a glass electrode equal to log
a
H
+ and to substitute the value of a
H
+ so obtained for
[H
+
] in defining the concentration equilibrium constant,
K
c
. It is also customary in most branches of chemistry
to use values of equilibrium constants and of Gibbs
energy which have not been extrapolated to = 0.
Thus most values of K and G, including many of those
in this book, are actually of K
c
and G
c
. An international
Commission on Biothermodynamics
21
recommended
that values of K
c
be measured with the lowest effective
buffer concentration and that the ionic strength be
brought to 0.1 with KCl.
Changes in equilibria with temperature. At
constant pressure G varies with absolute temperature
as follows:
d G T
dT
H T
( )
=
2
The corresponding variation in K is described by the
vant Hoff equation:
d K
dT
H
RT
d K
d T
H
R
H
d K
d T
ln
ln
( )
( )
. log
( )
=

=

=

2
1
10
1
0 01914
1
kJ mol
If H can be assumed constant over the temperature
range of an experiment, a plot of ln K vs 1/T provides
a convenient estimate of H (or H if ln K is plotted).
The slope of the line will be H/R. Since G can
be calculated from K, the method also permits evalua-
tion of S using Eq. 6-15. However, unless great care
is taken the method is of low accuracy
23
and it is prefera-
ble to establish H by direct calorimetry. Also, espe-
cially for proteins, the assumption that H is constant
over a significant temperature range may be erroneous.
From observations at only two temperatures, T
1
,
and T
2
, Eq. 6-37 becomes
H R K K T T T T =

ln ( )[ ( )]
2 1 1 2 2 1
1
kJ mol
6. Thermodynamics and Life Processes
Can thermodynamics be applied to living organ-
isms? Classical thermodynamics deals with equilibria,
but living beings are never in equilibrium. The laws
of thermodynamics are usually described as statistical
laws. How can such laws apply to living things, some
of which contain all of their genetic information in a
single molecule of DNA?
24,25
The ideal, reversible
reactions of classical thermodynamics occur at infini-
tesimal speeds. How can thermodynamics be applied
to the very rapid chemical reactions that take place in
organisms? One answer is that thermodynamics can
be used to decide whether or not a reaction is possible
under given conditions. Thus, if we know the steady-
state concentrations of reactants and products within
a cell, we can state whether a reaction will or will not
tend to go in a given direction.
We may still ask whether there are generalities
comparable to the laws of thermodynamics that apply
to the kind of steady state or dynamic equilibrium
that exists in organisms. Lars Onsager showed that
such relationships can be found for conditions that are
near equilibrium. Ilya Prigogine and associates extended
Onsagers findings and showed that under conditions
that are far from equilibrium the system tends to become
unstable and to spontaneously develop new structures,
which Prigogine calls dissipative structures.
26 28
Vortices in flowing water, tornados and hurricanes are
examples of dissipative structures. The maintenance
of dissipative structures depends upon a flow of energy
and matter through the system. The flow of energy is
sufficient to organize a system. Thus, if a flask of
water is placed on a hot plate, a cycle is established.
The water moves via cyclic convection currents that
develop as a result of the flow of energy through the
system. Morowitz reckoned that the 6 3 x 10
18
kJ/year
of solar energy that falls on the earth supplies the
organizing principle for life.
28
Just as it drives the great
cycles within the atmosphere and within the seas, it
gives rise to the branching and interconnecting cycles
of metabolism. This idea may even make the sponta-
neous development of the organized systems that we
call life from inanimate precursors through evolution
seem a little more understandable.
A characteristic of this nonequilibrium or irrever-
sible thermodynamics is that time is explicitly intro-
duced. Furthermore, open systems, in which materials
and energy flow into and out of the system, are con-
sidered. Clearly, a living organism is an open system
not a closed one of classical thermodynamics. Because
of the flow of materials concentration gradients are set
up and transport phenomena often become of primary
importance. Articles and books that provide an intro-
duction to nonequilibrium thermodynamics and to the
literature in the field include the following.
10,26,28 34
Whether these methods can be applied in a practical
way to metabolic systems has been debated.
35,36
A. Thermodynamics
(6-36)
(6-37)
or
or
(6-37a)
290 Chapter 6. Thermodynamics and Biochemical Equilibria
Acetaldehyde C
2
H
4
O 139.7 1123.5 171.5 28.3 10
Acetic acid C
2
H
4
O
2
369.4 866.8 169.2 9.3 8
Acetate

369.2 894.0 142.0 22.1 8


Acetyl-CoA 374.1* 889.1* 146.9* 13.0* 8
Acetyl-P 1218.4 901.7 134.3 25.6 8
Acetylene
c
C
2
H
2
209.2 1235.2 59.8 140.0 10
Acetoacetate

C
4
H
5
O
3

493.7 1795.4 276.5 3.2 16


Acetone C
3
H
6
O 161.2 1733.6 338.4 18.7 16
cis-Aconitate
3
C
6
H
3
O
6
3
920.9 2157.0 173.9 65.8 18
L-Alanine C
3
H
7
O
2
N 371.3 1642.0 300.4 0.8 15
L-Asparagine C
4
H
8
O
3
N
2
526.6 1999.7 331.2 28.4 18
L-Aspartate

C
4
H
6
O
4
N

700.7 1707.0 235.4 24.3 15


n-Butanol C
4
H
10
O 171.8 2591.7 516.2 36.7 24
n-Butyric acid C
4
H
8
O
2
380.2 2146.1 443.8 44.2 20
n-Butyrate

C
4
H
7
O
2

352.6 2173.7 416.2 56.6 20


Butyryl-CoA 357.5* 2168.8* 421.1* 21.5* 20
Caproate

C
6
H
11
O
2

329.7 3459.7 684.1 84.7 32


CO
2
(g) 394.4 0.0 0.0 0.0 0
CO
2
(aq) 386.2 8.2 8.2 8.2 0
HCO
3

587.1 44.5 44.5 4.6 0


CO (g) 137.3 257.1 1.9 38.1 2
Citrate
3
C
6
H
5
O
7
3
1166.6 2148.4 182.5 57.3 18
Creatine C
4
H
9
O
2
N
3
264.3 2380.6 338.8 80.8 21
Creatinine C
4
H
7
ON
3
28.9 2378.8 340.6 79.0 21
Crotonate

C
4
H
5
O
2

275.7 2013.4 317.5 2.1 18


Cysteine C
3
H
7
O
2
NS 339.8 2178.3 541.1 121.5 21
Cystine C
6
H
12
O
4
N
2
S
2
665.3 4133.8 1046.0 246.9 40
Dihydroxyacetone-P
d
1293.2 1458.4 95.5 144.2 12
Erythrose 4-P
d
1439.1 1944.1 127.8 191.9 16
Ethanol C
2
H
6
O 181.5 1318.8 235.1 4.6 12
Ethylene (g)
c
C
2
H
4
O 68.1 1331.3 222.7 17.1 12
Formaldehyde CH
2
O 130.5 501.0 16.9 63.0 4
Formic acid CH
2
O
2
356.1 275.5 16.5 56.5 2
Formate

CHO
2

350.6 281.0 22.0 22.0 2


Fructose C
6
H
12
O
6
915.4 2874.1 233.8 245.7 24
Fructose 6-P
d
1758.3 2888.1 219.8 259.7 24
Fructose di-P
d
2600.8 2902.5 205.4 274.1 24
Fumaric acid C
4
H
4
O
4
647.1 1404.8 149.2 90.6 12
Fumarate

C
4
H
3
O
4

604.2 1447.7 106.2 93.6 12


-D-Galactose C
6
H
12
O
6
923.5 2865.9 242.0 237.5 24
-D-Glucose C
6
H
12
O
6
917.2 2872.2 235.6 243.8 24
Glucose 6-P 1760.3 2886.0 221.8 257.6 24
L-Glutamate

C
5
H
8
O
4
N

696.8 2342.5 376.9 2.7 21


L-Glutamine C
5
H
10
O
3
N
2
524.8 2633.1 474.8 4.7 24
3-P glycerate
d
1515.7 1235.9 59.0 100.8 10
2-P glycerate
d
1509.9 1241.8 53.2 106.6 10
Glyceraldehyde 3-P
d
1285.6 1466.0 87.9 151.8 12
Glycerol C
3
H
8
O
3
488.5 1643.4 169.5 110.2 14
Glycerol-P 1336.2 1652.6 160.3 119.3 14
Glycine C
2
H
5
O
2
N 373.5 1008.3 157.2 22.6 9
Glycogen C
6
H
10
O
5
665.3 2887.0 220.9 258.6 24
Glycolate

C
2
H
3
O
3

523.4 739.7 37.2 42.7 6


Glyoxylate

C
2
HO
3

461.1 564.9 46.9 86.9 4


H
2
O (l) 237.2 0.0 0.0 0.0 0
OH

157.3 79.9 79.9 39.9 0


H
+
0.0 0.0 0.0 0
H
2
(g) 0.0 237.2 21.8 18.2 2
H
2
O
2
136.8 100.4 359.4 319.4 2
H
2
S 27.4 714.6 321.3 161.5 8
HS

12.6 754.5 281.4 161.5 8


-Hydroxybutyric acid C
4
H
8
O
3
531.4 1994.9 336.0 23.6 18
-Hydroxybutyrate

C
4
H
7
O
3

506.3 2020.0 310.9 8.8 18


Hydroxypyruvate C
3
H
4
O
4
615.9 1041.6 5.7 165.5 8
TABLE 6-4
Gibbs Energies of Formation and of Oxidation at 25C for Compounds of Biochemical Interest
a,b
For oxidation by NAD
+
Compound Formula
G
f

(kJ mol
1
)
G
c

(kJ mol
1
)
G
ox
(kJ mol
1
)
G
ox
(pH 7)
(kJ mol
1
)
Number of
electrons
291
Hypoxanthine C
5
H
6
O 89.5 2773.0 334.8 144.6 24
Isocitrate
3
C
6
H
5
O
7
3
1160.0 2155.1 175.8 63.9 18
-Ketoglutarate
2
C
5
H
4
O
5
2
798.0 1885.5 186.4 53.3 16
Lactate

C
3
H
5
O
3

516.6 1378.1 175.9 23.9 12


-Lactose C
12
H
22
O
11
1515.2 5826.5 389.3 569.7 48
L-Leucine C
6
H
13
O
2
N 356.3 3551.7 721.6 62.3 33
Mannitol C
6
H
14
O
6
942.6 3084.0 282.8 236.6 26
Malate
2
C
4
H
4
O
5
2
845.1 1444.0 109.9 49.9 12
Methane (g) CH
4
50.8 818.0 218.0 58.2 8
Methanol CH
4
O 175.2 693.5 83.4 36.4 6
NH
4
+
79.5 276.3 112.2 12.3 3
NO
2

34.5 84.1 472.6 372.7 3


NO (g) 86.7 86.7 345.7 305.7 2
NO
3

110.5 8.1 655.6 515.7 5


Oxalate
2
C
2
O
4
2
674.9 351.1 92.1 52.1 2
Oxaloacetate
2
C
4
H
2
O
5
2
797.2 1254.7 40.2 79.7 10
H
3
PO
4
(aq)
e
1147.3 0.0 0.0 0.0 0
H
2
PO
4

(aq)
e
1135.1 12.1 12.1 27.8 0
HPO
4
2
(aq)
e
1094.1 53.1 53.1 26.8 0
n-Propanol C
3
H
8
O 175.8 1956.1 374.8 15.2 18
Isopropanol C
3
H
8
O 185.9 1946.0 384.9 25.3 18
Propionate

C
3
H
5
O
2

360.0 1534.7 278.2 38.5 14


Pyruvate

C
3
H
3
O
3

474.5 1183.1 111.9 47.9 10


Phosphoenolpyruvate
3
1269.5 1245.0 50.0 109.8 10
Ribose 5-P
d
1599.9 2414.9 175.0 224.6 20
Ribulose 5-P
d
1597.6 2417.1 172.8 226.8 20
Sedoheptulose 7-P
d
1913.3 3364.6 261.2 298.2 28
Sedoheptulose di-P
d
2755.8 3379.0 246.9 312.5 28
Sorbitol C
6
H
14
O
6
942.7 3083.9 282.9 236.5 26
Succinate
2
C
4
H
4
O
4
2-
690.2 1598.9 214.1 14.3 14
Succinyl-CoA 686.7* 1602.4* 210.6* 29.2* 14
Sucrose C
12
H
22
O
11
1551.8 5789.9 425.9 533.1 48
SO
4
2
742.0 0.0 0.0 79.9 0
SO
3
2
497.1 244.9 14.1 54.0 2
S
2
O
3
2
513.4 733.4 302.5 222.6 8
L-Threonine C
4
H
9
O
3
N 514.6 2130.3 330.1 49.5 19
L-Tyrosine C
9
H
11
O
3
N 387.2 4466.8 842.5 23.4 41
Urea CH
4
ON
2
203.8 664.9 112.0 7.8 6
Uric acid C
5
H
4
O
3
N
4
356.9 2089.4 241.5 118.1 18
L-Valine C
5
H
11
O
2
N 360.0 2916.5 579.9 40.5 27
Xanthine C
5
H
5
O
2
N
4
139.3 2425.6 293.8 125.7 21
D-Xylulose C
5
H
10
O
5
748.1 2409.8 180.1 219.5 20
a
The quantities tabulated are G
f
, the standard free energy of formation from the elements; G
c
, the standard free energy of combustion;
G
ox
, the standard free energy of oxidation by NAD
+
to products NADH + H
+
, CO
2
, H
2
O, N
2
, HPO
4
2
, and SO
4
2
; G
ox
(pH 7), the apparent
standard free energy change at pH 7. All values are in kJ mol
1
at 25C in aqueous solution unless indicated otherwise. If a compound is
designated (g) the values are for the gaseous phase at 1 atm pressure. The number of electrons involved in complete oxidation to CO
2
,
H
2
O, N
2
, and H
2
SO
4
is given in the final column. If this number is negative, the compound must be reduced to obtain the products, e.g.,
2 NO
3

+ 10 e

+ 12 H
+
N
2
+ 6 H
2
0. The data for phosphate esters refer to the compounds with completely dissociated phosphate groups
( O PO
3
2
). The values of G
f
for many of these compounds were calculated as G
f
(nonphosphorylated compound) G for hydrolysis
(to HPO
4
2
, Table 6-6) G
f
for H
2
O (one molecule for each phosphate ester formed) + G
f
for HPO
4
2
(from this table). Data from Bassman
and Krause
d
were used directly. For acyl-CoA derivatives CoA ( SH) is treated as an element, i.e., the values of G
f
given and designated
with an asterisk (*) are for formation from the elements plus free CoA. The values of G
c
and G
ox
are for oxidation to the usual products
plus CoA. Values of G of hydrolysis (Table 6-6) were used in computing G
f
for each of these compounds from that of the corresponding
alcohol or carboxylate anion. Another source containing an extensive table of Gibbs energy values is Wilhoit, R. C. (1969) in Biochemical
Microcalorimetry (Brown, H. D. ed.), pp. 305 317. Academic Press, New York
b
The major source is Long, C., ed. (1961) Biochemists Handbook, pp. 90 92. Van Nostrand, Reinhold, Princeton, New Jersey. Most of the
values in this collection are from Burton, K. (1957) Ergeb. Physiol., Biol. Chem. Exp. Pharmakol. 49, 275 298
c
From Stull, D.R., Westrum, E. F., Jr., and Sinke, G. C. (1969) The Chemical Thermodynamics of Organic Compounds. Wiley, New York
d
Bassham, J. A. and Krause, G. H. (1969) Biochim. Biophys. Acta. 189, 207 221
e
Van Wazer, J. R. (1958) Phosphorus and Its Compounds, Vol. I, p. 889. Wiley (Interscience), New York
TABLE 6-4
(continued)
For oxidation by NAD
+
Compound Formula
G
f

(kJ mol
1
)
G
c

(kJ mol
1
)
G
ox
(kJ mol
1
)
G
ox
(pH 7)
(kJ mol
1
)
Number of
electrons
A. Thermodynamics
292 Chapter 6. Thermodynamics and Biochemical Equilibria
B. Tables of G Values for Biochemical
Compounds
1. Gibbs Energies of Formation
Table 6-4 gives, in the first column, standard values
of Gibbs energies of formation from the elements G
f

for a variety of pure solids, gases, and liquids as well


as values for substances in solution at the hypothetical
1 M activity. As an example, consider the value of G
f

for pure liquid acetic acid, 389.1 kJ mol


1
. The equa-
tion for its formation from the elements is:
2 2 1
1
389 1
2
2 2 4 2
1
C s H g, atm
O g, atm C H O l
kJ mol
f
( ) ( )
( ) ( )
.
+
+
=

G
To obtain the Gibbs energy of formation in aqueous
solution, we must have solubility data as well as activity
coefficients of acetic acid at various concentrations.
From these data the change in Gibbs energy for solu-
tion of the liquid acetic acid in water to give aqueous
acetic acid in the hypothetical 1 molal standard state
(Eq. 6-39) can be obtained.
Acetic acid l acetic acid aq
kJ mol
( ) ( )
.

=

G 7 3
1
Summing Eqs. 6-38 and 6-39 we obtain:
2 2
396 4
2 2
1
C H O acetic acid aq
kJ mol
f
+ +
=

( )
. G
In many computations it is convenient to have G
values for single ions, e.g., for acetate

. We can obtain
G
f
of acetate

(aq) from that of acetic acid (aq) by


making use of G of dissociation (Eq. 6-41).
Acetic acid H acetate
p
kJ mol at C
a a
+
= =
= +
+

G K K 5 708 5 708
27 2 25
1
. log .
.
By convention we define the Gibbs energy of formation of
H
+
as zero. Then, by summing Eqs. 6-40 and 6-41 we
obtain G
f
of acetate

= 369.2 kJ mol
1
.
2. Gibbs Energies of Dissociation of Protons
Table 6-5 gives thermodynamic dissociation con-
stants and values of G and H for a number of acids
of interest in biochemistry. Some of these values were
used in obtaining the values of G
f
for the ions of
Table 6-4. The data of Table 6-5 can also be used in
evaluation of Gibbs energy changes for reactions of
ionic forms not given in Table 6-4.
3. Group Transfer Potentials
Recall that the equilibria for reactions by which
monomers are linked to form biopolymers (whether
amides, esters, phosphodiesters, or glycosides) usually
favor hydrolysis rather than formation (condensation).
The equilibrium positions depend on the exact struc-
tures. Some linkages are formed easily if monomer
concentrations are high enough, but others are never
formed in significant concentrations. Likewise, hydro-
lysis may be partial at equilibrium or it may be 99.9%
or more complete.
Let us compare the hydrolysis of the two organic
phosphates adenosine triphosphate (ATP) and
glucose 6-phosphate (Eqs. 6-42 and 6-43).
HATP H O HADP H PO
kJ mol
kJ mol at C
3
2
2
2 4
1
1
32 9
22 6 25 0 25

+ +

=

G
H
.
. , .
- -Glucose -phosphate H O
glucose H PO
kJ mol at C
D 6
16 4 25
2
2 4
1

+
+
= G .
The decrease in Gibbs energy upon hydrolysis is
twice as large for ATP as it is for glucose 6-phosphate.
Glucose phosphate is thermodynamically more stable
than ATP. It would be easier to form than would ATP
by a reversal of the hydrolysis reaction and also easier
to form biosynthetically. From the Gibbs energies of
hydrolysis it follows that a phospho group could be
transferred spontaneously from ATP to glucose in the
presence of a suitable catalyst but not vice versa.
Because it reflects quantitatively the thermodynamic
tendency for a group to be transferred to another
(6-38)
(6-39)
(6-40)
(6-41)
(6-42)
(6-43)
P O
OH
O

Phospho (phosphoryl) group


293
TABLE 6-5
Values of pK
a
, G, and H for Ionization of Acids at 25C
a,b
Acid pK
a
G (kJ mol
1
) H (kJ mol
1
)
a
These are thermodynamic values (infinite dilution) except for those labeled apparent.
The latter apply at an ionic strength of 0.2 0.25.
b
Most data are from Jencks, W. P. and Regenstein, J. (1976) in Handbook of Biochemistry
and Molecular Biology, 3rd ed., Vol. I (Fasman, G. D. ed.), pp. 305 351, CRC Press,
Cleveland, Ohio.
c
Here, pK
1
is for K
1
= [H
+
][HCO
3

] / [CO
2
] + [H
2
CO
3
]. From Forster, R. E., Edsall, J. T.,
Otis, A. B., and Roughton, F. J. W., eds. (1969) NASA Spec. Publ. 188.
d
From Phillips, R. C., George, P., and Rutman, R. J. (1963) Biochemistry 2, 501 508.
e
From Alberty, R. A. (1969) J. Biol. Chem. 244, 3290 3302.
f
Values used by Alberty, R. A. (1972) Horizons of Bioenergetics, pp. 135 147, Academic
Press, New York, calculated for 0.2 ionic strength from equations of Phillips, R. C.,
George, P., and Rutman, R. J. (1966) J. Am. Chem. Soc. 88, 2631 2640.
Formic acid 3.75 21.4 0.04
Acetic acid 4.76 27.2 0.1
Propionic acid 4.87 27.8 0.6
Lactic acid 3.97 (35C) 23.4 2.2
Pyruvic acid 2.49 14.2 12.1
NH
4
+
9.25 52.8 52.2
CH
3
NH
4
+
10.59 60.4 55.4
Alanine
COOH 2.35 13.4 3.1
NH
3
+
9.83 56.1 45.4
-Alanine
COOH 3.55 20.3 4.5
NH
3
+
(apparent) 10.19 58.2
L-Alanyl-L-alanine 3.34 19.1 0.5
Aspartic acid
COOH 2.05 11.7 7.7
COOH 3.87 22.1 4.0
NH
3
+
10.60 60.5 38.8
H
2
CO
3
, pK
1
6.35
c
36.2 9.4
pK
2
10.33 59.0 15.1
H
3
PO
4
, pK
1
2.12 12.1 7.9
pK
2
7.18
d
41.0 3.8
(apparent) 6.78
e
38.7 3.3
pK
3
12.40 70.8 17.6
Glycerol 1-phosphate, pK
2
6.66 38.0 3.1
Glucose 6-phosphate 6.50 37.1 1.8
Pyrophosphoric acid, H
4
P
2
O
7
pK
3
6.7 38.1 1.3
(apparent) 6.12
e
34.9 0.5
pK
4
9.4 53.6 7.1
(apparent) 8.95
e
51.2 1.7
Adenosine 3.5 20.1 13.0
AMP
pK
1
(ring, apparent) 3.74
e
21.3 4.2
pK
2
(phosphate) 6.67
d
38.1 3.6
(apparent) 6.45
e
36.8 3.6
ADP
pK
2
(ring, apparent) 3.93
e
22.4 4.2
pK
3
(diphosphate) 7.20
d
41.1 5.7
(apparent) 6.83
f
39.0 5.7
ATP
pK
3
(ring, apparent) 4.06 23.2 0
pK
4
(triphosphate) 7.68
d
43.8 7.0
(apparent) 7.06
d
40.2 7.0
Pyridine 5.17 29.5 20.1
Phenol 9.98 56.9 23.6
nucleophile (see Chapter 12), the
Gibbs energy decrease (G) upon
hydrolysis is sometimes called the
group transfer potential. During
the hydrolysis of ATP (Eq. 6-42) the
phospho group of ATP is transferred
to a hydroxyl ion from water with
a value G = 32.9 kJ mol
1.
The
group transfer potential of this
phospho group is 32.9 kJ/mol and
that of the phospho group of glucose
6-phosphate is only 27.6 kJ mol
1
.
While the choice of water as the
reference nucleophile for expression
of the group transfer potential is
somewhat arbitrary, it is customary.
Transfer of groups is important in
energy metabolism and in biosyn-
thesis of polymers. Gibbs energies
of hydrolysis are given in Table 6-6
for several compounds.
4. Constants That Vary with
pH and Magnesium Ion
Concentrations
Equation 6-42 is written for
hydrolysis of HATP
3
to HADP
2
+
H
2
PO
4

, a stoichiometry that applies


well in the pH range around 6. How-
ever, at a pH above ~ 7 most of the
ATP is in the form ATP
4
and is
cleaved to HPO
4
2
according to Eq.
6-44.
ATP
4
+ H
2
O ADP
3
+ HPO
4
2
+ H
+
G + 5.4 kJ mol
1
at 25C, = 0.25
H 19.7 kJ mol
1
The value of G = + 5.4 kJ mol
1
for this reaction is hardly the large
negative number expected for a
highly spontaneous reaction. What
is the matter? The problem is that
H
+
is produced and that the stan-
dard state of H
+
is 1 M, not 10
7
M.
Because of this, biochemists often
prefer to use another kind of appar-
ent dissociation constant and an
apparent G such that the standard
state of H
+
is taken as that of the pH
at which the experiments were done,
usually pH 7. The symbol K has
often be used and is used in this
book to represent the following
B. Tables of G Values for Biochemical Compounds
(6-44)
294 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-46)
pH-dependent equilibrium constant (Eq. 6-45) which
will be a constant only at a single pH.
If one proton is produced in the reaction as in Eq. 6-44,
the following relationship will hold.
G = G 5.708 x pH kJ mol
1
at 25C
Note that G = RT ln K and that [H
+
] does not
appear in the expression for K given by Eq. 6-45.
From the value G = + 5.4 kJ mol
1
and applying Eq.
=

K
[ ][ ]
[ ]
ADP HPO
ATP
3
4
2
4
(6-45)
TABLE 6-6
Gibbs Energies of Hydrolysis at 25C (in kJ mol
-1
)
a
a
Unless indicated otherwise, the values are based on tables from Jencks, W. P. (1976) Handbook of Biochemistry and Molecular Biology, 3rd ed.,
Vol I (Fasman,G.D. ed.), pp. 296304. CRC Press, Cleveland, Ohio. For a reaction producing one proton at pH 7 G = 39.96 kJ mol
1.
b
Guynn, R.W. and Veech, R.L. (1973) J. Biol. Chem. 248, 69666972.
c
Based on +11.80 kcal mol
1
for hydrolysis to P
2
O
7
4
plus G of dissociation of HP
2
O
7
3
as quoted by Alberty, R. A. (1969) J.Biol.Chem. 244, 3290
3324. However, 1.017 kcal mol
1
was added to the value of 11.80 to make it consistent with that for hydrolysis of ATP to ADP. Reevaluation by
Frey and Arabshahi (1995) Biochemistry 34, 1130711310, indicates that G (pH 7) for hydrolysis of ATP to AMP + PP
i
is ~ 10 kJ mol
1
more
negative than is shown here.
d
Alberty, R.A. (1972) Horizons of Bioenergetics, Academic Press, New York, pp. 135147.
e
George, P. , Witonsky, R.J., Trachtman, M., Wu, C., Dorwart, W., Richman, L., Richman, W., Shurayh, F., and Lentz, B. (1970) Biochim. Biophys.
Acta. 223, 115.
f
Based on G

= 3.0 kcal mol


1
for acetyl phosphate + CoA acetyl-CoA + P
i
from Stadtman, E. R. (1973) The Enzymes, (Boyer, P.D., ed.), 3rd
ed., Vol. 8. pp. 149. Academic Press, New York, together with G (pH 7) for hydrolysis of acetyl-CoA.
g
Estimated from G for ATP hydrolysis + G = 19.9 kJ mol
1
for the 3-phosphoglycerate kinase reaction: Burton, K. and Krebs, H. A. (1953)
Biophys. J. 54, 94107; and (1955) Biophys. J. 59, 4446.
h
Estimated from data at pH 7.7 or 8.0 (tables of Jencks).
i
Guynn, R. W., Gelberg, H. J., and Veech, R. L. (1973) J. Biol. Chem. 248, 69576965 found G = 35.75 kJ mol
1
at 38C. Without data on H,
correction to 25C is difficult. Burton, K., (1955) Biophys. J. 59, 44 46, gave G (pH 7) for ATP
4
+ acetate

+ CoA ADP
3
+ acetyl-CoA +
HPO
4
2
as approximately zero at 25C. Guynn et al. found 0.56 kJ mol
1
at 38C. This same value (0.56 kJ) at 25C was assumed to obtain the
figure given here. This is equivalent to assuming H of hydrolysis as almost the same for ATP and acetyl-CoA, an unsupported assumption.
j
Assumed 2 kcal mol
1
more negative than that of acetyl-CoA as in tables of Jencks.
Compound Products G G(pH 7) H
H
2
N C
O
O

ATP
4
ADP
3
+ HPO
4
2
+ H
+
5.41
b
34.54 19.71
ATP
4
AMP
2
+ HP
2
O
7
3
+ H
+
2.54
c
37.4 19.0
MgATP
2
MgADP

+ HPO
4
2
+ H
+
16.0
d
24.0 14.2
ADP
3
AMP
2
+ HPO
4
2
+ H
+
3.67
c
36.3 13.5
AMP
2
Adenosine + HPO
4
2
9.6
e
9.6 0
ATP
4
Adenosine + HP
3
O
10
4
36.0
e
36.0 7.9
HP
2
O
7
3
2 HPO
4
2
+ H
+
6.54
c
33.4 12.6
Acetyl phosphate
2
Acetate

+ HPO
4
2
+ H
+
7.7
f
47.7
1,3-Diphosphoglycerate
4
3-Phosphoglycerate
3
+ HPO
4
2
+ H
+
14.5
g
54.5
Phosphoenolpyruvate
3
Pyruvate

+ HPO
4
2
61.9 61.9 25.1
Carbamoyl phosphate
2
+ HPO
4
2
+ H
+
11.5 51.5
Creatine phosphate

Creatine
+
+ HPO
4
2
43.1 43.1
Phosphoarginine

Arginine + HPO
4
2
38.1
h
(Mg
2+
present)
Glycerol phosphate
2
Glycerol + HPO
4
2
9.2 9.2
-D-Glucose 6-phosphate
2
-D-Glucose + HPO
4
2
13.8 13.8 2.5
Glucose 1-phosphate
2
Glucose + HPO
4
2
20.9 20.9
Maltose (or glycogen) 2-Glucose 16.7 16.7
Sucrose Glucose + fructose 29.3 29.3
UDP glucose
2
Glucose + UDP
3
+ H
+
9.4 30.5
N
10
-Formyltetrahydrofolic acid Formate

+ H
+
+ tetrahydrofolic acid 14.1 25.9
h
Acetic anhydride 2-Acetate

+ 2H
+
31.1 48.9
Acetyl-CoA Acetate

+ H
+
+ CoA 4.9 35.1
i
Succinyl-CoA

Succinate
2
+ H
+
+ CoA 3.5 43.5
j
Ethyl acetate Ethanol + acetate

+ H
+
20.2 19.7
Asparagine Aspartate

+ NH
4
+
15.1 15.1
Glycine ethyl ester
+
(39C) Glycine + ethanol + H
+
4.9 35.1
Valyl-tRNA
+
Valine + tRNA + H
+
4.9 35.1
295
6-46, we obtain for the hydrolysis of ATP at 25C,
= 0.25:
G (pH 7) = 34.5 kJ mol
1
= 8.26 kcal mol
1
An additional set of standard states is frequently
met in the biochemical literature. An equilibrium
constant, designated in this book as K

, is used to
relate the total concentrations of all ionic forms of the
components present at the pH of the experiment.
Thus,
BOX 6-A MEASUREMENT OF INTRACELLULAR pH
What is the pH within a cell? Is it constant or
does it vary with physiological conditions? Do all
cells operate at similar pH? The answers to these
important questions have been sought using a
variety of techniques.
a
Tiny microelectrodes with
tips only 1 m in diameter have been inserted into
cells. Indicator dyes have been diffused into cells
and either light absorption or fluorescence
b
mea-
sured. The distribution of a suitable radiolabeled
weak acid or weak base, such as [
14
C]methylamine,
that is able to permeate cells can be used to calculate
the difference between internal and external pH.
c
The activity of the enzyme carbonic anhydrase,
which is very pH sensitive, can be used to monitor
the pH of mitochrondia.
d
Since 1973 NMR methods
have been popular.
e l
The chemical shifts of
31
P in
inorganic phosphate (pK
a
= 6.9), ATP, glucose 6-
phosphate, and of other metabolites of
13
C in citrate
and bicarbonate,
l
give direct estimates of pH. How-
ever, caution must be exercised if internal pH values
good to 0.1 unit are to be obtained.
g
More sensitive
measurements over the pH range 1.3 9.1 can be
made by diffusing one of a series of aminophospho-
nates into cells and measuring the
31
P chemical
shift.
h
Fluorinated probes such as dimethylfluoro-
alanine (pK
a
= 7.3) are also useful because of their
low toxicity and high sensitivity of the
19
F NMR
signal. These alanine derivatives can be diffused
into cells as their methyl esters, which are rapidly
cleaved within cells, allowing the fluorinated amino
acids to accumulate.
i,m
The pH within cells appears to be tightly con-
trolled although small variations are sometimes
observed. Red blood cells, thymocytes, liver, skele-
tal muscles, and intact hearts all maintain a pH in
the range 7.0 7.3.
b,h,i
However, the pH can fall to 6.2
within 13 minutes of oxygen deprivation (ischemia)
and to 6.1 after exhaustive exercise.
n,o
The
31
P NMR
technique permits the monitoring of pH as well as
the state of the adenylate system (Section D) in human
limbs suffering from circulatory insufficiency.
c
In higher plants the pH of cytoplasm is 7.4 7.5
but vacuoles are acidic with a pH of 4.5 6.
l
The
cytoplasm of maize root tips has a pH of 7.1 but the
vacuoles are at a pH of 5.5.
f
The pH of granules of
the chromaffin cells of the adrenal cortex, which
accumulate high concentrations of ATP and
catecholamines, is also low, ~ 5.7.
p
The bacterium
Streptococcus faecalis maintains a higher internal pH
of ~ 8.0, even when the pH of the medium varies
from 6.5 to 8.0
q
while E. coli operates at pH 7.6, the
extremes of variation being 7.4 7.8 when the external
pH changes from 5.5 9.
r
Changes of internal pH during developmental
events such as fertilization of sea urchin eggs (+ 0.3
unit) have been recorded. However, the significance
of pH changes in metabolic regulation remains
uncertain.
c
a
Kotyk, A., and Slavik, J. (1989) Intracellular pH and its Measure-
ment, CRC Press, Boca Raton, Florida
b
Rogers, J., Hesketh, T. R., Smith, G. A., and Metcalfe, J. C. (1983)
J. Biol. Chem. 258, 59945997
c
Nuccitelli, R., and Deamer, D. W., eds. (1982) Intracellular pH:
Its Measurement, Regulation and Utilization in Cellular Functions,
Liss, New York
d
Dodgson, S. J., Forster, R. E., II, and Storey, B. T. (1982) J. Biol.
Chem. 257, 17051711
e
Moon, R. B., and Richards, J. H. (1973) J. Biol. Chem. 248, 7276
7278
f
Roberts, J. K. M., Ray, P. M., Wade-Jardetsky, N., and Jardetsky,
O. (1980) Nature (London) 283, 870872
g
Avison, M. J., Hetherington, H. P., and Shulman, R. G. (1986)
Ann. Rev. Biophys. Biophys. Chem. 15, 377402
h
Pietri, S., Miollan, M., Martel, S., Le Moigne, F., Blaive, B., and
Culcasi, M. (2000) J. Biol. Chem. 275, 1950519512
i
Taylor, J. S., and Deutsch, C. (1983) Biophys. J. 43, 261267
j
Bailey, I. A., Williams, S. R., Radda, G. K., and Gadian, D. G.
(1981) Biochem. J. 196, 171178
k
Barton, J. K., Den Hollander, J. A., Lee, T. M., MacLaughlin, A.,
and Shulman, R. G. (1980) Proc. Natl. Acad. Sci. U.S.A. 77, 2470
2473
l
Gout, E., Bligny, R., and Douce, R. (1992) J. Biol. Chem. 267,
1390313909
m
Deutsch, C. J., and Taylor, J. S. (1987) Ann. N.Y. Acad. Sci. 508, 33
n
Garlick, P. B., Radda, G. K., and Seeley, P. J. (1979) Biochem. J.
184, 547554
o
Pan, J. W., Hamm, J. R., Rothman, D. L., and Shulman, R. G.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 78367839
p
Pollard, H. B., Shindo, H., Creutz, C. E., Pazoles, C. J., and
Cohen, J. S. (1979) J. Biol. Chem. 254, 11701177
q
Kobayashi, H., Murakami, N., and Unemoto, T. (1982) J. Biol.
Chem. 257, 1324613252
r
Slonczewski, J. L., Rosen, B. P., Alger, J. R., and MacNab, R. M.
(1981) Proc. Natl. Acad. Sci. U.S.A. 78, 62716275
B. Tables of G Values for Biochemical Compounds
[ ][ ]
[ ]
ADP, all forms phosphate, all forms
ATP, all forms
(6-48)
(6-47)
K

=
296 Chapter 6. Thermodynamics and Biochemical Equilibria
and
G

= RT ln K

The Gibbs energy change G

can be related to G
by considering the relationship of K

to K. For ATP
hydrolysis in the pH range of 2 10, K

is given by Eq.
6-50.
In this equation K
HADP
2
, etc., are consecutive dissocia-
tion constants as given in Table 6-4. The expressions in
parentheses are the Michaelis pH functions, which
were considered in Chapter 3 (Eqs. 3-4 to 3-6). In Eq.
6-50 they relate the total concentration of each compo-
nent to the concentration of the most highly dissociated
form. Thus, for the pH range 2 10
[P
i
]
total
= [HPO
4
2
](1 + [H
+
] / K
H
2
PO
4
)
Using apparent pK
a
values ( = 0.2) for H
2
PO
4

, HADP
2
,
and HATP
3
of 6.78, 6.83, and 7.06 (Table 6-5) and taking
G at pH 7 as 34.5 kJ mol
1
, we compute G

= 35.0
kJ mol
1
at pH 7. The difference between G and G

in this case is small, but it would be larger if the ionic


forms in Eq. 6-44 were not the ones predominating at
pH 7.
To obtain the Gibbs energy change for a reaction
under other than standard conditions, Eq. 6-29 must be
applied. Thus, at pH 7 and 0.01 M activities of ADP
3
,
ATP
4
, and HPO
4
2
, G for hydrolysis of ATP according
to Eq. 6-44 is 34.5 (2 x 5.71) = 45.9 kJ mol
1
= 11.0
kcal mol
1
. We see that at concentrations existing in
cells (usually in the millimolar range) ATP has a sub-
stantially higher group transfer potential than under
standard conditions.
The reader should bear in mind that there is no
accepted standard usage of K and that K

is just for
this book! An international committee
21
has recom-
mended that K be used with the same meaning as K

in
this book and more changes may be coming (see the
next section).
To obtain G at a temperature other than 25C, we
must know H for the reaction. Using Eq. 6-37a it is
easy to show that G at temperature T
2
is related to
that at T
1
as follows:
The enthalpy of hydrolysis of ATP according to
Eq. 6-44 is approximately 19.7 kJ mol
1
. Using this
value and applying Eq. 6-52 we can calculate that G
(pH 7) for the hydrolysis of ATP at 38C is 35.2 kJ
mol
1
. The value of G (25C, pH 7), used in obtain-
ing this answer, is 35.54 kJ mol
1
which was computed
from the value of G (pH 7), 38C of 35.19 kJ mol
1
reported by Guynn and Veech
37
using Eq. 6-52. Bear
in mind that all of the foregoing Gibbs energy changes
are apparent values applying to solutions of ionic
strength ~ 0.25.
Both ADP and ATP as well as inorganic pyrophos-
phate form complexes with metal ions. Since the mag-
nesium complexes are often the predominant forms of
ADP and ATP under physiological conditions, we must
consider the following Gibbs energy changes. These
are apparent values for = 0.2 at 25C.
ATP
4
+ Mg
2 +
Mg ATP
2
G = 26.3; H = 13.8 kJ mol
1
ADP
3
+ Mg
2 +
Mg ADP

G = 19.8; H = 15.1 kJ mol


1
Combining the apparent G values for Eqs. 6-47, 6-53,
and 6-54, we obtain
MgATP
2
+ H
2
O MgADP

+ HPO
2
2
+ H
+
G (pH 7) = 28.0 kJ mol
1
The stoichiometry of Eq. 6-55 never holds exactly.
Some of the Mg
2 +
dissociates from the MgADP

; both
protons and Mg
2 +
bind to H
2
PO
4

; and HATP
3
and
HADP
2
are present and bind Mg
2 +
weakly.
38
Thus,
the observed value of G

varies with both pH and


magnesium concentration as well as with changes in
ionic strength. Tables and graphs showing the appar-
ent value of G

under various conditions have been


prepared by Alberty
38
and by Phillips et al.
39
An exam-
ple is shown in Fig. 6-2. From this graph we find that
G

for hydrolysis of ATP at pH 7, 25C, = 0.2, and


1 mM Mg
2 +
(a relatively high intracellular concentra-
tion
40
) is 30.35 kJ mol
1
( 7.25 kcal mol
1
).
Figure 6-2 was drawn using the equations of
Alberty, but the value of G (pH 7) of hydrolysis of
ATP = 34.54 kJ mol
1
at [Mg
2 +
] = 0 based on results
of Guynn and Veech
37
was used. The pK
a
values and
formation constants of Mg
2 +
complexes were those of
Alberty.
38
Note that George et al.
40
provided formation
constants of these complexes at infinite dilution where
the values of G of formation are considerably more
negative than those given in Eqs. 6-53 to 6-55.
From the foregoing considerations we see that
complexing with Mg
2 +
somewhat decreases the group
transfer potential of the phospho group of ATP. Fur-
thermore, changes in the concentration of free Mg
2 +
with time and between different regions of a cell may
K
K K K K
K K K
[ ] [ ] [ ]
[ ] [ ]
+ +

+ +

+ + +
+ +


1 1
1
2 2
2 4
2
3 3
2
2
2
2
H H H
H H
HADP HADP H ADP HPO
HATP HATP H ATP (6-50)
(6-49)
(6-51)


G
T G T T H
T
2
2 1 2 1
1

( )
(6-52)
(6-53)
(6-54)
(6-55)
K

=
297
40
44
48
52
36
32
28
24
10
1
0.1
0
4 5 6 7 8 9 10
pH
k
J

m
o
l

1
have significant effects.
41
While Mg
2 +
is a principal
cation in tissues, it is by no means the only one. Thus,
Ca
2 +
, Mn
2 +
, and even K
+
will affect equilibria involving
polyphosphates such as ATP.
It is not easy to measure the group transfer poten-
tial of ATP and published values vary greatly. George
et al.
40
reported G for Eq. 6-42 as 39.9 kJ mol
1
at
25C, = 0.2, and as 41.25 kJ mol
1
at infinite dilution.
They regarded these values as good to 4 kJ mol
1
.
However, most other estimates have been at least 4 kJ
mol
1
less negative.
42 44
The self-consistent set of thermo-
dynamic data used throughout this book are based in
part on the value of G for hydrolysis of ATP obtained
by Guynn and Veech.
37
5. A New Standard for Biochemical
Thermodynamics?
Because of the complexities of the equilibria in-
volved in biochemical reactions Alberty and others
45 47
and a Panel on Biochemical Thermodynamics
48
have
suggested that tables of thermodynamic properties for
biochemical use be tabulated for the following condi-
tions: T = 298.15 K, P = 1 bar (0.1 MP
a
0.987 atm),
pH = 7, pMg = 3 ([Mg
2+
] = 10
3
M), and ionic strength
I ( in this chapter) = 0.25 M. There appears to be both
advantages and disadvantages to this. The proponents
suggest that the symbols G, K, etc. be used for this
Figure 6-2 Plots of the apparent Gibbs energy G

for
hydrolysis of ATP as a function of pH at a series of different
concentrations of free magnesium ions. Millimolar [Mg
2 +
] is
indicated by the numbers by the curves. Computer-drawn
graphs courtesy of Carol M. Metzler.
new standard. To change all of the numbers in this
book, which uses a self-consistent set of thermodynamic
quantities, is impractical. However, it is worthwhile to
compare the value of Alberty and Goldberg for G
for hydrolysis of ATP to ADP + P
i
under the proposed
biochemical standard conditions with other values
given in this chapter, all at 25C.
G = 32.49 kJ mol
1
(new proposed biochemical
standard)
G

(pH 7) 35.0 kJ mol


1
(Eq. 6-50)
G (pH 7) 34.5 kJ mol
1
(for ATP
4
ADP
3
+
HPO4
2
; Eq. 6-47)
G (pH 7) 28.0 kJ mol
1
(for MgATP
2

MgADP

+ HPO4
2
; Eq. 6-55)
6. Bond Energies and Approximate Methods
for Estimation of Thermodynamic Data
For approximate estimation of enthalpy changes
during reactions, use can be made of empirical bond
energies (Table 6-7) which represent the approximate
enthalpy changes (H) for formation of compounds
in a gaseous state from atoms in the gas phase. Other
more comprehensive methods of approximation have
been developed.
49,50
7. Gibbs Energies of Combustion by O
2
and
by NAD
+
Since oxidation processes are so important in the
metabolism of aerobic organisms, it is often convenient
to discuss Gibbs energies of combustion. These are
easily derived from the Gibbs energies of formation.
For example, G
c
for acetate ion (aqueous) may be
obtained as follows:
Acetate

+ H
+
2 C + 2 H
2
+ O
2
G = + 369.2 kJ mol
1
= G
f
of acetate (from Table 6-3)
2 H
2
+ O
2
2 H
2
O
G = 2 x 237.2 = 474.4 kJ mol
1
= 2 x G
f
of H
2
O
2 C + 2 O
2
2 CO
2
G = 2 x 394.4 = 788.8 kJ mol
1
= 2 x G
f
of CO
2
Acetate

(1 M) + H
+
(1 M) + 2 O
2
(1 atm)
2 CO
2
(1 atm) + H
2
O (l)
G = 894.0 kJ mol
1
= G
c
of acetate

B. Tables of G Values for Biochemical Compounds


(6-56)
298 Chapter 6. Thermodynamics and Biochemical Equilibria
Benzene 155
Naphthalene 314
Styrene 155 + 21
Phenol 155 + 29
Benzaldehyde 155 + 17
Pyridine 180
Pyrrole 130
Indole 226
117
100
88
Urea 155
CO 439
CO
2
151
TABLE 6-7
Empirical Bond Energies and Resonance Energies
a
Energy values
for single
bonds
(kJ mol
1
)
Energy values
for multiple
bonds
(kJ mol
1
)
C=C 615
N=N 418
O=O 402 ( state)
C=N 615
C=O 686 (formaldehyde)
715 (aldehydes)
728 (ketones)
C=S 477
CC 812
NN 946 (N
2
)
CN 866 (HCN)
891 (nitriles)
Empirical
resonance
energy values
(kJ mol
1
)
HH 436
CC 346
Si Si 177
NN 161
OO 139
SS 213
CH 413
Si H 295
NH 391
PH 320
OH 463
SH 339
CSi 290
CN 292
CO 351
CS 259
CF 441
CCl 328
CBr 276
CI 240
Si O 369
a
From Pauling, L. ( 1960) The Nature of the Chemical Bond, 3rd ed., pp. 85, 189, and
195198, Cornell Univ. Press, Ithaca, New York
C OH
O
C OR
O
C NH
2
O
Table 6-4 lists values of G
c
as well as those of G
f
.
Besides CO
2
and H
2
O, the other products assumed in
calculating the values of G
c
in Table 6-4 are N
2
(1atm),
H
3
PO
4
(1 M), and H
2
SO
4
(1 M).
Much of the oxidation occurring in cells is carried
out by the biological oxidizing agent nicotinamide
adenine dinucleotide (NAD
+
) or by the closely related
NADP
+
(Chapter 10). It is convenient to tabulate values
of G for complete oxidation of compounds to CO
2
using NAD
+
rather than O
2
as the oxidizing agent.
These values, designated G
ox
and G
ox
(pH 7), are
also given in Table 6-3. Notice that these values are
relatively small, corresponding to the fact that little
energy is made available to cells by oxidation with
NAD
+
; for example (Eq. 6-57):
Acetate

+ 2 H
2
O + 4 NAD
+
2 CO
2
+ 4 NADH +
3 H
+
(10
7
M)
G
ox
(pH 7) = + 22.1 kJ mol
1
When the reduced NAD
+
(designated NADH) so formed
is reoxidized in mitochondria (Eq. 6-58), a large amount
of energy is made available to cells.
4 NADH + 4 H
+
+ 2 O
2
4 H
2
O + 4 NAD
+
G
ox
(pH 7) = 876.1 kJ mol
1
The sum of Eqs. 6-57 and 6-58 is the
equation for combustion of acetate
by O
2
(Eq. 6-59) and the two G
values sum to G
c
for acetate

.
Acetate

+ H
+
+ 2 O
2

2 CO
2
+ 2 H
2
O
G
c
= 894.0 kJ mol
1
G
c
(pH 7) = 854.0 kJ mol
1
The values of G
ox
(Table 6-4)
not only give an immediate indica-
tion of the relative amounts of energy
available from oxidation of substrate
with NAD
+
but also are very conve-
nient in evaluating G for fermenta-
tion reactions. For example, consider
the fermentation of glucose to ethanol
(Eq. 6-60):
-D-Glucose 2 CO
2
+ 2 ethanol
The Gibbs energy change G
(pH 7) for fermentation of glucose
to ethanol and CO
2
can be written
immediately from the data of Table
6-4 (Eq. 6-61).
G (pH 7) = 243.8 2 ( 4.6)
= 234.6 kJ mol
1
The values of G
ox
for H
2
O, CO
2
, and H
+
are always
zero and need not be considered. The same computa-
tion can be made using the awkwardly large values of
G
c
or using values of G
f
The latter are also large,
and CO
2
and water must be considered in the equa-
tions. Table 6-4 can be used to obtain values of G for
many metabolic reactions considered later in the book.
Data from any column in the table may be used for
this purpose, but for simplicity try the values in the
G
ox
column.
Note, however, that for reactions involving oxida-
tion by O
2
or by any oxidant, other than NAD
+
, not
appearing in Table 6-4, the following two-step proce-
dure is necessary if the G
ox
values are used. From the
G
ox
values compute G or G for the reaction under
consideration using NAD
+
as the oxidant. Then add
to this the Gibbs energy of oxidation by O
2
(or other
oxidant) of the appropriate number of moles of NADH
formed. The latter is given for O
2
in Table 6-8 and can
also be evaluated for a number of other oxidants, such
as Fe
3+
and cytochrome c, from the data in Table 6-8.
(6-57)
(6-58)
(6-59)
(6-60)
(6-61)
299
BOX 6-B MAGNESIUM
After potassium ion Mg
2+
is the most abundant
cation present in tissues
a,b
. The average adult ingests
~10 12 mmol of magnesium ion daily (~ 1/ 4 g). Of
this, about one-third is absorbed from the digestive
tract. An equivalent amount is excreted in the urine
to maintain homeostasis. Sixty percent of the mag-
nesium in the body is found in the bones. The total
Mg
2+
concentration in serum is ~ 1 mM, while vari-
ous tissues contain as much as 5 17 mM.
c
The con-
centration of free Mg
2+
is difficult to measure. A
variety of measurements using
31
P NMR spectroscopy
of ATP,
d f 19
F NMR of added fluorine-containing
chelators,
g
fluorescent chelators,
h
ion selective
electrodes,
c
and other indirect procedures
g,i,j
indicate
that the free magnesium concentration is almost the
same in extracellular fluids, cytosol, and mitochondria.
h
Most values have been about 0.5 mM, somewhat
less in erythrocytes. However, the most recent
estimates indicate an intracellular [Mg
2+
] of 0.8 1.1
mM.
c,f
The additional Mg
2+
present in cells is bound to
proteins, nuclei acids, and soluble compounds such
as ATP, ADP, citrate,
k
and other phosphate- and
carboxylate-containing substances. The binding is
reversible and equilibration is usually rapid. It has
been suggested that [Mg
2+
], like [H
+
], remains rela-
tively constant within cells and that these two ions
are in free equilibrium in the blood serum.
l
Never-
theless, there are instances in which at least tempo-
rary alterations in concentrations of both free Mg
2+
and H
+
occur.
m
During rapid catabolism of carbo-
hydrates the formation of lactic acid by glycolysis
leads to acidification of muscle cells, the pH falling
from 7.3 to as low as 6.3. This drop in pH causes a
large decrease in the extent of binding of Mg
2+
to
molecules such as ATP and to a transient increase
in [Mg
2+
]. Similarly, the release of bisphosphogly-
cerate from hemoglobin upon oxygenation leads to
a decreased concentration of free Mg
2+
as the latter
coordinates with bisphosphoglycerate.
n
The 0.25
mM [Mg
2+
] of aerobic red blood cells rises to 0.67
mM under anaerobic conditions.
d
Such changes in
the free Mg
2+
concentrations will affect many equi-
librium
o
and may be of significance in metabolic
regulation.
The magnesium ion has a smaller radius than
Ca
2+
, a fact that may account for its more ready
entry into cells. Mg
2+
can often be replaced by Mn
2+
with full activity for enzymes that require it. On the
other hand, high concentrations of Ca
2+
are often
antagonistic to Mg
2+
. This antagonism is clearly
seen in the effect of the two ions on irritability of
protozoa.
p
Both deficiency of magnesium and excess
of calcium in the surrounding medium cause in-
creased irritability. Excess magnesium leads to
anesthesia. The Mg
2+
concentration is high in hiber-
nating animals.
Over 300 enzymes are dependent upon Mg
2+
,
the largest single group being the phosphotrans-
ferases, for which MgATP complex may be regarded
as the substrate.
q
Magnesium has a special role in
photosynthesis as a component of chlorophyll.
One of the most toxic metals is beryllium. It
has been suggested that Be
2+
competes with Mg
2+
at many enzyme sites, including those of phospho-
glucomutase and of phosphatases. However, as
pointed out by Petsko,
r
because of its small size
beryllium tends not to form Be
2+
ions but, rather,
covalent complexes such as BeF
3
OH
2

, BeF
4
2
, and
MgADP-BF
2
(OH). In the latter complex beryllium
is covalently linked to the oxygen atom of the
phospho group of ADP to give an analog of MgATP,
which inhibits many enzymes. See for example, Fig.
19-16A, in which MgADPBeF
x
occupies the active
site of myosin.
a
Cowan, J. A., ed. (1995) The Biological Chemistry of Magnesium,
VCH Publ., New York
b
Strata, P., and Carbone, E., eds. (1991) Mg
2+
and Excitable
Membranes, Springer-Verlag, Berlin and New York
c
McGuigan, J. A. S., Blatter, L. A., and Buri, A. (1991) in Mg
2+
and
Excitable Membranes (Strata, P., and Carbone, E., eds), Springer-
Verlag, Berlin and New York
d
Gupta, R. K., and Moore, R. D. (1980) J. Biol. Chem. 255, 3987
3993
e
Garfinkel, L., and Garfinkel, D. (1984) Biochemistry 23, 3547
3552
f
Clarke, K., Kashiwaya, Y., King, M. T., Gates, D., Keon, C. A.,
Cross, H. R., Radda, G. K., and Veech, R. L. (1996) J. Biol. Chem.
271, 2114221150
g
Levy, L. A., Murphy, E., Raju, B., and London, R. E. (1988)
Biochemistry 27, 40414048
h
Jung, D. W., Apel, L., and Brierley, G. P. (1990) Biochemistry 29,
41214128
i Corkey, B. E., Duszynski, J., Rich, T. L., Matschinsky, B., and
Williamson, J. R. (1986) J. Biol. Chem. 261, 25672574
j
Magneson, G. R., Puvathingal, J. M., and Ray, W. J., Jr. (1987)
J. Biol. Chem. 262, 1114011148
k
Kwack, H., and Veech, R. L. (1992) Curr. Top. Cell. Regul. 33,
185207
l
Veloso, D., Guynn, R. W., Oskarrson, M., and Veech, R. L. (1973)
J. Biol. Chem. 248, 48114819
m
Purich, D. L., and Fromm, H. J. (1972) Curr. Top. Cell. Regul. 6,
131167
n
Bunn, H. F., Ransil, B. J., and Chao, A. (1971) J. Biol. Chem. 246,
52735279
o Cornell, N. W. (1979) J. Biol. Chem. 254, 65226527
p
Meli, J., and Bygrave, F. L. (1972) Biochem. J. 128, 415420
q
Vink, R., McIntosh, T. K., and Faden, A. I. (1991) in Mg
2+
and
Excitable Membranes (Strata, P., and Carbone, E., eds), Springer-
Verlag, Berlin and New York
r
Petsko, G. A. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 538540
B. Tables of G Values for Biochemical Compounds
300 Chapter 6. Thermodynamics and Biochemical Equilibria
C. Electrode Potentials and Gibbs Energy
Changes for OxidationReduction
Reactions
We live under a blanket of the powerful oxidant O
2
.
By cell respiration oxygen is reduced to H
2
O, which is a
very poor reductant. Toward the other end of the scale
of oxidizing strength lies the very weak oxidant H
+
,
which some bacteria are able to convert to the strong
reductant H
2
. The O
2
H
2
O and H
+
H
2
couples define
two biologically important oxidationreduction (redox)
systems. Lying between these two systems are a host
of other pairs of metabolically important substances
engaged in oxidationreduction reactions within cells.
There are two common methods for expressing the
oxidizing or reducing powers of redox couples in a
quantitative way. On the one hand, we can list values
of G for oxidation of the reduced form of a couple to
the oxidized form by O
2
. A compound with a large
value of G for this oxidation will be a good reductant.
An example is H
2
for which G of combustion at pH 7
(Table 6-4) is 237 kJ / mol. Poor reductants such as Fe
2 +
are characterized by small values of G of oxidation
( 8.5 kJ mol
1
for 2 Fe
2 +
2 Fe
3 +
). The Gibbs energies
of oxidation of biological hydrogen carriers, discussed
in Chapter 15, for the most part fall between those of
H
2
and Fe
2 +
.
A second way of expressing the same information
is to give electrode potentials (Table 6-8). Electrode
potentials are also important in that their direct mea-
surement sometimes provides an experimental
approach to the study of oxidationreduction reac-
tions within cells. To measure an electrode potential
it must be possible to reduce the oxidant of the couple
by flow of electrons (Eq. 6-62) from an electrode sur-
face, often of specially prepared platinum.
A + 2 H
+
+ 2 e

AH
2
Equation 6-62 represents a reversible reaction taking
place at a single electrode. A complete electrochemical
cell has two electrodes and the reaction occurring is
the sum of two half-reactions. The electrode potential
of a given half-reaction is obtained from the measured
electromotive force of a complete cell in which one
half-reaction is that of a standard reference electrode
of known potential. Figure 6-3 indicates schematically
an experimental setup for measurement of an electrode
potential. The standard hydrogen electrode consists of
platinum over which is bubbled hydrogen gas at one
atmosphere pressure. The electrode is immersed in a
solution containing hydrogen ions at unit activity (a
H
+
= 1). The potential of such an electrode is convention-
ally taken as zero. In practice it is more likely that the
reference electrode will be a calomel electrode or some
other electrode that has been established experimentally
as reliable and whose potential is accurately known.
The standard electrode is connected to the experi-
mental electrode compartment by an electrolyte-filled
bridge. In the experimental compartment the reaction
represented by Eq. 6-62 occurs at the surface of another
electrode (often platinum). The voltage difference
between the two electrodes is measured with a poten-
tiometer. The difference between the observed voltage
and that of the reference electrode gives the electrode
potential of the couple under investigation. It is im-
portant that the electrode reaction under study be
strictly reversible. When the electromotive force (emf)
of the experimental cell is balanced with the potenti-
ometer against an external voltage source, no current
flows through the cell. However, for a reversible
reaction a slight change in the applied voltage will
lead to current flow. The flow will be in either of the
two directions, depending upon whether the applied
voltage is raised or lowered.
Not all redox couples are reversible. This is espe-
cially true of organic compounds; for example, it is not
possible to determine readily the electrode potential
for an aldehyde alcohol couple. In some cases, e.g.,
with enzymes, a readily reducible dye with a potential
similar to that of the couple being measured can be
added. A list of suitable dyes has been described by
Dutton.
51
If the dye is able to rapidly exchange elec-
trons with the couple being studied, it is still possible
to measure the electrode potential directly. In many
cases electrode potentials appearing in tables have
been calculated from Gibbs energy data. The student
should be able to calculate many of the potentials in
Table 6-8 from Gibbs energy data from Table 6-4. If
A, H
+
, and AH
2
are all present at unit activity in the
experimental cell, the observed potential for the half-
reaction will be the standard electrode potential E.
If the emf of the hypothetical cell with the standard
hydrogen electrode is positive when electron flow is
in the direction indicated by the arrow in Fig. 6-3, the
potential of the couple A / AH
2
is also taken as positive
(and is often called a reduction potential). This is the
convention used in establishing Table 6-8, but poten-
tials of exactly the opposite sign (oxidation potentials)
are used by some chemists. To avoid confusion in
reading it is best to be familiar with values of one
or two potentials such as those of the O
2

H
2
O and
NAD
+
NADH couples.
When electrons flow in the external circuit the
maximum amount of work that can be done per mole
of electrochemical reaction (G) is given by Eq. 6-63
G = nEF = nE x 96.487 kJ mol
1
V
1
= nE x 23.061 kcal mol
1
V
1
where F equals the number of coulombs per mole
of electrons (Avogadros number multiplied by the
charge on the electron = 96,487 coulombs). E is mea-
sured in volts and represents the difference of the
(6-63)
(6-62)
301
(6-64)
electrode potentials of the two half-cells. In the case
of a cell using the standard hydrogen electrode, E is
the electrode potential of the experimental couple.
The number of moles of electrons transferred in the
reaction equation (n) is usually 1 or 2 in biochemical
reactions (2 for Eq. 6-62).
Since the reactants and products need not be at
unit activity, we must define the observed electrode
potential E as a function of E and the activities (con-
centrations) of A, AH
2
, and H
+
(Eq. 6-64).
In the biochemical literature values of the appar-
ent standard electrode potential at pH 7 (E) are
usually tabulated instead of values of E (Table 6-8,
second column). Note that E (pH 7) for the hydro-
gen electrode is not zero, as it is at pH 0, but 0.414 V.
Values of E are related to E by Eq. 6-64 with [H
+
]
2
O
2
+ 4 H
+
+ 4e

2 H
2
O + 1.229 + 0.815 0.0
Fe
3+
+ e

Fe
2+
0.771 0.771 8.5
NO
3

+ 2 H
+
+ 2e

NO
2

+ H
2
O0.42176.0
Cytochrome f (Fe
3+
) + e

cytochrome f (Fe
2+
) 0.365 86.8
Fe (CN)
6
3
(ferricyanide) + e

Fe (CN)
6
4
0.36 87.8
O
2
+ 2H
+
+ 2 e

H
2
O
2
0.709 0.295 100.3
Cytochrome a (Fe
3+
) + e

Cytochrome a (Fe
2+
) 0.29 101.3
p-Quinone + 2 H
+
+ 2 e

hydroquinone 0.699 0.285 102.3


Cytochrome c (Fe
3+
) + e

cytochrome c (Fe
2+
) 0.254 108.3
Adrenodoxin (Fe
3+
) + e

adrenodoxin (Fe
2+
) 0.15 128.3
Cytochrome b
2
(Fe
3+
) + e

cytochrome b
2
(Fe
2+
) 0.12 134.1
Ubiquinone + 2 H
+
+ 2 e

ubiquinone H
2
0.10 138.0
Cytochrome b (Fe
3+
) + e

cytochrome b (Fe
2+
) 0.075 142.8
Dehydroascorbic acid + 2 H
+
+ 2 e

ascorbic acid 0.058 146.1


Fumarate
2
+ 2 H
+
+ 2 e

succinate
2
0.031 151.3
Methylene blue + 2 H
+
+ 2 e

leucomethylene blue (colorless) 0.011 155.2


Crotonyl-CoA + 2 H
+
+ 2 e

butyryl-CoA 0.015 160.2


Glutathione + 2 H
+
+ 2 e

2-reduced glutathione ~ 0.10 176.6


Oxaloacetate
2
+ 2 H
+
+ 2 e

malate
2
0.166 189.3
Pyruvate

+ 2 H
+
+ 2 e

lactate
1
0.185 193.0
Acetaldehyde + 2 H
+
+ 2 e

ethanol 0.197 195.3


Riboflavin + 2 H
+
+ 2 e

dihydroriboflavin 0.208 197.4


Acetoacetyl-CoA + 2 H
+
+ 2 e

-hydroxybutyryl-CoA 0.238 (38C) 203.2


S + 2 H
+
+ 2 e

H
2
S 0.14 0.274 210.2
Lipoic acid + 2 H
+
+ 2 e

dihydrolipoic acid 0.29 213.2


NAD
+
+ H
+
+ 2 e

NADH 0.113 0.32 219.0


NADP
+
+ H
+
+ 2 e

NADPH 0.324 219.8


Ferredoxin (Fe
3+
) + e

ferredoxin (Fe
2+
) (Clostridia) 0.413 237.0
2 H
+
+ 2 e

H
2
0 0.414 237.2
CO
2
+ H
+
+ 2 e

formate

0.42 (30C) 238.3


Ferredoxin (Fe
3+
) + e

ferredoxin (Fe
2+
) (spinach) 0.432 240.6
TABLE 6-8
Reduction Potentials of Some Biologically Important Systems
a,b
G (pH 7)
(kJ mol
1
)
for oxidation by O
2
(per 2 electrons) Half-reaction
E
(pH 7)
(V) E (V)
a
A compound with a more positive potential will oxidize the reduced form of a substance of lower potential with a standard free energy
change G = nF E = n 96.49 kJ mol
1
where n is the number of electrons transferred from reductant to oxidant. The temperature
is 25C unless otherwise indicated. E refers to a standard state in which the hydrogen ion activity = 1; E refers to a standard state of pH 7,
but in which all other activites are unity.
b
The major source is Loach, P. A. (1976) in Handbook of Biochemistry and Molecular Biology 3rd ed. Vol. I (Fasman, G. D. ed.),
pp. 122130, CRC Press, Cleveland, Ohio.
E E
RT
F
n
E E
= +
=
= +
+
+
n
ln
[ ][ ]
[ ]
. log
[ ][ ]
[ ]
A H
AH
If
A H
AH
volts at C
2
2
2
2
2
0 0296 25
C. Electrode Potentials and Gibbs Energy Changes for OxidationReduction Reactions
302 Chapter 6. Thermodynamics and Biochemical Equilibria
Figure 6-3 Device for measurement of electrode potentials.
The electrode reactions are indicated below each half-cell.
The maximum electrical work that can be done by such a cell
on its surroundings is G = nEF, where E = V
2
V
1
as measured
by a potentiometer. If A is reduced to AH
2
by H
2
, electrons
will flow through an external circuit as indicated. A will be
reduced in the right-hand cell. H
2
will be oxidized to H
+
in
the left-hand cell. Protons will flow through the gel bridge
from left to right as one of the current carriers in the internal
circuit.
Gel-filled
bridge
Direction of
electron flow
through wire if
emf is positive
Standard
hydrogen
electrode
Platinum
electrodes
H
2
2H
+
+ 2e

A + 2H
+
+ 2e

2H
2
AH
2
A
+
a
H
+
=1
H
2
(1 atm)
V
1
V
2
E V
2
V
1
deleted from the numerator (since the term in log [H
+
]
is contained in E ). On the scale of E (pH 7) the poten-
tial of the oxygenwater couple is 0.815 V, while that
of the NAD
+
NADH couple is 0.32 V.
D. The Adenylate System
Of central importance to the energy metabolism
of all cells is the adenylate system which consists of
adenosine 5'-triphosphate (ATP), adenosine 5'-diphos-
phate (ADP), and adenosine 5'-monophosphate
(AMP) together with inorganic phosphate (P
i
),
pyrophosphate (PP
i
), and magnesium ions. Remem-
ber that P
i
refers to the mixture of ionic forms of phos-
phoric acid present under experimental conditions.
Between pH 4 and pH 10 this will be mainly H
2
PO
4

N
N
NH
2
N
N
O
OH HO
CH
2
O
P O
P
O
O
Adenosine triphosphate
ATP
4
O
O
O
P
O
O
O

Hydrolysis here
yields AMP +
pyrophosphate (PP
i
)
Hydrolysis here yields
ADP + inorganic
phosphate (P
i
)


5'
(pK
a
= 6.8) and HPO
2
2
. Likewise, the symbols AMP,
ADP, and ATP refer to mixtures of ionic forms and PP
i
refers to a mixture of the ions of pyrophosphoric acid.
Above pH 4.4 only H
2
P
2
O
7
2
(pK
a
= 6.1), HP
2
O
7
3
(pK
a
= 9.0), and P
2
O
7
4
contribute appreciably to PP
i
.
1. Storage and Utilization of Energy
ATP is a thermodynamically unstable molecule
with respect to hydrolysis to either ADP or AMP as
is indicated in the foregoing diagram. The standard
Gibbs energy of hydrolysis, G, of ATP
4
to ADP
3
+
HPO
4
2
at pH 7 is 34.5 kJ mol
1
and that of hydrolysis
of ADP
3
to AMP
2
+ HPO
4
2
is 36.3 kJ mol
1
at 25C
(Table 6-6). The exact value of these changes in Gibbs
energy depends on pH and on the concentration of
Mg
2 +
as is detailed in Sections B, 4, 5. Rates of reaction
of components may also depend upon metal ions.
Magnesium ion is especially important and complexes
such as MgATP
2
are regarded as the true substrates
for many ATP-utilizing enzymes.
The large Gibbs energy decreases upon hydrolysis
(high group transfer potentials) enable cells to use ATP
and ADP as stores of readily available energy. Energy
from the adenylate system is used for many purposes
including biosynthesis, transport of ions and molecules
across membranes, and for doing mechanical work.
The mechanisms by which this energy is utilized are
considered later (see Chapters 10, 12, and 17 19). The
first step most often involves transfer of either the
terminal -phospho group of ATP to a site on a differ-
ent molecule or transfer of the entire AMP portion of
the molecule onto another group. Thus the products
of cleavage of ATP in these energy-utilizing processes
may be either ADP + P
i
or AMP + PP
i
. In the latter
case the pyrophosphate is usually hydrolyzed rapidly
to two molecules of P
i
by pyrophosphatases. This
process, too, serves an essential function in the adeny-
late system, because it removes a product of the initial
ATP cleavage reaction, shifting the overall equilibrium
of the reaction sequence in the direction of the products.
The adenylate system provides the major store
of rapidly available energy but the whole family of
nucleoside triphosphates that are related to ATP in
structure have similar functions. These include gua-
nosine triphosphate (GTP), uridine triphosphate (UTP),
cytidine triphosphate (CTP), and deoxyribonucleotide
triphosphates (Table 5-1). These compounds are formed
by successive transfer to the nucleoside monophosphate
(GMP, UMP, CMP, dGMP, etc.) from two different mole-
clues of ATP of two phospho groups. The resulting
compounds are used to provide energy for a variety of
specific biosynthetic processes, including synthesis of
RNA and DNA. Inorganic polyphosphates, linear
polymers of orthophosphate (P
i
), are present in nearly
all living forms.
52 54
Like ATP, they can also store
303
energy and in some organisms substitute for ATP in
certain enzymatic reactions.
2. Synthesis of ATP
The phosphate anhydride (pyrophosphate)
linkages of ATP are generated by the joining of
ADP and inorganic phosphate by means of special
phosphorylation reactions. The most important of
the latter occur in the photosynthetic membranes of
chloroplasts (photosynthetic phosphorylation) and
in oxygen-utilizing membranes of bacteria and of
mitochondria (oxidative phosphorylation). Con-
version of AMP to ADP is accomplished by transfer of
the terminal phospho group from an ATP molecule to
AMP in a reaction catalyzed by the extremely active
enzyme adenylate kinase (Chapter 12) which is found
in all cells. The following equations indicate how one
of the special phosphorylation reactions must be used
twice for the conversion of one molecule of AMP into
one molecule of ATP.
Various measures of the phosphorylating potential
of the adenylate system within cells have been pro-
posed. One measure is the product [ATP] / [ADP][P
i
],
which will be called the phosphorylation state ratio
or R
p
in this book. It is also sometimes called the phos-
phorylation potential. This ratio directly affects the
Gibbs energy of hydrolysis of ATP, as is shown by Eq.
6-29. The value of R
p
may be as high as 10
4
to 10
5
M
1
within cells
55
adding 22.8 kJ mol
1
to G of hydrolysis
of ATP. Another quantity that is sometimes cited is the
energy charge, the mole fraction of adenylic acid
charged by conversion to ATP. ADP is regarded as
half-charged.
56,57
The energy charge varies from 0 if only AMP is
present to 1.0 if all of the AMP is converted to ATP.
AMP
Adenylate
kinase
Phosphorylation system
ATP
ADP
P
i
H
2
O
ATP
ADP
P
i
H
2
O
(6-65)
Energy charge
ATP ADP
ATP ADP AMP
=
+
+ +
[ ] [ ]
[ ] [ ] [ ]
1
2
(6-66)
Measurements on a variety of cells and tissues show
that the energy charge is usually between 0.75 and
0.90. Although it is easy to calculate its numerical
value, the energy charge cannot be used in chemical
equations and the idea that the energy charge of a cell
plays a key role in regulation of metabolism has been
challenged.
58
3. Creatine Phosphate, an Energy Buffer
Although ATP provides the immediate source of
energy for operating muscle, its concentration is only
about 5 mM. However, muscle contains, in addition,
a phosphagen, an N-phospho derivative of a guanidin-
ium compound. In mammalian muscle, the phosphagen
is creatine phosphate. Related compounds including
arginine phosphate serve in various invertebrates.
The group transfer potential (G of hydrolysis) for
creatine phosphate is 43.1 kJ mol
1
. Thus, the transfer
of a phospho group from creatine phosphate to ADP to
form ATP (Eq. 6-67) is spontaneous with G = 8.6 kJ
mol
1
. Recent values under a variety of conditions
have been reported by Taugue and Dobson.
59
Creatine
phosphate, which is present at a concentration of 20
mM, provides a reserve of high-energy phospho groups
and keeps the adenylate system of muscle buffered at
a high value of R
p
.
4. Phosphorus-31 NMR
A spectacular development is the ability to observe
components of the adenylate system as well as phos-
phocreatine and other phosphate esters in living cells
by
31
P NMR.
60 61a
Spectra can be recorded on suspen-
sions of cells
60
or of mitochondria,
62
on individual
excised muscles (Fig. 6-4),
63,64
or on perfused organs
65,66
including beating rat hearts and surgically exposed
animal organs.
61,61a
Metabolism can be observed in
human erythrocytes
67
and even in human limbs, liver,
hearts, and brain.
68 70
The method can play a valuable
role in understanding human diseases.
63,67,70
As is
seen in Fig. 6-4, each phosphorus atom of ATP gives
a distinct resonance. The area of the P

peak provides
a direct measure of the ATP concentration, and
D. The Adenylate System
COO
CH
2
N C
N H
3
C
NH
2
P
O
O
O
H
COO
CH
2
N C
NH
2
H
3
C
NH
2

Creatine phosphate
+
Creatine

G' = 8.6 kJ mol
1
ADP ATP
(6-67)
304 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-72)
(6-70)
(6-71)
(6-68)
(6-69)
phosphocreatine (creatine-P) and P
i
can be estimated
in a similar way. Knowing the concentrations of ATP,
P
i
, and creatine-P, the amount of ADP present, usually
too low to be estimated by NMR, may be calculated.
61
Barnacle muscle contains, instead of phosphocreatine,
a high concentration of phosphoarginine which has
also been measured by NMR.
71
Usually no ADP can be seen in NMR spectra,
although there may be about 0.5 mM ADP according
to chemical analysis of rapidly frozen tissues. It has
been concluded that most ADP is bound to proteins.
In muscle the proteins myosin and actin (Chapter 20)
hold most of the ADP leaving only about 0.02 mM
free.
62,72
The same conclusion has been reached on
the basis of other evidence.
55
If a perfused heart in an NMR spectrometer is
deprived of oxygen, the level of phosphocreatine
drops rapidly and that of P
i
increases while that of ATP
remains relatively constant until the phosphocreatine
is gone. Then it too falls and becomes undetectable
after 17 min.
72
Similar changes occur when a tourniquet
is placed on a human arm in the NMR spectrometer.
Study of the rate and extent of recovery of the adeny-
late system when oxygen is readmitted is helping to
provide a better means of protecting kidneys and
other organs during transplantation operations. Diag-
nosis of circulatory ailments in human limbs by
31
P
NMR may soon be routine. Use of radiofrequency
coils placed on the body surface allows monitoring
of the adenylate system in the heart, brain, and other
tissues deep within the body.
69,73
Changes of concen-
tration in the adenylate system and of phosphocreatine
can be monitored very rapidly and evenly throughout
the cardiac heartbeat cycle.
74
P
i
sugar
phosphates
Creatine-P
P

20 10 0 -10
Chemical shift (ppm)
ATP
Figure 6-4 Phosphorus-31 NMR spectrum of an excised
rat muscle (vastus lateralis) in Ringer solution at 15C. The
spectrum represents the accumulation of 400 scans. From
P. J. Seeley et al.
64
E. Complex Biochemical Equilibria
The binding of small molecules to larger ones is
basic to most biological phenomena. Substrates bind
to enzymes and hormones bind to receptors. Metal
ions bind to ATP, to other small molecules, and to
metalloproteins. Hydrogen ions bind to amino acids,
peptides, nucleotides, and most macromolecules. In
this section we will consider ways of describing math-
ematically the equilibria involved.
The strength of bonding between two particles can
be expressed as a formation constant (or association
constant) K
f
. Consider the binding of a molecule X to
a second molecule P, which may be a protein or some
other macromolecule. If there is on the surface of P
only one single binding site for X, the process can be
described by Eq. 6-68 and the equilibrim constant K
f
by Eq. 6-69.
X + P PX
K
f
= [PX] / [P][X]
The units for K
f
are liters per mole (M
1
). The constant
K
f
is a direct measure of the strength of the binding:
The higher the constant, the stronger the interaction.
This fact can be expressed in an alternative way by
giving the standard Gibbs energy change (G) for the
reaction (Eq. 6-70). The more negative G
f
, the stronger
the binding.
G
f
= RT ln K
f
= 2.303RT log K
f
= 5.708 log K
f
kJ mol
1
at 25C
To avoid confusion, it is important to realize that the
frequently used dissociation constants (K
d
) are recip-
rocal association constants (Eq. 6-71). The use of
K
d
= 1 / K
f
association constants and of dissociation constants is
firmly entrenched in different parts of the chemical
literature; be sure to keep them straight. Dissociation
constants are customarily used to describe acidbase
chemistry, while formation constants are more often
employed to describe complexes with metal ions or
associations of macromolecules (Section C). However,
both types of equilibria can be described using either
formation constants or dissociation constants.
Logarithms of formation constants, which are
proportional to the Gibbs energies of association, are
often tabulated. The logarithms of formation constants
and pK
a
values of dissociation constants are identical
(Eq. 6-72) and are a measure of the standard
log K
f
= log K
d
= pK
d
305
(6-74)
Gibbs energy decrease in the association reaction. The
difference in G corresponding to a change of one
unit in log K
f
or pK
d
is 5.7 kJ mol
1
, a handy number
to remember.
1. Effects of pH on Equilibria
Compounds that contain several acidic or basic
groups can exist in a number of different ionic forms,
H
n
A, H
n1
A, H
2
A, HA, A, etc., as is indicated in the
following equation, which is identical in form to Eq. 3-3.
The dissociation constants K
1
- - - - K
n
for a multi-
protic acid H
n
A are defined as stepwise or macroscopic
constants (also called molecular constants). For some
compounds, e.g. alanine, the pK
a
values are far apart
(pK
1
and pK
2
are 2.4 and 9.8, respectively). The macro-
scopic constants can be assigned specifically, K
1
to the
carboxyl group and K
2
to the protonated amino group.
At the isoelectric pH of 6.1 the alanine exists almost
entirely as the dipolar ion. However, for compounds
in which the macroscopic pK
a
values are closer togeth-
er, they cannot be assigned to specific groups. We will
consider some specific examples in the next section.
2. Microscopic Dissociation Constants and
Tautomerization
A microscopic constant applies to a single site.
Consider the dissociation of a simple carboxylic acid:
R COOH R COO

+ H
+
The dissociation constant is about 1.7 x 10
5
and pK
a
=
4.8. Since there is only one proton, the observed disso-
ciation constant is also the microscopic dissociation
constant. Now consider the cation of pyridoxine
which has two dissociable protons bound to distinctly
different sites, the phenolic oxygen and the ring nitrogen.
(6-73)
N
HOH
2
C
CH
3
OH
CH
2
OH
+
H
pK
B
* = 5.62
K
B
* = 2.04 10
6
pK
A
* = 5.04
K
A
* = 7.9 10
6
Pyridoxine
(vitamin B
6
)
H
n
A H
n1
A H
2
A HA A
K
1
K
n+1
K
n
Either of the two protons might dissociate first as
the pH is raised (Eq. 6-75). However, the two micro-
scopic dissociation constants pK
A
* and pK
B
* are dis-
tinctly different. The result is that at 25C in the neutral
(monoprotonated) form 80% of the molecules carry a
proton on the N, while the other 20% are protonated
on the less basic O

. Notice that the subscripts a and b


used in this discussion do not refer to acidic and basic but
to the individual dissociation steps shown in Eq. 6-75.
Microscopic constants will always be indicated with
asterisks in this discussion.
The two monoprotonated forms of pyridoxine are
the tautomeric pair shown in Eq. 6-75 and whose con-
centrations are related by the tautomeric ratio, R =
[neutral form]/[dipolar ion], a pH-independent equilibrium
constant with a value of 0.204/0.796 = 0.26 at 25C.
75
Evaluation of microscopic constants for dissociation
of protons from compounds containing non-identical
groups depends upon measurement of the tautomeric
ratio, or ratios if more than two binding sites are present.
In the case of pyridoxine, a spectrophotometric method
was used to estimate R.
To calculate microscopic constants from stepwise
constants and tautomeric ratios, consider Eq. 6-76 in
which [HP]
A
and [HP]
B
are the concentrations of the
two tautomers and K
1
is the first stoichiometric or
macroscopic dissociation constant for the diprotonated
species H
2
P.
pK
1
= 4.94; K
1
= 1.15 x 10
5
= 9.1 x 10
6
+ 2.4 x 10
6
E. Complex Biochemical Equilibria
N
HOH
2
C
CH
2
OH
OH
CH
3
N
HOH
2
C
CH
2
OH
O
CH
3
N
HOH
2
C
CH
2
OH
O
CH
3
N
HOH
2
C
CH
2
OH
OH
CH
3
H
+

pK
C
* = 8.79
pK
D
* = 8.21
+
pK
B
* = 5.62
H
R = 0.26
pK
A
* = 5.04
pK
1
= 4.94 pK
2
= 8.89
(6-75)
K K K
1
2
=
+
= +
+

([ ] [ ] )[ ]
[ ]
HP HP H
H P
A B
A B
(6-76)
306 Chapter 6. Thermodynamics and Biochemical Equilibria
(6-78)
(6-79)
(6-80)
(6-81)
(6-77)
We see that K
1
is just the sum of the two microscopic
constants K
A
* and K
B
* for dissociation of H
2
P to the
pair of tautomers HP(A) and HP(B). In a similar
fashion it can be shown that the second stoichiometric
constant K
2
is related to the microscopic constants K
C
*
and K
D
* for dissociation of HP(A) and HP(B) to form P
(Eq. 6-77).
1 / K
2
= 1 / K
C
* + 1 / K
D
*
Since the tautomeric ratio R equals [HP]
B
/ [HP]
A
, Eqs.
6-76 and 6-77 can be rearranged to Eqs. 6-78 to 6-81.
These allow the evaluation of all of the microscopic
constants from the two stoichiometric constants K
1
and K
2
plus the tautomeric ratio R.
pK
A
* = pK
1
+ log (l + R)
pK
B
* = pK
A
* log R
pK
C
* = pK
2
log (l + R)
pK
D
* = pK
C
* + log R
For pyridoxine pK
1
and pK
2
were determined
spectrophotometrically as 4.94 and 8.89. These values,
together with that of R given above, were used to
estimate the microscopic constants that are given in
Eq. 6-74.
75
Notice that the microscopic constants of
Eq. 6-74 are not all independent; if any three of the five
equilibrium constants are known the other two can be
calculated readily. In describing and measuring such
equilibria it is desirable to select one pathway of disso-
ciation, e.g., H
2
P HP(A) P, and to relate the species
HP(B) to it via the pH-independent constant R.
Often more complex situations arise in which
additional tautomers or other forms arise via pH-
independent reactions. These can all be related back
to the reference ionic species by additional ratios R,
which may describe equilibria for tautomerization,
hydration, isomerization, etc. (Eq. 6-82).
76
In the case
illustrated, only one of the ratios, namely R
2
or R
3
, is
likely to be a tautomerization constant because, as a
rule, H
2
P and P will not have tautomers. Equations
analogous to Eqs. 6-76 to 6-82 can be written easily to
derive K
C
*, K
D
* and any other microscopic constants
desired from the stoichiometric constants plus the ratios
R
1
to R
4
. While it is easy to describe tautomerism by
equations such as Eqs. 6-76 and 6-82 it is often difficult
HP(A)
HP(B) H
2
P(B)
P(A)
K
a
*
R
1
HP(C)
R
4
H
2
P(A)
R
3
R
2
K
b
*
P(B)
(6-82)
to measure the tautomeric ratios R.
77
In favorable cases
measurements of spectra of one kind or another allow
their evaluation. However, because tautomerism may
be extremely rapid, NMR spectra will often show only
one peak for a proton present in a mixture of tautomers.
As was pointed out in Chapter 2, tautomerization
ratios are often affected strongly by changes in solvent.
Tautomerism among monoprotonated forms of cysteine,
glutathione, and histidine (Eq. 2-6) has received con-
siderable attention by biochemists
77 79
as has tautom-
erism in binding of protons and other small ligands to
proteins.
80,81
For cysteine,
78,79
for which the following
species coexist in the alkaline pH range, the distribution
of the various ionic species including the two tautomers
is shown in Fig. 6-5. A similar situation is met in the
small protein thioredoxin (Box 15-C) which has a buried
aspartate carboxylate that interacts with a nearby
cysteine SH group,
82
in papain where SH and imi-
dazole groups interact (Fig. 12-15),
83
and in carbohy-
drases where two or more carboxyl and carboxylate
side chains interact (Chapter 12).
84
OOC C
H
CH
2
S
NH
3
OOC C
H
CH
2
SH
NH
2
and

8.53 10.36
8.86 10.03
6 7 8 9 10 11 12
0
0.2
0.4
0.6
0.8
1.0
pH
F
r
a
c
t
i
o
n
a
l

c
o
n
c
e
n
t
r
a
t
i
o
n
Total S
NH
2
S

OOC

NH
3
+
OOC

NH
2
SH
OOC

SH
OOC

NH
3
+
Figure 6-5 Distribution of various ionic species of cysteine
as a function of pH. The function in each ionic and tauto-
meric form is plotted. Microscopic pK
a
values are given
numerically and macroscopic pK
a
s are indicated by the
vertical lines. From Dixon and Tipton.
79
307
Because it is usually difficult to measure tautomeric
ratios, dissociation constants are frequently assumed
identical to those of compounds in which one of the
acidic groups has been modified by methylation or
esterification or in some other manner to prevent
dissociation of a proton. For example, the cation of
1-methyluracil can be dissociated to the two tautomers
shown in Eq. 6-83.
The apparent pK
a
values were estimated for dissociation
of the following two dimethylated cations which
resemble that of Eq. 6-83.
It can reasonably be assumed that these pK
a
values
approximate pK
A
* and pK
B
* as indicated. Thus, applying
Eq. 6-85, log R 3.25 0.65 + 3.90. This result indi-
cates that tautomer A of Eq. 6-89 is overwhelmingly
predominant in water.
85,86
It also suggests that, within
experimental error, the pK
a
for 1-methyluracil will
equal pK
A
*, namely, 3.25. In fact, it is close to this
( 3.40).
85
N
N
OH
O
CH
3
H
N
N
O
O
CH
3
H
N
N
O
O
CH
3
H
+
R
K
A
*
K
B
*
A
B
(6-83)
3. The Binding of Metal Ions
Equilibria in the formation of complex ions with
metals are treated exactly as is the binding of small
molecules and ions to macromolecules.
87 89
Stepwise
constants are defined for the formation of complexes
containing one, two, or more ligands L bound to a
central metal ion M. The binding constants K
f
s are
usually referred to as s as in Eq. 6-84.

1
= K
f1

n
= K
f 1
K
f 2
... K
f n
Many important questions can be asked about the
binding of metal ions within living cells. For example,
What fraction of a given metal ion is free and what
fraction is bound to organic molecules? To what ligands
is a metal bound? Since many metal ions are toxic in
excess, it is clear that homeostatic mechanisms must
exist. How do such mechanisms sense the free metal
ion activity within cells? How does the body get rid of
unwanted metal ions? Answers to all these questions
depend upon the quantitative differences in the binding
of metal ions to the variety of potential binding sites
found within a cell.
Table 6-9 gives formation constants for 1:1 complexes
of several metal ions and a number of inorganic as well
as organic ligands.
89
Only the values of log K
1
are
given when a series of stepwise constants have been
established. However, in many cases two or more
ligands can bind to the same metal ion. Thus for
cupric ion and ammonia there are four constants.
Cu
2 +
+ NH
3
log K
i
= 4.0, 3.3, 2.7, 2.0
They are all separated by more than the statistical
distance, which in this case is less than the 0.6 loga-
rithmic units for two equivalent binding sites (see
Chapter 7, Section A,2). Thus, the second ligand binds
less tightly than the first and anticooperativity in
binding of successive ligands is observed. Most metal
ions will also bind two or three successive amino acids.
In the case of copper, whose preferred coordination
number is four, two ligands may be bound. Again, a
distinct anticooperativity is evident in the spread of
the two constants.
Cu
2 +
+ alanine log K
i
= 8.1, 6.8
Factors affecting the strength of binding of a
metal in a complex. More basic ligands tend to bind
metal ions more tightly just as they do protons. How-
ever, the strength of bonding to metal ions to a group
is more nearly proportional to the nucleophilic char-
acter (Chapter 12) which is only partly determined by
basicity to protons.
E. Complex Biochemical Equilibria
(6-84)
N
N
O
H
H
3
C
O
CH
3
N
N
OCH
3
O
CH
3
pK
a
= +0.65
pK
A
*
+
pK
a
= 3.25
H
pK
B
*
+
pK
A
* pK
A
*
308 Chapter 6. Thermodynamics and Biochemical Equilibria
BOX 6-C LINEAR GIBBS ENERGY RELATIONSHIPS
Organic functional groups exert characteristic
electronic effects upon other groups to which they
are attached. The quantitative expression of such
effects can sometimes be correlated by linear Gibbs
energy relationships. The best known of these is the
Hammett equation, which deals with the trans-
mission of electronic effects across a benzene or
other aromatic ring. Consider the acid dissociation
constants of three classes of compounds:
The values of pK
a
given in the following table have
been observed for the parent compounds and for
the meta-chloro- and the meta-nitro-substituted
compounds.
changes in the dissociation constant of benzoic acid
define the substituent constants , which are used
in the Hammett equation and are given in the fol-
lowing table. In this equation we use the symbol
pK
0
to represent the pK
a
of the unsubstituted parent
compound and pK to represent the pK
a
of the sub-
stituted molecule.
For substituted benzoic acids: pK
0
pK =
The decreases in the pK of phenylacetic acid
occasioned by replacement of the meta-hydrogen
with Cl or NO
2
are 0.18 and 0.34, respectively,
substantially less than for the benzoic acids. On the
other hand, for the phenols the differences amount
to 0.90 and 1.53, much greater than for the benzoic
acids. The Hammett equation asserts that for reac-
tions such as the dissociation of protons from phe-
nylacetic acids or from phenols, the changes in G
occasioned by meta substitutions are proportional
to the values, i.e., to the changes in G for the
standard reaction dissociation of a proton from
benzoic acid.
b d
Since pK
a
is the negative logarithm of a dissoci-
ation constant, it follows from Eq. 6-30 that values
of pK
a
are directly proportional to values of G for
dissociation of protons. In the Hammett treatment
differences in pK
a
values, rather than differences in
G, are considered. When a hydrogen atom in the
meta position of benzoic acid is replaced by the
electron-withdrawing Cl or NO
2
, pK
a
is lowered,
i.e., the basicity of the conjugate base of the acid is
decreased. The decrease in pK
a
amounts to 0.365
for m-chlorobenzoic acid and 0.742 for m-nitroben-
zoic acid. The Hammett treatment asserts that these
changes in pK
a
are a measure of the electron-with-
drawing power of the meta substituent.
b
Thus, the
nitro group is about twice as strong as the chloro
group in this respect. The numerical values of these
Substituent Constants for Use in
the Hammett and Taft Equations
b-d
Substituent
m

p

p

p
+
* (Taft)
H (parent compound) 4.202 4.31 9.92
Cl 3.837 4.13 9.02
NO
2
3.460 3.97 8.39
Values of pK
a
for Unsubstituted and Substituted
Benzoic Acids, Phenylacetic Acids, and Phenols
* For an independent set of pK
a
values for substituted benzoic
acids see Bolton and Fleming.
a
The values of calculated from
them are slightly different from those given here.
Meta
substituent
Benzoic
acids*
Phenylacetic
acids Phenols
C
OH
O
C
OH
O X
Benzoic acids
CH
2
X
OH
X
Phenol
Phenylacetic
O

0.71 0.52
NH
2
0.16 0.66 1.11 0.62
OH 0.121 0.37 0.85 1.34
OCH
3
0.115 0.27 0.78 1.81
CH
3
0.069 0.17 0.31 0.00
NH COCH
3
0.21 0.01 0.25
H 0 0 0 0.49
CH
2
OH 0.08 0.08 0.56
COO

0.10 0.00 0.11 1.06


SO
3

0.05 0.09 0.12


SH 0.25 0.15 0.019 1.68
CH
2
Cl 0.18 1.05
CONH
2
0.28 0.36
F 0.337 0.06 0.02 0.07
I 0.352 0.18 0.135
Cl 0.373 0.227 0.114
CHO 0.36 0.22 0.37
COCH
3
0.376 0.502 0.85
COOH 0.37 0.45 0.42 2.08
COOCH
3
0.39 0.31 0.49
SO
2
NH
2
0.55 0.62 0.61
CN 0.56 0.66 0.89 0.66
C CH 2.18
CF
3
0.43 0.54 0.61 2.61
CCl
3
2.65
NO
2
0.710 0.778 1.25 0.790 4.0
NH
3
+
1.13 1.70 3.76
N (pyridine) 0.73 0.83
NH
+
(pyridine) 2.18 2.42 4.0
309
BOX 6-C (continued)
log (K/K
0
) = pK
0
pK =
The Hammett equation
The proportionality constant , which also appears
in the equation, is a measure of the sensitivity of the
reaction to the presence of electron-withdrawing or
electron-donating substituents in the ring. For ben-
zoic acid, is taken as 1.00. Using the data from the
accompanying table together with many other data,
an average value of = 0.49 has been found for
phenylacetic acids. Likewise, = 2.23 for phenols,
and = 5.7 for dissociation of protons from substi-
tuted pyridinium ions. The sensitivity to substitu-
ent changes is highest (highest ) in the latter case
where proton dissociation is directly from an atom
in the ring and is lowest when the basic center is
removed farthest from the ring (phenylacetic acid).
Through knowledge of the value of for a
given reaction, it is possible to predict the effect of
a substituent on pK
a
using the tabulated values of .
In many cases the effects are additive for multisub-
stituted compounds.
pK
a
= pK
0

Since substituents in o, m, and p positions have quan-
titatively different influences, different substituent
constants are defined for each position. Moreover,
since special complications arise from ortho inter-
actions, it is customary to tabulate values only for
meta and para positions. These are designated
m
and
p
. Apparent values for ortho substituents are
also available.
d
An additional complication is that
certain reactions are unusually sensitive to para
substituents that are able to interact by resonance
directly across the ring. An example is the acid
dissociation of phenols. While
p
for the nitro
group is ordinarily 0.778, a correct prediction of the
effect of the p-nitro group on dissociation of phenol
is given only if
p
is taken as 1.25. This higher value
is designated
p

. The resonance in the phenolate


anion giving rise to this enhanced effect of the nitro
group may be indicated as follows:
(Here the curved arrows represent the direction of
flow of electrons needed to convert from the one
resonance structure to the other.) For similar rea-
sons, some reactions require the use of special
p
+
constants for strongly electron-donating substitu-
ents such as OH which are able to interact across
the ring by resonance. Thus
p
for the OH group is
0.37, while
p
+
is 0.85.
e
For the methoxyl group
( OCH
3
)
p
= 0.27,
p
+
= 0.78, and
m
= + 0.12.
The use of different kinds of substituent con-
stants complicates the application of the Hammett
equation and over 20 different sets of values have
been proposed. A simplification is the representation
of substituent constants as linear combinations of
two terms, one representing field or inductive
effects and the other resonance effects.
e,f
Many other linear Gibbs energy relationships
have been proposed; for example, the acid strengths
of aliphatic compounds can be correlated using the
Taft polar substituent constants *.
log (K/K
0
) = * *
For example, the following give good approximations
of pK
a
values.
d
for R COOH pK
a
= 4.66 1.62 *
for R CH
2
COOH pK
a
= 5.16 0.73 *
While the examples chosen here concern only
dissociation of protons, the Hammett equation has
a much broader application. Equilibria for other
types of reactions can be treated. Furthermore,
since rates of reactions are related to Gibbs energies
of activation, many rate constants can be correlated.
For these purposes the Hammett equation can be
written in the more general form in which k may be
either an equilibrium constant or a rate constant.
b
The subscript j denotes the reaction under consider-
ation and i the substituent influencing the reaction.
log k
ij
log k
0j
=
i

j
An example of a linear Gibbs energy relationship
that is widely used in discussing mechanisms of
enzymatic reactions is the Brnsted plot (Eqs. 9-90
and 9-91).
a
Bolton, P. D., Fleming, K. A., and Hall, F. M. (1972) J. Am. Chem.
Soc. 94, 1033 1034
b
Hammett, L. P. (1970) Physical Organic Chemistry, 2nd ed.,
p. 356, McGraw-Hill, New York
c
Wells, P. R. (1963) Chem. Rev. 63, 171 219
d
Barlin, G. B., and Perrin, D. D. (1966) Q. Rev., Chem. Soc. 20,
75101
e
Swain, C. G., and Lupton, E. C., Jr. (1968) J. Am. Chem. Soc. 90,
4328 4337
f
Hansen, L. D., and Hepler, L. G. (1972) Can. J. Chem. 50, 1030
1035
E. Complex Biochemical Equilibria
N O

O
O
N O
O
O

310 Chapter 6. Thermodynamics and Biochemical Equilibria


The pH of the medium always has a strong effect
on metal binding. Competition with protons means
that metal complexes tend to be of weak stability at
low pH. Anions of carboxylic acids are completely
protonated below a pH of ~ 4 and a metal can combine
only by displacing a proton. However, at pH 7 or
higher, there is no competition from protons. On the
other hand, in the case of ethylenediamine, whose pK
a
values are 10.2 and 7.5 (Table 6-9), protons are very
strong competitors at pH 7, even with a strongly com-
plexing metal ion such as Cu
2 +
. At high pH there may
be competition between the ligand and hydroxyl ion.
At pH 7 about one-half of Cu
2 +
dissolved in water is
complexed as CuOH
+
. Aluminum forms soluble
complexes Al(OH)
2 +
, Al(OH)
2
+
, and Al(OH)
4
+
(Box 12-F).
TABLE 6-9
Logarithms of Binding Constants for Some 1:1 Metal
Complexes at 25C
a
Ligand H
+
Mg
2+
Ca
2+
Mn
2+
Cu
2+
Zn
2+
a
All values are for log K
1
. Included is the highest pK
a
for protons. Data for amino acids
are from Martell, A.E. and Smith, R.M. (1974, 1975) Critical Stability Constants, 3 vols.,
Plenum, New York. Others are from Silln, L.G. and Martell, A.E. (1964) Stability Constants
of Metal Ion Complexes, Spec. Publ. No. 17, Chemical Society, London. Most constants
for amino acids are for ionic strength 0.1. Some (designated by asterisks) are for zero
ionic strength. The values shown for other ligands have been selected from a large
number reported without examination of the original literature.
b
Ethylenediaminetetraacetic acid (EDTA), a chelating agent widely used by biochemists
for
preventing unwanted reactions of metal ions. The high formation constants ensure
that most metal ions remain bound to the EDTA.
c
EGTA is similar to EDTA but has the group CH
2
CH
2
O CH
2
CH
2
O CH
2
CH
2

joining the two nitrogen atoms in place of CH


2
CH
2
of EDTA. Note that EGTA has
a higher selectivity for Ca
2+
compared to Mg
2+
than does EDTA.
Hydroxide, OH

14.0 2.5 1.4 6.5 4.4


Acetate

4.7 ~0.65 0.5 ~1.0 2.0 1.5


Lactate

3.8 ~1.0 ~1.2 1.3 3.0 2.2


Succinate
2
5.2 1.2 1.2 3.3 2
NH
3
9.3 ~0 0.2 0.8 4.0 2.4
Ethylenediamine 10.2 0.4 2.8 10.8 6.0
Glycine

9.6 2.2 1.4* 2.8 8.2 5.0


Glycine amide 8.1 ~1.5 5.3 3.3
Alanine

9.7 2.0* 1.2* 3.0* 8.1 4.6


Aspartate
2
9.6 2.4 1.6 8.6 5.8
Glycylglycine

8.1 1.2* 2.2* 5.5 3.4


Pyridine 5.2 0.1 2.5 1
Imidazole 7.5 1.6 4.6 2.6
Histidine 9.1 3.3 10.2 6.6
Adenine 9.8 8.9 6.4
Citrate
3
5.6 3.2 4.8 3.5 ~4 4.7
EDTA
4 b
10.2 8.8 10.6 13.8 18.7 16.4
EGTA
4 c
9.4 5.3 10.9 12.2 17.6 12.6
ATP
4
6.5 4.2 3.8 4.8 6.1 4.9
CH
2
CH
2
N N
H
2
C
H
2
C
CH
2
CH
2
COO
COO
OOC
OOC

One of the most important


factors in determining the affinity
of organic molecules for metal ions
is the chelate effect. The term
chelate (pronounced keel-ate) is
from a Greek word meaning crabs
claw. It refers to the greatly enhanced
binding of metal ions resulting from
the presence of two or more com-
plexing groups in the same organic
molecule. The chelate effect has been
exploited by nature in the design of
many important metal-binding
molecules, including porphyrins
(Fig. 16-5), chlorophyll (Fig. 23-20),
the siderophores (Fig. 16-1), and
metal-binding proteins. Structures
of two chelate complexes are shown
here. Notice from Table 6-9 that
many simple compounds, such as
the -amino acids and citric acid,
often form strong chelate complexes
with metal ions.
Co
N
N
C
CH
2
C
CH
2
C
H
2
C
N
O
O
3+

H
H
H
H
O

O
H
H
O
Co
N
N
C
CH
2
C
CH
2
C
H
2
O
O
3+

O
C
O
O

CH
2
C
O
O
CH
2
CH
2
311
How properties of the metal ion affect chelation.
The charge, the ionic radius (Table 6-10), the degree
of hydration, and the geometry of orbitals used in
covalent bonding between metal and chelating groups
all affect the formation constants of a complex. Multi-
charged ions form stronger complexes than do mono-
valent ions, which have a lower charge density.
Among ions of a given charge type (e.g., Na
+
vs
K
+
; Mg
2 +
vs Ca
2 +
), the smaller ions are more strongly
hydrated than are the larger ions in which the charge
is dispersed over a greater surface area. Most cations,
except for the large ones, have a primary hydration
sphere containing about six molecules of water. Four
molecules of water can be placed around the ion in
one plane as shown in the following drawing of water
molecules coordinated to Mg
2 +
.
One additional water molecule can be bound above
and another below to provide six molecules in an
O
H H
O
H
H
O
H H
H
H
O
array of octahedral geometry. An alternative tetra-
hedral arrangement of four molecules of water around
the ion has been suggested for Li
+
and Na
+
.
90
In either
case additional solvent molecules are held in a looser
secondary sphere. For instance, electrochemical trans-
ference experiments indicate a total of ~ 16 molecules
of water around Na
+
and ~ 10 around K
+
.
To form a chelate complex a metal ion must usually
lose most of its hydration sphere. For this reason, the
larger, less hydrated metal ions often bind more strongly
than do the smaller, more hydrated ones. For example,
Ca
2 +
binds to EDTA more tightly than does Mg
2 +
(Table
6-9). However, the reverse order may be observed with
negatively charged ligands such as OH

in which the
charge in highly concentrated. The same is true for
ATP
4
, which binds Mg
2 +
more tightly than Ca
2 +
(Table
6-9; Section B,5).
Differences in both the charge density and the
hydration of ions determine the Hofmeister series
(lyotropic series).
91
Ba
2 +
> Ca
2 +
> Mg
2 +
> Li
+
> Na
+
> K
+
> Cs
+
> NH
4
+
This was originally defined as the order of effectiveness
in precipitating colloids or protein molecules. The ions
to the left are less hydrated than those to the right. A
similar series can be defined for anions. The following,
a well-known sequence of the stabilities of complexes
of metals of the first transition series, applies to many
different types of complexes including those of the
-amino acids, as is shown in Fig. 6-11.
Mn
2 +
< Fe
2 +
< Co
2 +
< Ni
2 +
< Cu
2 +
> Zn
2 +
Simple electrostatic theory based upon differences
in the ionization potentials or electronegativity of the
ions would predict a gradual monotonic increase in
chelate stability from manganese to zinc. In fact, with
nitrogen-containing ligands Cu
2 +
usually forms by far
the strongest complexes. Cobalt, nickel, and iron ions
also show an enhanced tendency to bind to nitrogen-
containing ligands. The explanation is thought to lie
in the ability of the transition metals to supply d orbitals
which can participate in covalent bond formation by
accepting electrons from the ligands. Iron, copper,
and cobalt are often located in the centers of nitrogen-
containing structures such as the heme of our blood
(iron, Fig. 16-5) and vitamin B
12
(Box 16-B).
Metal binding sites in cells. Functional groups
that participate in metal binding include negatively
charged carboxylate COO

, thiolate S

, phenolate O

,
and phosphate

as well as uncharged amino, imida-


zole, OH, and the polarizable carbonyl groups of
peptide and amide side chains. To which of these
ligands will specific ions tend to bind? The alkali
metal ions Na
+
and K
+
are mostly free within cells
E. Complex Biochemical Equilibria
Mn
2+
0.080
Li
+
0.060 Fe
2+
0.076 H

0.21
Na
+
0.095 Co
2+
0.074 F

0.136
K
+
0.133 Ni
2+
0.069 Br

0.195
Rb
+
0.148 Cu
2+
0.072
b
I

0.216
Zn
2+
0.074
Cd
2+
0.097
Be
2+
0.031
Mg
2+
0.065 Al
3+
0.050
Ca
2+
0.099 Fe
3+
0.064
Sr
2+
0.113 Mo
4+
0.070
Ba
2+
0.135 Mo
6+
0.062
TABLE 6-10
Ionic Radii in Nanometers for Some Metallic and
Nonmetallic Ions
a
a
Radii are calculated according to the method of Pauling and
are taken from Cotton, F.A. and Wilkinson, G. (1972) Advanced
Inorganic Chemistry, 3rd ed. Wiley (Interscience), New York .
b
From Ahrens, L. H. as given by Sienko, M. J. and Plane, R. A.
(1963) Physical Inorganic Chemistry, pp. 68 69. Benjamin, New
York.
312 Chapter 6. Thermodynamics and Biochemical Equilibria
(Box 5-A) but are in part bound to defined sites in
proteins. Both Ca
2 +
and Mg
2+
tend to remain partially
free and complexed with the numerous phosphate and
carboxylate ions present in cells. However, they may
find very specific tight binding sites such as that of
Mg
2 +
in chlorophyll (Fig. 23-20). The heavier metal
ions, including those of zinc, copper, iron, and other
transition metals, usually bind to nitrogen or sulfur
atoms.
92
For example, small peptides react with Cu
2 +
to form chelate complexes in which the peptide carbonyl
oxygen binds to the metal (Eq. 6-85, step a).
93,94
By
losing a proton the peptide NH can sometimes also
function as a metal ligand (Eq. 6-85, step b).
In many metalloproteins the metals are found
in prosthetic groups such as the porphyrin of heme
proteins and the molybdopterin of molybdenum con-
taining enzymes (Fig. 16-31). Very often clusters of
carboxylate, imidazole, and other groups are used to
create binding sites. In carboxypeptidase A (Fig. 12-16)
two imidazole groups and one carboxylate of a gluta-
mate site chain hold Zn
2 +
. In carbonic anhydrases,
three imidazoles hold a zinc ion (Fig. 13-1), while in
one kind of superoxide dismutase both Zn
2 +
and Cu
2 +
are bound in adjacent locations with six imidazoles and
one carboxylate group participating in the binding. In
contrast, Zn
2 +
and Cd
2 +
are bound in metallothioneins
by clusters of thiol groups from cysteine side chains
(Box 6-E). While Fe
2 +
is bound by from four to six
nitrogen atoms in heme proteins, it is also found attached
to oxygen atoms of tyrosine side chains, as is shown
for a transferrin in Fig. 16-2. One imidazole, one
carboxylate group, and a bicarbonate ion also bind to
the iron. This site is also quite satisfactory for Al
3+
(Box 12-F), which tends to occupy a fraction of the
transferrin sites in blood, although it binds much more
weakly than does Fe. Several other binding sites for
transition metals are pictured in Chapter 16.
OOC
HN
NH
2
O
HN
N
O
C
O
O
Cu
2
OH
2
OH
2
O
N
N
C
O
O
Cu
OH
2
OH
2

Glycylglycine anion
Cu(H
2
O)
n

H
2

H
2
+
+
a
b
H
+
(6-85)
25 26 27 28 29 30
12
10
8
6
4
2
0
L
o
g
K
f
Mn Fe Co Ni Cu Zn
ATP
4
Gly

His

Figure 6-6 Logarithms of formation constants of metal


complexes of histidine, glycine, and ATP plotted against the
atomic number of elements from manganese to zinc.
Calcium-binding proteins. Much information
about the functions of transition metal ions is given in
Chapter 16. Here and in Box 6-D we consider calcium
ions which, because of their broad distribution and
range of functions, will be discussed in virtually every
chapter of this book. The concentration of Ca
2+
varies
greatly in different parts of a cell and also with time
(Box 6-D). Many calcium-binding proteins participate
in mediating the physiological effects of these changes
and also in buffering the calcium ion concentration.
These include a large family of helixloophelix or
EF-hand proteins.
95 97
The first of these to be discov-
ered was parvalbumin, a 108-residue protein from
carp muscle.
96,98-99a
The structure of the pair of metal-
binding sites of the protein is shown in Fig. 6-7.
Each consists of two helices that are almost per-
pendicular and connected by a loop that forms the
Ca
2+
-binding site. In the site at the left side in Fig. 6-7A
the Ca
2+
is bound by four carboxylate groups from
aspartate and glutamate side chains, a hydroxyl group
of serine, and the residue 57 carbonyl oxygen of the
peptide backbone.
99b
The same peptide group is hydro-
gen bonded to a carbonyl group of another segment of
peptide chain near the second Ca
2 +
site (to the right in
Fig. 6-7A). This site contains four carboxylate ions, one
of which coordinates the Ca
2+
with both oxygen atoms,
and another peptide carbonyl group. By attaching
itself to several different side chain groups, the metal
ion can induce a substantial change in conformation
from that present in the calcium-free protein.
97
313
Figure 6-7 (A) Part of the 108-residue peptide chain of the calcium-binding protein of carp muscle. The two calcium-binding
loops are shown together with a hydrogen bond between them. (B) A view of the intricate network of hydrogen bonds linking
two segments of the peptide chain in the interior of the molecule. Note especially the bonding of the guanidinium group from
arginine-75 to the carboxylate of glutamic acid 81 and to the peptide carbonyl of residue 18. Note that the carboxylate group
also interacts with two different peptide NH groups. From Kretsinger and Nockolds.
98
Kretzinger, who discovered the structure of parval-
bumin, named the Ca
2+
-binding helixloophelix motif
an EF-hand because it is formed by helices E and F and
resembles a hand with pointer finger extended along
the E helix and the thumb in the direction of the F helix,
the flexed middle finger forming the Ca
2+
-binding
loop. Helices C and D also form a hand. The resulting
pair of hands can be visualized better at the top of the
calmodulin structure in Fig. 4-8. The EF-hand motif
has been identified in over 1000 proteins.
100
Parvalbumins, which are also found in other verte-
brates, are high-affinity Ca
2+
-buffers.
99
Additional
calcium buffers with EF-hand structures are the vitamin
D-induced calbindins. One 9-kDa calbindin is found
in mammalian intestinal tissue and in skin. It has
two helixloophelix Ca
2+
-binding sites of differing
affinity
101,102
that presumably function in the absorption
of calcium. A 28-kDa vitamin D-dependent protein
from chicken intestine contains six similar Ca
2 +
-binding
loops.
97,103
Another group of calcium-buffering and storage
proteins with remarkable Ca
2+
-binding properties are
the 40 - to 45-kDa calsequestrins, which are found in
the lumen of the ER (sarcoplasmic reticulum) of skeletal
muscle. Calsequestrins are not typical EF-hand proteins
but have a high content of glutamate and aspartate.
They bind ~ 50 Ca
2+
per molecule of protein with K
d
~ 1 mM.
104,105
Similar proteins called calreticulins
are found in most non-muscle cells.
106,107
A very large number of EF-hand proteins have
signaling functions. The best known of these is the
148-residue calmodulin, which regulates many enzymes
and cellular processes (Box 6-D; also Chapter 11).
108,109
The protein, which is present in all eukaryotes, has a
conserved sequence that forms two pairs of helixloop
helix Ca
2 +
-binding sites that are separated by a long
helix (Fig. 6-8).
108,110,111
Two of the sites bind Ca
2 +
tightly and cooperatively,
110,112
with K
d
values in the
micromolar range. Calmodulin from almost all species
contains the modified amino acid -N-trimethyllysine
at position 115. However, octopus calmodulin has
ordinary lysine at this position and seems to function
well.
108
Calmodulins controlling functions result from
Ca
2+
-induced conformational changes that modify
its affinity for other proteins whose activity may be
increased or decreased by bound calmodulin.
109,113
A protein with a similar dumbell shape and structure
is troponin C of skeletal muscles.
114,115
Troponin C
binds to a complex of proteins that assemble on the
thin actin filaments of muscle fibers and control con-
traction in response to changes in the calcium ion con-
centration (Chapter 19).
116
Other proteins that contain
EF-hand motifs and are therefore responsive to Ca
2+
include spectrin of cell membranes,
117
clathrin light
chains from coated vesicles,
118,119
the extracellular
osteonectin of bones and teeth,
120
and a birch pollen
antigen.
121
Another group of 17 or more small S100
EF-hand proteins play a variety of other roles.
122123a
One of these, which has a high affinity for Zn
2+
, has
been named psoriasin because of its 5-fold or greater
E. Complex Biochemical Equilibria
A B
314 Chapter 6. Thermodynamics and Biochemical Equilibria
BOX 6-D CALCIUM
The essentiality of calcium ions to living things
was recognized in the last century by S. Ringer, who
showed that ~1 M Ca
2+
was needed to maintain
the beat in a perfused frog heart. Later, calcium was
shown essential for repair of ruptures in the cell
membrane of the protozoan Stentor and for the
motion of amebas. The animals quickly died in its
absence. The role in the frog heart was traced to
transmission of the nerve impulse from the nerve to
the heart muscle. More recently it has been recog-
nized that Ca
2+
is required for blood clotting and is
involved in triggering many responses by cells. Cal-
cium ions are also an integral part of many enzymes
and have structural roles in proteins, carbohydrate
gels, and biological membranes.
Like Na
+
, the calcium ion is actively excluded
from cells. Indeed, 99% of the calcium in the human
body is present in the bones.
a d
The blood serum
concentration of Ca
2+
is ~3 mM, of which ~1.5 mM
is free. The rest is chelated by proteins, carbohydrates,
and other materials. Within cells the concentration
of free Ca
2+
is < 1 M and typically ~0.05 0.2 M
for unexcited cells.
d f
However, the total intracellular
Ca
2+
is considerably higher and may be in excess of
1 mM. Approximate total concentrations are: red
blood cells, 20 M; liver, 1.6 mM; and heart, 4 mM.
A gradient in [Ca
2+
] of 10
3
or more is maintained
across membranes by the calcium ion pump (Chapter
8). The action of this pump is counteracted by a very
slow diffusion of the external Ca
2+
back through the
membrane via an Na
+
Ca
2+
exchange into the cells.
g
Free Ca
2+
lacks spectroscopic properties suitable
for its direct measurement at the low concentrations
present in cells. However, it can be measured indi-
rectly by the use of chelating agents that are relatively
specific for Ca
2+
and which have a measurable prop-
erty that changes upon calcium-binding. The fluo-
rescent photoprotein aequorin (Chapter 23), which
may be injected into cells or synthesized within cells
from transferred genes is often employed.
h,i
Various
synthetic Ca
2+
indicators also fluoresce brilliantly
upon chelation.
j n
Others contain fluorine or a suit-
ably placed atom of
13
C which changes its NMR
chemical shift upon chelation with Ca
2+
.
o
These
compounds may be carried into cells in the form of
esters which pass through membranes but are then
hydrolyzed leaving the indicators trapped in the
cytoplasm.
j
Chelate compounds that bind Ca
2+
within cells and release it rapidly upon irradiation
with ultraviolet light have also been developed.
p,q
Consistent with their role in signaling, calcium
ions are unevenly distributed within cells. Mito-
chondria, endoplasmic reticulum, Golgi, and nuclei
may all take up calcium ions. Cytoplasmic Ca
2+
may sometimes be sequestered in microvesicles
(calciosomes) or in intracellular granules
c,r,s
in
which the [Ca
2+
] is ~ 0.5 1 mM but may reach 5 10
mM.
f
Many regulatory mechanisms exist. For
example, calcitonin and parathyroid hormone
interact with vitamin D and its metabolites in the
small intestine, bones, and kidneys to control the
deposition of calcium in bones, a topic considered
in Box 22-C.
A characteristic function of Ca
2+
in living things
is activation of various metabolic processes. This
occurs when a sudden change in permeability of
the plasma membrane or in the membranes of the
endoplasmic reticulum (ER) allows Ca
2+
to diffuse
into the cytoplasm. For example, during the con-
traction of muscle, the Ca
2+
concentration rises from
~ 0.1 to ~10 M as a result of release from storage in
the calciosomes of the ER. The calcium ions bind to
troponin C initiating muscle contraction (Chapter
19).
t
The ER membranes of muscle are rich in a Ca
2+
pump protein
u,v
(Fig. 8-26), and in a series of calcium-
binding proteins such as calsequestrin (see text).
f
Their combined action soon restores the [Ca
2+
] to
the original low value.
Skeletal muscle is activated by nerve impulses
which induce Ca
2+
release through the action of a
voltage sensor, a protein also known as the dihy-
dropyridine receptor,
w,x
together with a calcium
release channel known as the ryanodine
Calcium chelate compound that releases free calcium ion within
cells upon irradiation with ultraviolet light.
p,q
O
O
O
ON
N
COO COO
HN
COO
COO
N
O
O
O O
N N
O
O
O
O

O
O

O
O

Ca
2+
(free)

Ca
2+
315
BOX 6-D (continued)
receptor.
w,y aa
Inositol trisphosphate (Fig. 11-9),
cyclic ADP-ribose, as well as pH changes
bb
are
involved in controlling these channels,
cc,dd
a topic
discussed in Chapter 11, E. The release of Ca
2+
can
be visualized using high-speed digital imaging
microscopy and fluorescent [Ca
2+
] indicators.
ee,ff
Release of Ca
2+
stored in sea urchin eggs is induced
by cyclic ADP-ribose and by nicotinic acid adenine
dinucleotide phosphate (NAADP
+
, Eq. 15-16).
dd,gg
In a similar manner, when a nerve impulse
reaches a neuron ending (synapse) calcium ions are
released into the cytoplasm and provide the trigger
that causes stored neurotransmitters to be dumped
into the narrow synaptic cleft that separates the
endings of two communicating neurons.
y,hh
The
released neurotransmitter initiates an impulse in the
postsynaptic neuron usually again with an inflow
of Ca
2+
(see Chapter 30).
e
Hormones and various
other compounds often stimulate the flow of calcium
ions into cells.
ii
There Ca
2+
regulates enzymes
jj,kk
(Chapter 11), microtubules, clathrin of coated vesicles
(Chapter 8), K
+
channels in nerve membranes, and
events within mitochondria,
ll,mm
in the ER,
op
and in the
nucleus.
nn,oo
A substantial fraction of these responses
are mediated by calcium-binding regulatory
proteins. Among the most prominent of these is
calmodulin (Fig. 6-8), which, upon binding of Ca
2+
activates a host of metabolic processes
pp
as indicat-
ed in the following scheme, which is modified from
that of Cheung.
qq
In many cases metabolic control by Ca
2+
is mod-
ulated by phosphorylation and dephosphorylation
or other covalent modification of proteins. Such
modification may alter the affinity for Ca
2+
, allow-
ing the latter to either bind and induce a conforma-
tional change or remain unbound,
rr
without a change
in [Ca
2+
]. Nevertheless, the free Ca
2+
concentration
within cells can change greatly and very rapidly.
For example, the oscillatory change in intracellular
[Ca
2+
] shown above was observed in pancreatic
insulin-secreting cells responding to stimulation
by the agonist carbamoylcholine. The free [Ca
2+
]
was evaluated from fluorescence measurements
using the Ca
2+
indicator dye fura 2 (From Prentki
et al
ss
). Oscillations in [Ca
2+
] have been observed
under many circumstances.
ff,oo,tt ww
These are of particular interest
because of the possible relation-
ship to the initiation of oscillatory
nerve conduction (Chapter 30).
uu
Released Ca
2+
often appears
to move across cells in waves
and sometimes to be released
as puffs or sparks.
ee,ff,oo
Adenylate
cyclase
Phosphodiesterase
Phospholipase A
2
Ca
2+
ATPase (calcium pump)
Microtubule disassembly
Membrane phosphorylation
IRS-1 (insulin signalling)
Neurotransmitter release
Myosin light chain kinase
Phosphorylase kinase
Guanylate cyclase
Ca
2+
-dependent protein kinase
NAD
kinase
Calcineurin (phosphatase)
Smooth muscle activation
NO synthase Calmodulin
a
Bianchi, C. P. (1968) Cell Ca
++
, Appleton, New York
b
Means, A. R., ed. (1994) Calcium Regulation of Cellular Function,
Lippincott-Raven, Hagerstown, Maryland
c
Volpe, P., Krause, K.-H., Hashimoto, S., Zorzato, F., Pozzan, T.,
Meldolesi, J., and Lew, D. P. (1988) Proc. Natl. Acad. Sci. U.S.A.
85, 10911095
d
Carafoli, E. (1987) Ann. Rev. Biochem. 56, 395433
e
Ghosh, A., and Greenberg, M. E. (1995) Science 268, 239247
f
Meldolesi, J., and Pozzan, T. (1998) Trends Biochem. Sci. 23,
1014
g
Khananshvili, D. (1991) J. Biol. Chem. 266, 1376413769
(continued)
E. Complex Biochemical Equilibria
60
90
120
150
180
F
l
u
o
r
e
s
c
e
n
c
e

i
n

a
r
b
i
t
r
a
r
y

u
n
i
t
s
0 50 100 150 200 250 300 350 400
Time (s)
Calcium oscillations observed with six cultured pancreatic cells
after a single infusion of 0.2 mM carbamoylcholine. The fluores-
cence intensity of the Ca
2+
indicator dye fura 2, with excitation at
380 nm, was recorded versus time. From Pretki et al.
ss
316 Chapter 6. Thermodynamics and Biochemical Equilibria
BOX 6-D (continued)
h
Rizzuto, R., Simpson, A. W. M., Brini, M., and Pozzan, T.
(1992) Nature (London) 358, 325327
i
Knight, M. R., Campbell, A. K., Smith, S. M., and Trewavas,
A. J. (1991) Nature (London) 352, 524 526
j
Tsien, R. Y., Pozzan, T., and Rink, T. J. (1984) Trends Biochem.
Sci. 9, 263266
k
Davies, P. L., and Hew, C. L. (1990) FASEB J. 4, 24602468
l
Ganz, M. B., Rasmussen, J., Bollag, W. B., and Rasmussen, H.
(1990) FASEB J. 4, 16381644
m
Lloyd, Q. P., Kuhn, M. A., and Gay, C. V. (1995) J. Biol. Chem.
270, 2244522451
n
Etter, E., Minta, A., Poenie, M., and Fay, F. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 53685373
o
Robitaille, P.-M. L., and Jiang, Z. (1992) Biochemistry 31, 12585
12591
p
Ellis-Davies, G. C. R., and Kaplan, J. H. (1994) Proc. Natl. Acad.
Sci. U.S.A. 91, 187191
q
Ellis-Davies, G. C. R., Kaplan, J. H., and Barsotti, R. J. (1996)
Biophys. J. 70, 10061016
r
Burgoyne, R. D., and Cheek, T. R. (1991) Trends Biochem. Sci.
16, 319320
s
Golovina, V. A., and Blaustein, M. P. (1997) Science 275, 1643
1648
t
Gagn, S. M., Li, M. X., and Sykes, B. D. (1997) Biochemistry 36,
43864392
u
MacLennan, D. H., Rice, W. J., and Green, N. M. (1997) J. Biol.
Chem. 272, 2881528818
v
Yonekura, K., Stokes, D. L., Sasabe, H., and Toyoshima, C.
(1997) Biophys. J. 72, 9971005
w
Yano, M., El-Hayek, R., and Ikemoto, N. (1995) J. Biol. Chem.
270, 30173021
x
Mitterdorfer, J., Sinnegger, M. J., Grabner, M., Striessnig, J.,
and Glossmann, H. (1995) Biochemistry 34, 93509355
y
McPherson, P. S., and Campbell, K. P. (1993) J. Biol. Chem. 268,
1376513768
z
Sakube, Y., Ando, H., and Kagawa, H. (1997) J. Mol. Biol. 267,
849864
aa
Zorzato, F., Fujii, J., Otsu, K., Phillips, M., Green, N. M., Lai, F.
A., Meissner, G., and MacLennan, D. H. (1990) J. Biol. Chem.
265, 22442256
bb
Donoso, P., Beltrn, M., and Hidalgo, C. (1996) Biochemistry 35,
1341913425
cc
Lee, M. G., Xu, X., Zeng, W., Diaz, J., Wojcikiewicz, R. J. H.,
Kuo, T. H., Wuytack, F., Racymaekers, L., and Muallem, S.
(1997) J. Biol. Chem. 272, 1576515770
dd
Graeff, R. M., Franco, L., De Flora, A., and Lee, H. C. (1998)
J. Biol. Chem. 273, 118125
ee
Isenberg, G., Etter, E. F., Wendt-Gallitelli, M.-F., Schiefer, A.,
Carrington, W. A., Tuft, R. A., and Fay, F. S. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 54135418
ff
Horne, J. H., and Meyer, T. (1997) Science 276, 16901693
gg
Lee, H. C., and Aarhus, R. (1997) J. Biol. Chem. 272, 20378
20383
hh
Miller, R. J. (1992) J. Biol. Chem. 267, 14031406
ii
Quitterer, U., Schrder, C., Mller-Esterl, W., and Rehm, H.
(1995) J. Biol. Chem. 270, 19921999
jj
Suzuki, K. (1987) Trends Biochem. Sci. 12, 103105
kk
Martin, B. L., and Graves, D. J. (1986) J. Biol. Chem. 261, 14545
14550
ll
Moudy, A. M., Handran, S. D., Goldberg, M. P., Ruffin, N.,
Karl, I., Kranz-Eble, P., DeVivo, D. C., and Rothman, S. M.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 729733
mm
Kasumi, T., Tsumuraya, Y., Brewer, C. F., Kersters-Hilderson,
H., Claeyssens, M., and Hehre, E. J. (1987) Biochemistry 26,
30103016
nn
Santella, L., and Carafoli, E. (1997) FASEB J. 11, 10911109
oo
Lipp, P., Thomas, D., Berridge, M. J., and Bootman, M. D.
(1997) EMBO J. 16, 71667173
op
Corbett, E. F., and Michalak, M. (2000) Trends Biochem. Sci. 25,
307311
pp
Ikura, M., Clore, G. M., Gronenborn, A. M., Zhu, G., Klee, C.
B., and Bax, A. (1992) Science 256, 632638
qq
Cheung, W. Y. (1980) Science 207, 1927
rr
Geisow, M. J. (1987) Trends Biochem. Sci. 12, 120121
ss
Prentki, M., Glennon, M. C., Thomas, A. P., Morris, R. L.,
Matschinsky, F. M., and Corkey, B. E. (1988) J. Biol. Chem. 263,
1104411047
tt
Tang, Y., and Othmer, H. G. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 78697873
uu
Putney, J. W., Jr. (1998) Science 279, 191192
vv
Berridge, M. J. (1997) Nature (London) 386, 759760
ww
Hajnczky, G., and Thomas, A. P. (1997) EMBO J. 16, 35333543
increase above the normal level in keratinocytes of
patients with the skin disease psoriasis.
123b
Calcium ions are often involved in holding nega-
tively charged groups together, for example, in the
binding of proteins to phospholipid membranes. Among
these membrane-associated proteins are the vitamin K-
dependent proteins, all of which contain several residues
of the chelating amino acid -carboxyglutamate (Chapter
15) at their calcium-binding sites.
124,125
Many of these
are involved in the clotting of blood (Chapter 12). Some
of the membrane-binding proteins called annexins
(Chapter 8) are also Ca
2+
-dependent ion channels.
126,127
Other Ca
2+
-requiring lipid-binding proteins include the
lipocortins, calpactins, and calelectrins.
128
Cadherins
bind Ca
2+
and help provide cohesion between cells.
129
Many proteins contain bound Ca
2+
in precisely defined
sites where it plays a structural role. These include the
galactose-binding protein of bacterial transport and
chemotaxis (Fig. 4-18)
130
and -lactalbumin of milk.
Although -lactalbumin does not contain the helix
loophelix pattern, its Ca
2 +
-binding site does consist
of three carboxylate groups and two backbone carbonyl
oxygen atoms.
131
The -amylases, thermolysin, and
staphylococcal nuclease (Chapter 12), and the lectin
favin (Fig. 2-15) all contain bound Ca
2+
. Calcium ions
also bind to anionic groups in carbohydrates, e.g., to the
sulfate groups in carageenin gels (Chapter 4) where they
provide structural stability.
317
Figure 6-8 Stereoscopic backbone
trace of a 148-residue recombinant
calmodulin. The two helixturn
helix (EF-hand) loops and their bound
Ca
2+
(as concentric circles) are at the
top and the two near the C terminus
are at the bottom. The long central
helix, seen in this crystal structure,
may undergo conformational changes
during the functioning of this Ca
2+
-
sensing molecule.
132
From Chatto-
padhyaya et al.
111
Courtesy of F. A.
Quiocho.
E. Complex Biochemical Equilibria
BOX 6-E METALLOTHIONEINS
If animals ingest excessive
amounts of Zn(II), Cd (II), Hg(II),
or Cu(I) their livers and kidneys
accumulate these metals as com-
plexes of proteins called metallo-
thioneins.
a e
In mammals at least
three related genes encode these
metal-binding proteins. The best
known, metallothionein II, has
a highly conserved 61-residue
sequence containing 20 cysteine
residues and no aromatic residues.
The protein is organized into two
domains, each able to bind a cluster
of metal ions via thiolate side chains.
The three-dimensional structure of
rat liver metallothionein containing
five Cd
2+
and two Zn
2+
ions is shown
in the accompanying stereoscopic
diagram.
f
The 61 alpha carbons,
the beta carbon and sulfur atoms
(green) of cysteine residues and
the bound metal ions are indicated.
The N-terminal domain (residues 1 29) contains one
Cd
2+
and two Zn
2
and nine cysteine sulfurs which
bind the metal ions. Three of the sulfur atoms form
bridges between pairs of metals. The second cluster
contains four Cd
2+
held by 11 cysteine sulfur atoms,
Metallothionein containing bound Cd
2+
and Zn
2+
. From Robbins et al.
f
five of which bridge between pairs of metals. All of
the metal ions are tetrahedrally coordinated. The
polypeptide chains of metallothioneins consist pre-
dominantly of beta turns.
g
Important techniques in
the study of these proteins include
113
Cd NMR,
h,i
318 Chapter 6. Thermodynamics and Biochemical Equilibria
BOX 6-E METALLOTHIONEINS (continued)
a
Mehra, R. K., Garey, J. R., Butt, T. R., Gray, W. R., and Winge,
D. R. (1989) J. Biol. Chem. 264, 1974719753
b
Kgi, J. H. R., and Kojima, Y., eds. (1987) Metallothionein II,
Birkhuser, Basel, Berlin
c
Fischer, E. H., and Davie, E. W. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 3333 3334
d
Kgi, J. H. R., and Schffer, A. (1988) Biochemistry 27, 85098515
spectroscopic methods,
j,k
and X-ray absorption.
l
Transcription of metallothionein genes is induced
by metal ions, and toxic metals such as Cd and Hg
accumulate as metallothionein complexes, suggesting
that one function is to protect against metal toxicity.
m
However, synthesis is also induced by glucocorticoid
hormones,
n
and accumulation of a high concentration
of copper and zinc in fetal metallothionein suggests a
role in storage of these essential metals.
c
Another metal
that binds to metallothioneins is Au(I),
o
which is widely
used in thiolate salts as a chemotherapeutic agent for
rheumatoid arthritis.
Metallothioneins are also found in insects, lower
invertebrates, and even in bacteria.
i,p
Nevertheless,
there are other metal-binding proteins.
q,r
For example,
albacore tuna contain a 66-kDa glycoprotein that con-
tains eight mole percent histidine and binds three Zn
2+
,
each by a cluster of three His.
s
Plants and fungi contain
phytochelins, peptides consisting largely of repeated
-glutamylcysteine units.
a,t,u
These appear to protect
plants against toxicity of cadmium in the same manner
as do the metallothioneins in our bodies.
e
Hamer, D. H. (1986) Ann. Rev. Biochem. 55, 913951
f
Robbins, A. H., McRee, D. E., Williamson, M., Collett, S. A.,
Xuong, N. H., Furey, W. F., Wang, B. C., and Stout, C. D. (1991)
J. Mol. Biol. 221, 12691293
g
Pande, J., Pande, C., Gilg, D., Vask, M., Callender, R., and
Kgi, J. H. R. (1986) Biochemistry 25, 55265532
h
Cismowski, M. J., Narula, S. S., Armitage, I. M., Chernaik, M.
L., and Huang, P. C. (1991) J. Biol. Chem. 266, 2439024397
i
Vask, M., Hawkes, G. E., Nicholson, J. K., and Sadler, P. J.
(1985) Biochemistry 24, 740747
j
Willner, H., Vask, M., and Kgi, J. H. R. (1987) Biochemistry 26,
62876292
k
Lu, W., and Stillman, M. J. (1993) J. Am. Chem. Soc. 115, 3291
3299
l
George, G. N., Byrd, J., and Winge, D. R. (1988) J. Biol. Chem.
263, 81998203
m
Sadhu, C., and Gehamu, L. (1988) J. Biol. Chem. 263, 26792684
n
Karin, M., Haslinger, A., Holtgreve, H., Richards, R. I., Krauter,
P., Westphal, H. M., and Beato, M. (1984) Nature (London) 308,
513519
o
Laib, J. E., Shaw, C. F., III, Petering, D. H., Eidsness, M. K.,
Elder, R. C., and Garvey, J. S. (1985) Biochemistry 24, 19771986
p
Freedman, J. H., Slice, L. W., Dixon, D., Fire, A., and Rubin, C. S.
(1993) J. Biol. Chem. 268, 25542564
q
Waalkes, M. P., and Peratoni, A. (1986) J. Biol. Chem. 261, 13097
13103
r
Willuhn, J., Schmitt-Wrede, H. P., Greven, H., and Wunderlich,
F. (1994) J. Biol. Chem. 269, 2468824691
s
Dyke, B., Hegenauer, J., Saltman, P., and Laurs, R. M. (1987)
Biochemistry 26, 32283234
t
Reese, R. N., and Winge, D. R. (1988) J. Biol. Chem. 263, 12832
12835
u
Clemens, S., Kim, E. J., Neumann, D., and Schroeder, J. I. (1999)
EMBO J. 18, 3325 3333
319
1. Lewis, G. N., and Randall, M. (1961)
Thermodynamics and the Free Energy of Chemical
Substances, 2nd ed., McGraw-Hill, New York
2. Mahan, B. H. (1963) Elementary Chemical
Thermodynamics, Benjamin, New York
3. Everdell, M. H. (1965) Introduction to Chemical
Thermodynamics, Norton, New York
4. Dickerson, R. E. (1969) Molecular Thermody-
namics, Benjamin, New York
5. Jones, M. N., ed. (1988) Biochemical Thermody-
namics, 2nd ed., Elsevier, Amsterdam
6. Hinz, H.-J., ed. (1986) Thermodynamic Data for
Biochemistry and Biotechnology, Springer-
Verlag, Berlin
7. Gutfreund, H., and Edsall, J. T. (1983)
Biothermo-dynamics, Wiley, New York
8. Harold, F. M. (1986) The Vital Force: A Study of
Bioenergetics, Freeman, San Francisco,
California
9. Garby, L., and Larsen, P. S. (1995) Bioenerget-
ics; Its Thermodynamic Foundations, Cambridge
Univ. Press, London and New York
10. Jou, D., and Llebot, J. E., eds. (1990)
Introduction to the Thermodynamics of Biological
Processes, Prentice Hall, Englewood Cliffs,
New Jersey
10a. Wads, I. (1985) Eur. J. Biochem. 153, 429434
11. Sturtevant, J. M. (1971) in Techniques of
Chemistry, Vol. I, Part V (Weissberger, A., ed),
pp. 347425, Wiley, New York
12. Brown, H. D., ed. (1969) Biochemical
Microcalorimetry, Academic Press, New York
13. Peters, K. S., and Snyder, G. J. (1988) Science
241, 10531057
14. White, A., Handler, P., and Smith, E. L. (1968)
Principles of Biochemistry, 4th ed., McGraw-
Hill, New York (pp. 291301)
15. Mosteller, R. D. (1988) N. Engl. J. Med. 318,
1130
16. Donata, K., and Hegsted, D. M. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 48664870
17. Ben-Naim, A. (1992) Statistical Thermodynam-
ics for Chemists and Biochemists, Plenum, New
York
18. Donovan, J. W. (1984) Trends Biochem. Sci. 9,
340344
19. Tanaka, A., Flanagan, J., and Sturtevant, J. M.
(1993) Protein Sci. 2, 567576
20. Pace, C. N., and Laurents, D. V. (1989)
Biochemistry 28, 25202525
21. Interunion Commission on
Biothermodynamics. (1976) J. Biol. Chem. 251,
68796885
22. Phillips, R. C., George, P., and Rutman, R. J.
(1963) Biochemistry 2, 501508
23. Liu, Y., and Sturtevant, J. M. (1995) Protein Sci.
4, 25592561
24. Halling, P. J. (1989) Trends Biochem. Sci. 14,
317318
25. rstan, A. (1990) Trends Biochem. Sci. 15, 137
138
26. Nicolis, G., and Prigogine, I. (1977) Self-
organization in Non-equilibrium Systems: From
Dissipative Structure to Order Through
Fluctuations, Wiley-Interscience, New York
27. Procaccia, I., and Ross, J. (1977) Science 198,
716717
28. Morowitz, H. J. (1968) Energy Flow in Biology,
Academic Press, New York
29. Caplan, S. R., and Essig, A. (1983) Bioenergetics
and Linear Nonequilibrium Thermodynamics/ The
Steady State, Harvard Univ. Press, Cambridge,
Massachusetts
30. Peacocke, A. R. (1983) An Introduction to the
Physical Chemistry of Biological Organization,
Clarendon Press, Oxford
31. Blumenfeld, L. A. (1983) Physics of Bioenergetic
Processes, Springer-Verlag, Berlin
References
32. Katchalsky, A., and Curran, P. F. (1965) Non-
equilibrium Thermodynamics in Biophysics,
Harvard Univ. Press, Cambridge, Massachu-
setts
33. Prigogine, I. (1967) Introduction to Thermody-
namics of Irreversible Processes, 3rd ed., Wiley,
New York
34. Coveney, P. V. (1988) Nature (London) 333,
409415
35. Wilson, D. F. (1982) Trends Biochem. Sci. 7,
275278
36. Westerhoff, H. V. (1982) Trends Biochem. Sci. 7,
275279
37. Guynn, R. W., and Veech, R. L. (1973) J. Biol.
Chem. 248, 69666972
38. Alberty, R. A. (1969) J. Biol. Chem. 244, 3290
3302
39. Phillips, R. C., George, P., and Rutman, R. J.
(1969) J. Biol. Chem. 244, 33303342
40. George, P., Phillips, R. C., and Rutman, R. J.
(1963) Biochemistry 2, 508512
41. Purich, D. L., and Fromm, H. J. (1972) Curr.
Top. Cell. Regul. 6, 131167
42. Jencks, W. P. (1968) in Handbook of Biochemistry
(Sober, H. A., ed), pp. J-148, CRC, Cleveland,
Ohio
43. Bassham, J. A., and Krause, G. H. (1969)
Biochim. Biophys. Acta. 189, 207221
44. Rosing, J., and Slater, E. C. (1972) Biochim.
Biophys. Acta. 267, 275290
45. Alberty, R. A. (1992) Biophysical Chem. 42, 117
131
46. Alberty, R. A., and Cornish-Bowden, A. (1993)
Trends Biochem. Sci. 18, 288291
47. Alberty, R. A., and Goldberg, R. N. (1992)
Biochemistry 31, 1061010615
48. Alberty, R. A. (1994) Pure & Appl. Chem. 66,
16411666
49. Stokes, G. B. (1988) Trends Biochem. Sci. 13,
422424
50. Mavrovouniotis, M. L. (1991) J. Biol. Chem.
266, 1444014445
51. Dutton, P. L. (1971) Biochim. Biophys. Acta. 226,
6380
52. Pepin, C. A., and Wood, H. G. (1986) J. Biol.
Chem. 261, 44764480
53. Wood, H. G., and Clark, J. E. (1988) Ann. Rev.
Biochem. 57, 235260
54. Crooke, E., Akiyama, M., Rao, N. N., and
Kornberg, A. (1994)
J. Biol. Chem. 269, 62906295
55. Veech, R. L., Lawson, J. W. R., Cornell, N. W.,
and Krebs, H. A. (1979) J. Biol. Chem. 254,
65386547
56. Swedes, J. S., Sedo, R. J., and Atkinson, D. E.
(1975) J. Biol. Chem. 250, 69306938
57. Chapman, A. G., Fall, L., and Atkinson, D. E.
(1971) J. Bacteriol. 108, 10721086
58. Purich, D. L., and Fromm, H. J. (1973) J. Biol.
Chem. 248, 461466
59. Teague, W. E., Jr., and Dobson, G. P. (1992) J.
Biol. Chem. 267, 1408414093
60. Cohen, S. M., Ogawa, S., Rottenberg, H.,
Glynn, P., Yamane, T., Brown, T. R., and
Shulman, R. G. (1978) Nature (London) 273,
554556
60a. Ugurbil, K., Kingsley-Hickman, P. B., Sako, E.
Y., Zimmer, S., Mohanakrishnan, P., Robitaille,
P. M. L., Thoma, W. J., Johnson, A., Foker, J. E.,
and From, A. H. L. (1987) Ann. N.Y. Acad. Sci.
508, 265 286
61. Brosnan, M. J., Chen, L., Van Dyke, T. A.,
and Koretsky, A. P. (1990) J. Biol. Chem. 265,
2084920855
61a. Saupe, K. W., Spindler, M., Hopkins, J. C. A.,
Shen, W., and Ingwall, J. S. (2000) J. Biol. Chem.
275, 19742 19746
62. Hutson, S. M., Williams, G. D., Berkich, D. A.,
LaNoue, K. F., and Briggs, R. W. (1992)
Biochemistry 31, 13221330
63. Burt, C. T., Glonek, T., and Brny, M. (1977)
Science 195, 145149
64. Seeley, P. J., Sehr, P. A., Gadian, D. G., Garlick,
P. B., and Radda, G. K. (1977) in NMR in
Biology (Dwek, R. A., Campbell, I. D.,
Richards, R. E., and Williams, R. J. P., eds), pp.
247275, Academic Press, London
65. Masson, S., and Quistorff, B. (1992)
Biochemistry 31, 7488 7493
66. Jeffrey, F. M., Storey, C. J., Nunnally, R. L., and
Malloy, C. R. (1989) Biochemistry 28, 53235326
67. Kagimoto, T., Higaki, T., Nagata, K., Morino, Y.,
and Takatsuki, K. (1989) NMR in Biomedicine 2,
9397
68. Park, J. H., Brown, R. L., Park, C. R., Cohn,
M., and Chance, B. (1988) Proc. Natl. Acad. Sci.
U.S.A. 85, 87808784
69. Radda, G. K. (1992) FASEB J. 6, 30323038
70. Radda, G. K. (1986) Science 233, 640645
71. Burt, C. T. (1985) Trends Biochem. Sci. 10, 404
406
72. Grove, T. H., Ackerman, J. J. H., Radda, G. K.,
and Bore, P. J. (1980) Proc. Natl. Acad. Sci.
U.S.A. 77, 299302
73. Bottomley, P. A. (1985) Science 229, 769772
74. Fossel, E. T., Morgan, H. E., and Ingwall, J. S.
(1980) Proc. Natl. Acad. Sci. U.S.A. 77, 3654
3658
75. Metzler, D. E., Harris, C. M., Johnson, R. J.,
Siano, D. B., and Thomson, J. A. (1973)
Biochemistry 12, 53775392
76. Johnson, R. J., and Metzler, D. E. (1970)
Methods Enzymol. 18, 433471
77. Dixon, H. B. F. (1992) Essays in Biochemistry 27,
161176
78. Vander Jagt, D. L., Hansen, L. D., Lewis, E. A.,
and Han, L. B. (1972) Arch. Biochem. Biophys.
153, 5561
79. Dixon, H. B. F., and Tipton, K. F. (1973)
Biochem. J. 133, 837842
80. Klotz, I. M., and Hunston, D. L. (1975) J. Biol.
Chem. 250, 30013009
81. Weber, G. (1975) Adv. Prot. Chem. 29, 183
82. Chivers, P. T., Prehoda, K. E., Volkman, B. F.,
Kim, B.-M., Markley, J. L., and Raines, R. T.
(1997) Biochemistry 36, 1498514991
83. Johnson, F. A., Lewis, S. D., and Shafer, J. A.
(1981) Biochemistry 20, 4448
84. McIntosh, L. P., Hand, G., Johnson, P. E.,
Joshi, M. D., Krner, M., Plesniak, L. A., Ziser,
L., Wakarchuk, W. W., and Withers, S. G.
(1996) Biochemistry 35, 99589966
85. Katritzky, A. R., and Waring, A. (1962) J.
Chem. Soc., 15401548
86. Katritzky, A. R., and Karelson, M. (1991) J.
Am. Chem. Soc. 113, 15611566
87. Bjerrum, J. (1941) Metal Amine Formation in
Aqueous Solution, Haase & Son, Copenhagen
88. Eichhorn, G. L., ed. (1973) Inorganic
Biochemistry, 2 vols, Elsevier, Amsterdam
89. Martell, A. E., and Smith, R. M. (1975) Critical
Stability Constants, Plenum, New York
90. Michaelian, K. H., and Moskovits, M. (1978)
Nature (London) 273, 135136
91. Lewsin, S. (1974) Displacement of Water and its
Control in Biochemical Reactions, Academic
Press, London
92. Karlin, S., Zhu, Z.-Y., and Karlin, K. D. (1997)
Proc. Natl. Acad. Sci. U.S.A. 94, 1422514230
93. Freeman, H. C., Healy, M. J., and Scudder, M.
L. (1977) J. Biol. Chem. 252, 88408847
94. Torrado, A., Walkup, G. K., and Imperiali, B.
(1998) J. Am. Chem. Soc. 120, 609610
95. Nakayama, S., and Kretsinger, R. H. (1994)
Annu. Rev. Biophys. Biomol. Struct. 23, 473507
320 Chapter 6. Thermodynamics and Biochemical Equilibria
References
96. Kretsinger, R. H. (1976) Ann. Rev. Biochem. 45,
239266
96a. Pawlowski, K., Bierzynski, A., and Godzik, A.
(1996) J. Mol. Biol. 258, 349366
97. Ikura, M. (1996) Trends Biochem. Sci. 21, 1417
98. Kretsinger, R. H., and Nockolds, C. E. (1973) J.
Biol. Chem. 248, 33133326
99. McPhalen, C. A., Sielecki, A. R., Santarsiero,
B. D., and James, M. N. G. (1994) J. Mol. Biol.
235, 718732
99a. Declercq, J.-P., Evrard, C., Lamzin, V., and
Parello, J. (1999) Protein Sci. 8, 2194 2204
99b. Biekofsky, R. R., Martin, S. R., Browne, J. P.,
Bayley, P. M., and Feeney, J. (1998) Biochemis-
try 37, 7617 7629
100. Drake, S. K., Zimmer, M. A., Miller, C. L., and
Falke, J. J. (1997) Biochemistry 36, 99179926
101. Akke, M., Forsn, S., and Chazin, W. J. (1995)
J. Mol. Biol. 252, 102121
102. Denisov, V. P., and Halle, B. (1995) J. Am.
Chem. Soc. 117, 84568465
103. Heizmann, C. W., and Hunziker, W. (1991)
Trends Biochem. Sci. 16, 98103
104. Ikemoto, N., Ronjat, M., Mszros, L. G., and
Koshita, M. (1989) Biochemistry 28, 67646771
105. Meldolesi, J., and Pozzan, T. (1998) Trends
Biochem. Sci. 23, 1014
106. Randolph, J. T., McClure, K. F., and
Danishefsky, S. J. (1995) J. Am. Chem. Soc. 117,
57125719
107. Fliegel, L., Burns, K., MacLennan, D. H.,
Reithmeier, R. A. F., and Michalak, M. (1989)
J. Biol. Chem. 264, 2152221528
108. Cohen, P., and Klee, C. B., eds. (1988)
Calmodulin, Elsevier, Amsterdam
109. James, P., Vorherr, T., and Carafoli, E. (1995)
Trends Biochem. Sci. 20, 3842
110. Lafitte, D., Capony, J. P., Grassy, G., Haiech, J.,
and Calas, B. (1995) Biochemistry 34, 13825
13832
111. Chattopadhyaya, R., Meador, W. E., Means,
A. R., and Quiocho, F. A. (1992) J. Mol. Biol.
228, 11771192
112. Pedigo, S., and Shea, M. A. (1995) Biochemistry
34, 1067610689
113. da Silva, A. C. R., and Reinach, F. C. (1991)
Trends Biochem. Sci. 16, 5357
114. Gagn, S. M., Li, M. X., and Sykes, B. D. (1997)
Biochemistry 36, 43864392
115. Trigo-Gonzalez, G., Awang, G., Racher, K.,
Neden, K., and Borgford, T. (1993) Biochemis-
try 32, 98269831
116. Spyracopoulos, L., Li, M. X., Sia, S. K., Gagn,
S. M., Chandra, M., Solaro, R. J., and Sykes, B.
D. (1997) Biochemistry 36, 1213812146
117. Trav, G., Lacombe, P.-J., Pfuhl, M., Saraste,
M., and Pastore, A. (1995) EMBO J. 14, 4922
4931
118. Nthke, I., Hill, B. L., Parham, P., and Brodsky,
F. M. (1990) J. Biol. Chem. 265, 1862118627
119. Pley, U. M., Hill, B. L., Alibert, C., Brodsky, F.
M., and Parham, P. (1995) J. Biol. Chem. 270,
23952402
120. Engel, J., Taylor, W., Paulsson, M., Sage, H.,
and Hogan, B. (1987) Biochemistry 26, 6958
6965
121. Engel, E., Richter, K., Obermeyer, G., Briza, P.,
Kungl, A. J., Simon, B., Auer, M., Ebner, C.,
Rheinberger, H.-J., Breitenbach, M., and
Ferreira, F. (1997) J. Biol. Chem. 272, 28630
28637
122. Schfer, B. W., and Heizmann, C. W. (1996)
Trends Biochem. Sci. 21, 134140
123. Drohat, A. C., Baldisseri, D. M., Rustandi, R.
R., and Weber, D. J. (1998) Biochemistry 37,
2729 2740
123a. Gribenko, A. V., and Makhatadze, G. I. (1998)
J. Mol. Biol. 283, 679 694
123b. Brodersen, D. E., Nyborg, J., and Kjeldgaard,
M. (1999) Biochemistry 38, 1695 1704
124. Christiansen, W. T., Tulinsky, A., and
Castellino, F. J. (1994) Biochemistry 33, 14993
15000
125. Schwalbe, R. A., Ryan, J., Stern, D. M., Kisiel,
W., Dahlbck, B., and Nelsestuen, G. L. (1989)
J. Biol. Chem. 264, 2028820296
126. Demange, P., Voges, D., Benz, J., Liemann, S.,
Gttig, P., Berendes, R., Burger, A., and Huber,
R. (1994) Trends Biochem. Sci. 19, 272276
127. Saurel, O., Czanne, L., Milon, A., Tocanne,
J.-F., and Demange, P. (1998) Biochemistry 37,
14031410
128. Moss, S. E., and Crumpton, M. J. (1990) Trends
Biochem. Sci. 15, 1112
129. Shapiro, L., Fannon, A. M., Kwong, P. D.,
Thompson, A., Lehmann, M. S., Grbel, G.,
Legrand, J.-F., Als-Nielsen, J., Colman, D. R.,
and Hendrickson, W. A. (1995) Nature
(London) 374, 327336
130. Vyas, N. K., Vyas, M. N., and Quiocho, F. A.
(1988) Science 242, 12901295
131. Stuart, D. I., Acharya, K. R., Walker, N. P. C.,
Smith, S. G., Lewis, M., and Phillips, D. C.
(1986) Nature (London) 324, 8487
132. Williams, R. J. P. (1991) Trends Biochem. Sci. 16,
206
321
Study Questions
1. a) From G for hydrolysis of sucrose (Table 6-6)
calculate the equilibrium constant
= [glucose][fructose] / [sucrose]
at 25C. Call this hydrolysis reaction 1.
b) Is the sucrose in a 1 solution stable? Explain.
c) If acid is added to a 1 sucrose solution to
catalyze its hydrolysis, what will be the final
sucrose concentration at equilibrium? (Assume
that concentrations equal activities for the
purpose of these calculations.)
d) Reaction 2 is the hydrolysis of -D-glucose
1-phosphate to glucose and inorganic phos-
phate (P
i
). Using G for this reaction (Table
6-6) calculate the equilibrium constant.
e) Sucrose phosphorylase from the bacterium
Pseudomonas saccharophila catalyzes the follow-
ing reaction (reaction 3):
Sucrose + P
1

-D-glucose 1-phosphate + fructose


Calculate the equilibrium constant and the
standard Gibbs energy change at 25C for
reaction 3 from the equilibrium constants
obtained above for reactions 1 and 2. Show
that G for reaction 3 = G of reaction 1 - G
of reaction 2.
f) Could the bacterium carry out reaction 3 in the
following two consecutive steps? Explain.
Sucrose glucose + fructose
Glucose + P
i
glucose 1-phosphate
2. For each of the following reactions, state whether
the equilibrium constant will be between 0.1 and
10 (i.e., about one), greater than 100, or less that
0.01. Assume that the pH is constant at 7.0.
a) 2 ADP
3
ATP
4
+ AMP
2
b) ATP
3
+ glucose glucose 6-phosphate
2
+
ADP
2
+ H
+
c) ADP
2
+ HPO
4
2
+ H
+
ATP
3
d) Glucose 6-phosphate
2

fructose 6-phosphate
2
e) Phosphoenolpyruvate
3
+ glucose glucose
1-phosphate
2
+ pyruvate

3. The combustion of 1 mol of solid urea to liquid


water and gaseous carbon dioxide and nitrogen
(N
2
) in a bomb colorimeter at 25C (constant
volume) liberated 666 kJ of heat energy. Calculate
H, the change in heat content (enthalpy), for this
reaction.
4. Using data of Table 6-4 calculate G (pH 7) for the
following reactions:
a) Glucose 2 lactate

+ 2 H
+
b) 2 NH
4
+
+ HCO
3

urea + 2 H
2
O + H
+
c) 2-oxoglutarate
2
+ 1/ 2 O
2
+ CoA + H
+

succinyl-CoA + H
2
O + CO
2
5. What is the ionic strength of a 0.2 Msolution of
NaCl? of 0.2 M Na
2
SO
4
?
6. The [ATP] / [ADP] ratio in an actively respiring
yeast cell is about 10. What would be the intracel-
lular [3-phosphoglycerate] / [1,3-bisphospho-
glycerate] ratio have to be to make the phospho-
glycerate kinase reaction (Fig. 9-7, reaction 7)
proceed toward 1,3-bisphosphoglycerate synthesis
at 25C, pH 7?
7. a) Using data from Table 6-8 determine the equi-
librium constant for the reaction between
malate and methylene blue, assuming all
reactants present initially at the same concen-
tration. Indicate clearly the direction of the
reaction for which the Gibbs energy change is
written.
b) Calculate the percentage of the reduced (leuco)
form of methylene blue present at pH 7 and
25C in a system for which the measured
electrode potential is 0.065 V.
8. NAD
+
is a coenzyme for both pyruvate dehydro-
genase and ethanol dehydrogenase. Using the
values of
o
from Table 6-8 calculate the Gibbs
energy change and the equilibrium constant for
the reaction.
Lactate

+ acetaldehyde pyruvate

+ ethanol
9. Consider the oxidation of acetate at 25C:
CH
3
COO

(0.1 M) + 2 O
2
(g, 0.2 atm) +
H
+
(10
7
M) 2 H
2
O (l) + 2 CO
2
(g)
a) What is the equilibrium pressure of CO
2
if the
reaction is not coupled to any other reaction?
b) What is the equilibrium pressure of CO
2
if the
reaction is coupled to the formation of 0.01 M
ATP from 0.02 MADP and 0.01 MHPO
4
2-
in
the citric acid cycle?
c) What do the above calculations tell you about
the prospects of gaining 100% efficiency of
energy storage in ATP from the citric acid cycle?
d) If the actual pressure of CO
2
is 0.01 atm, what
is the efficiency of energy storage under the
conditions in (b)?
322 Chapter 6. Thermodynamics and Biochemical Equilibria
10. The equilibrium constant for the following reac-
tion, which is catalyzed by creatine kinase, has
been determined by chemical analysis. The data
are given below. [S. A. Kuby and E. A. Noltman, in
The Enzymes, 2nd ed. (P. D. Boyer, H. Lardy, and K.
Myrbck, eds), Vol. VI, pp. 515 602. Academic
Press, New York, 1962]
ATP
4
+ creatine ADP
3
+ creatine phosphate
2
+ H
+
t (C) K
20 6.30 x 10
9
30 5.71 x 10
9
38 5.47 x 10
9
a) What are G, H, and S for the reaction at
25C?
b) What are G, H, and S (pH 7) for the
reaction at 25C?
c) What are G, H, and S (pH 7) for the
hydrolysis of creatine phosphate at 25C?
11. The following reaction was carried out in a
calorimeter at 25C in 0.1 M phosphate buffer at
pH 7.4 in the presence of a particulate suspension
containing the mitochondrial electron transport
system [M. Poe, H. Gutfreund, and R. W.
Estabrook, ABB 122, 204 211 (1967)]:
NADH + H
+
+ 1/ 2 O
2
(aq., sat.)
NAD
+
+ H
2
O
The oxygen consumption was monitored continu-
ously with an oxygen electrode. The temperature
was monitored simultaneously with a thermo-
couple immersed in the solution. At the start of
the reaction 96 mol NADH was added to 29.0 ml
buffer containing O
2
. A nearly zero-order reaction
was observed with the rate of O
2
consumption of
6.87 mol/min and the rate of temperature rise of
0.01171 K/ min. The heat capacity of the calorim-
eter and contents was 254.6 J/ K. What is H for
the above reaction? NOTE: The H
+
is supplied by
the phosphate buffer, which has a H of dissocia-
tion of 5.4 kJ mol
1
.
12. Enthalpy and Gibbs energy changes for the
following reaction at 25C are given in Table 6-6.
ATP
4
(1 M) + H
2
O
ADP
3
(1 M) + H
+
(10
7
M) + HPO
4
2
(1 M)
a) How much heat is evolved at constant tem-
perature and pressure if the reaction takes
place in a test tube without doing any work
other than p V work?
b) How much heat is evolved or absorbed by the
foregoing reaction if it is coupled with 100%
efficiency to an endergonic reaction?
c) What efficiency of coupling to an endergonic
reaction is required in order that the foregoing
reaction neither evolve nor absorb heat?
13. Microorganisms use a great variety of fermenta-
tion reactions for obtaining energy. Could the
following reaction be used for such a purpose?
Explain the reasons for your answer.
C
4
H
6
O
4
(succinic acid) + H
2
O
C
3
H
8
O
3
(glycerol) + CO
2
14. a) Calculate the work done in kJ and in kcal by a
70 kg person in climbing up stairs three stories
(13 m).
b) Calculate how much ATP (in mmol and in
grams) will be needed for the climb if muscles
can use the ATP with 50% efficiency and if the
phosphorylation state R
p
= [ATP] / [ADP] [P
i
] is
10
3
M
1
. The standard value of G

(pH 7) for
hydrolysis of MgATP to MgADP and P
i
may be
taken as 31 kJ/ mol at 37C.
c) How much of this ATP could be provided by
transfer of phospho groups from the stored
creatine phosphate (Cr-P) to ADP in muscle?
Assume that [Cr-P] = 20 mM and may fall to
10 mM during the climb. G

(pH 7) for
hydrolysis of creatine phosphate is about
43 kJ/ mol. If the creatine kinase reaction
attains equilibrium what will the value of R
p
be for the adenylate system?
15. Acid base titration gave the following three pK
a
values for cysteine: 1.70, 8.36, and 10.53. Spectro-
photometric data allowed H. B. F. Dixon and K. F
Tipton (1973, Biochem J. 133, 837 842) to estimate
the following ratio of tautomeric species at pH 9.4
[

OOC C(NH
3
+
) CH
2
S

] / [

OOC C(NH
2
)
CH
2
SH] = 2.12, as is also shown in Fig. 6-5. Verify
and assign the four microscopic pK
a
values.
Study Questions
323
16. The coenzyme pyridoxal phosphate (PLP; structure
on p. 740) has pK
a
values of 3.62, 6.10, and 8.33,
as determined by acid base titration or by spec-
trophotometric titration. The pK
a
of 6.10 belongs
primarily to the phosphate group and is nearly
independent of the others. However, the pK
a
values of 3.62 and 8.33 are shared by the proto-
nated ring nitrogen and the phenolic OH group.
PLP exists as an equilibrium mixture of aldehyde
together with its covalent hydrate (see Eq. 13-1).
The equilibrium constants for hydrate formation
(K
h
= [hydrate] / [aldehyde]) are independent of
pH but differ for each ionization state of the ring.
Consider only the equilibria in the pH range 7 12.
The UV-visible absorption spectrum of the
monoprotonated form of PLP was divided math-
ematically into individual bands for the aldehyde
with dipolar ionic ring (), the aldehyde tautomer
with an uncharged ring (0) and the hydrate of the
dipolar ion. The following fractions were esti-
mated (Harris et al., 1976, Biochem. Biophys. Acta.
521, 181 194.
Aldehyde () 52%
Aldehyde (0) 28%
Hydrate () 16%
Hydrate (0) 4%, estimated indirectly
Study Questions
HPLP

(aldehyde, )
HPLP

(hydrate, )
K
H
HPLP

(hydrate, 0)
K
T(H)
HPLP

(aldehyde, 0)
K
T(A)
K
B
K
A
PLP
2
(aldehyde)
K
H

K
C
PLP
2
(hydrate)
K
D
From 300 MHz
1
H NMR spectra areas for the
following hydrogen atoms were estimated by
Robitaille.
pH 7 pH 12
4'-H aldehyde .86 .900
4'-H hydrate .14 .012
6-H, aldehyde .96 1.059
6-H, hydrate .17 .014
2'-CH
3
, aldehyde 2.49 2.96
2'-CH
3
, hydrate .51 .038
Evaluate the tautomerization constants K
T(A)
and
K
T(H)
, the hydration constants K
H
and K
H
, and the
microscopic pK
a
values pK
A
, pK
B
, pK
C
, and pK
D
in
the foregoing scheme. Assume that the hydration
ratios K
h
and K
h
are the same in H
2
O (spectropho-
tometric data) and in D
2
O (NMR data).
324 Chapter 7. How Macromolecules Associate
339
360
363
371
329
355
367
Boxes
Box 7-A Life and Death for Proteins:
Chaperonins and Proteasomes
Box 7-B Sickle Cell Disease, Malaria, and
Blood Substitutes
Box 7-C The T-Even Bacteriophages
Box 7-D Mitosis, Tetraploid Plants, and
Anticancer Drugs
Tables
Table 7-1 Binding Constants of Protons to Dianions
of Dicarboxylic Acids
Table 7-2 Thermodynamic Functions for
Oxygenation of Hemoglobin
Table 7-3 A Few Well-Known Structural Domains
325
325
327
328
329
330
332
332
332
334
334
334
335
336
337
337
338
342
344
345
348
349
349
350
350
351
352
352
353
353
353
355
357
358
359
362
362
362
367
368
369
369
370
373
377
A. Describing Binding Equilibria
1. Analyzing Data
2. Multiple Binding Sites on a Single Molecule
Microscopic binding constants and statistical effects
Electrostatic repulsion: anticooperativity
3. Cooperative Processes
B. Complementarity and the Packing of
Macromolecules
1. Rings and Helices
Molecules with cyclic symmetry
Helical structures
Filamentous bacteriophages
A rod-shaped plant virus
Bacterial pili
The thin filaments of muscle
2. Oligomers with Twofold (Dyad) Axes
Paired interactions
Dihedral symmetry
Oligomers with cubic symmetry (polyhedra)
Asymmetry and quasi-equivalence in oligomers
Quasi-equivalence in virus coats
Regulatory subunits and multienzyme complexes
C. Cooperative Changes in Conformation
1. Unequal Binding of Substrate and Induced Fit
2. Binding Equilibria for a Dimerizing Protein
The MonodWymanChangeux (MWC) model
The induced fit model
One conformational state dissociated
3. Higher Oligomers
D. The Oxygen-Carrying Proteins
1. Myoglobin and Hemoglobin
The binding of oxygen
Structural changes accompanying oxygen binding
The Bohr effect and allosteric regulators
Carbon monoxide, cyanide, and nitric oxide
2. Abnormal Human Hemoglobins
3. Comparative Biochemistry of Hemoglobin
E. Self-Assembly of Macromolecular Structures
1. Bacteriophages
2. Kringles and Other Recognition Domains and
Motifs
F. The Cytoskeleton
1. Intermediate Filaments
2. Microfilaments
3. Microtubules
References
Study Questions
Contents
Some ways in which protein subunits associate. (Left) The 3.66 MDa hemoglobin of the earthworm Lumbricus
terrestris contains 144 globin subunits organized as 12 cylindrical disulfide-linked dodecamers. Two 6-dodecamer
layers, each a ring with 6-fold cyclic symmetry, lie back-to-back. This reconstructed particle also contains three
types of linker proteins in the center region. From Lamy et al. (2000) J. Mol. Biol. 298, 638. (Center) The iron storage
protein ferritin is formed from 24 19- to 21-kDa 4-helix-bundle subunits with cubic symmetry. As many as 4500
atoms of iron, as hydrated iron oxide, may be stored in the internal cavity. See Fig. 7-13. From Trikha et al. (1995)
J. Mol. Biol. 248, 954. Courtesy of Elizabeth Theil. (Right) The 2 MDa molecular chaperone GroEL consists of two
back-to-back 7-subunit rings, each subunit formed from domains E, I and A. A 7-subunit cap of the smaller
GroES may cover either end to form a compartment in which polypeptides fold. See Box 7-A.
A
I
E E
I
A
E
I
A A
I
E
13.7 nm
GroEL Chaperonin
14.6 nm
4.5 nm
Hydrophobic binding
patches
Ferritin shell
Earthworm hemoglobin
325
How Macromolecules Associate
7
The complicated shapes and internal structures
of cells are determined to a large extent by the way in
which proteins and other macromolecules are bonded
one to another. In addition, intimate association of
macromolecules is essential to such biological processes
as the motion of flagella, the contraction of muscle, the
action of antibodies, the transmission of nerve impulses,
the replication of DNA, and the synthesis of proteins.
Equally important is the binding of small molecules to
large ones. In this chapter we will first examine methods
of measuring binding with an emphasis on protons
and small molecules. Then we will consider the ways
in which macromolecules stick together as well as the
role of conformational changes within macromolecules.
A. Describing Binding Equilibria
In previous discussions of pH we have dealt with
dissociation constants, but in this section we will use
formation constants K
f
, where K
f
= 1/K
d
. Measurement
of the strength of association of molecules is an every-
day aspect of modern biochemical research. It may be
important to know how strongly a hormone binds to
a receptor in a cell membrane or how well a feedback
inhibitor binds to an enzyme to determine whether the
interaction is significant physiologically. The binding
of O
2
to hemoglobin and other oxygen carriers is vitally
important, but the description of these oxygenation
reactions is mathematically complex. This is especially
so because we must consider effects of pH changes and
of changing concentrations of allosteric effectors on
the binding equilibria.
In considering such equilibria we must first exam-
ine the individual interactions of different domains of
a protein, one with another. These can be described by
association constants or, alternatively, by the Gibbs
energy changes for the association reaction.
1,2
The
average kinetic energy of motion of a molecule in
solution is about 3 / 2k
B
T, where k
B
T is Boltzmanns
constant. For one mole the kinetic energy is 3 / 2RT
or 3.7 kJ (0.89 kcal) mol
1
at 25C. Thus, if K
f
= 10 M
1
(G = 5.7 kJ mol
1
or 1.36 kcal mol
1
) the binding
energy is only slightly in excess of the thermal energy
of the molecules and the complex is weakly bound. In
this instance, if X and P are both present in 10
4
molar
concentrations (typical enough for biochemical systems),
only 0.1% of the molecules will exist as the complex
([complex] = K
f
[X][P]). If the formation constant is
higher by a factor of 1000, i.e., K
f
= 10
4
M
1
(G = 22.8
kJ mol
1
), 38% of the molecules will exist as the com-
plex; while if K
f
= 10
7
M
1
(extremely strong binding,
G = 40 kJ or 9.55 kcal mol
1
), 97% of the molecules
will be complexed.
1. Analyzing Data
The extent of binding of a molecule X to another
molecule P (Eqs. 7-1, 7-2) is measured by varying the
concentrations of X and P and observing changes
in the concentration of the complex [PX]. The first
K
f
= [ PX ] / [ P ] [ X ])
prerequisite is to find a measurable property that is
different for the complex than for either of the free
components. For example, the complex may be colored
and the components colorless. More commonly, the
complex simply has a different light absorbance (A) at
X + P PX
(7-2)
(7-1)
A
I
E E
I
A
E
I
A A
I
E
13.7 nm
GroEL Chaperonin
14.6 nm
4.5 nm
Hydrophobic binding
patches
Ferritin shell
Earthworm hemoglobin
326 Chapter 7. How Macromolecules Associate
a certain wavelength than do the components. Like-
wise, the circular dichroism or the chemical shift of a
peak in the NMR spectrum may change. If P is an
enzyme, only the complex PX will undergo decompo-
sition to products. Sometimes (but not always) the rate
of breakdown of PX (the enzymesubstrate complex)
to form products will be relatively slow compared to
the rate at which the equilibrium between X, P, and PX
is established. In this case the concentration of complex
PX will be proportional to the observed rate of forma-
tion of product.
Whatever change of property is measured, its
value will increase with increasing concentrations of
X if the total concentration of the macromolecule P is
kept constant. In the usual experimental design, the
molar concentration of P is small and it is possible to
increase the concentration of X to quite large values.
When this is done, it is usually observed that at high
enough values of [X] almost all of the P is converted
to PX, and the change being measured (e.g., A for
increased light absorption) no longer increases. This
effect is known as saturation and is observed in most
binding studies and also in many physiological phe-
nomena.
The property being measured (A) reaches a
maximum value A
max
at saturation and when all of
compound P has been converted to PX. The ratio of
[PX] to the total concentration of all forms of P present
[P]
t
is known as the saturation fraction and is often
given the symbol Y. If P has more than one binding
site for X, Y is defined as the fraction of the total bind-
ing sites occupied. If n is the number of sites per
molecule, the total number of sites is n[P]. The value
of Y is often taken as A/A
max
, an equality that holds
for multisite macromolecules only if the change in A is
the same for each successive molecule of X added. This
is not always true, but when it is Eq. 7-3 is followed.
Here i represents the number of ligands X bound to P
and may vary from 0 to n. When n = 1 the saturation
fraction Y and A are related to the concentration of
free unbound X and the formation constant as follows:
A plot of Y or A against [X] is shown in Fig. 7-1.
This kind of plot is sometimes called an adsorption
isotherm because it describes binding only at a constant
temperature. Notice, from both Fig. 7-1 and Eq. 7-4,
that Y reaches a value of 0.5 when [X] is just equal to
1/K
f
(or to K
d
). Note also that as [X] increases saturation
is reached slowly and that even at the point represent-
ing the highest concentration of X (8 /K
f
in Fig. 7-1)
saturation is less than 90%. Since in the usual experi-
mental situation, we do not know Y but only A, it is
difficult to estimate the limiting value A
max
from a
plot of this type unless K
f
is very high. However, we
need to know A
max
to evaluate K
f
. For this reason,
plots like that of Fig. 7-1 are seldom used, this one
being included mainly to illustrate a point of nomen-
clature. The curve shown in Fig. 7-1 is a rectangular
hyperbola, and the type of saturation curve shown is
frequently referred to as hyperbolic. This is in con-
trast to certain other binding curves (Section 3) which,
when plotted in this way, are sigmoidal (S-shaped).
A better type of plot is often that of Y against log
[X] (Fig. 7-2). It has the following features. (1) The
curve is symmetric about the midpoint at log [X] = log
K
f
. (2) No matter how high or low the concentration
range used in the experiments, it is easy to choose a
scale that puts all the points on the same sheet of
paper. (3) Spacing between points tends to be more
uniform than in a plot against [X]; e.g., compare Figs.
7-1 and 7-2 for which the experimental points repre-
sent the same data and for which values of [X] for
successive points are each twofold greater than the
preceding one. (4) The same logarithmic scale can be
used for all compounds, no matter how strong or weak
the binding, and the same shape curve is obtained for
all 1:1 complexes. The midpoint slope, dY/dlog [X],
is 0.576; the change in log [X] in going from 10 to 90%
saturation is 1.81. The curve is familiar to most chem-
ists because it is frequently used for pH titration curves
in which pH substitutes for log [X]. To represent a
complex with tighter binding, the curve is simply moved
to the left, and for weaker binding, it is moved to the
right.
i
i
i
n
A
A
= =
[ ]
[ ]
max
PX
P
t
Y

(7-3)
Y
f
X
f
X
f
X
f
X
=
+
=
+
K
K
A
A K
K
[ ]
[ ]
max
[ ]
[ ] 1 1


(7-4)
Figure 7-1 An adsorption isotherm, a plot of the saturation
fraction Y or of some change in a measured property A vs
[X], the concentration of a substance that binds reversibly to
a macromolecule. The curve is hyperbolic and [X] = 1/K
f
when Y = 0.5.
[x]
A
max
A
0.5
1.0
0
1
K
f
(in units of 1/K
f
)
2 0 4 6 8
Y
327
Figure 7-2 A saturation curve plotted on a logarithmic scale
for [X]. The data points are the same as those used in Fig. 7-1.
Tighter
binding
Looser
binding
A
max
A
0.5
1.0
log K
f
1
0
log K
f
log K
f
+ 1
log[X]
Y
A. Describing Binding Equilibria
Saturation data are often plotted in yet another form
known as the Scatchard plot (Fig. 7-3). The value of
A/ [X] (or of Y/ [X]) is plotted against A (or Y) and
a straight line is fitted to the points, preferably using
the method of least squares. The intercept on the
x axis and the slope of the fitted line give values of
A
max
/K
f
and K
f
, respectively, as indicated by Eq. 7-5,
which follows directly from Eq. 7-4.
Y/ [X] = K
f
YK
f
A/ [ X ] = A
max
K
f
AK
f
The Scatchard plot is the best of the various linear
transformations of the saturation equation and is pre-
ferred to double reciprocal plots analogous to that
shown in Fig. 9-3.
Scatchards original equation was formulated to
(7-5)
deal with the binding of two or more ligands to a
single macromolecule.
3 5
If we let [X]
b
represent the
concentration of bound X and [P]
t
the total molar
concentration of the protein or other macromolecule
and if there is only one binding site on the protein,
[X]
b
/ [P]
t
will equal Y. However, if there are n inde-
pendent binding sites that have the same binding
constant K
f
Eq. 7-5a will hold.
If [X]
b
/ [P]
t
[X] is plotted against [X]
b
/ [P]
t
the resulting
linear plot will have an intercept of K
f
on the y axis and
n on the x axis. Thus, n is directly apparent, whereas
in Eq. 7-5 it is incorporated into Y. A problem arises if,
as discussed in the next section, the multiple binding
sites are not independent but interact. Curved Scat-
chard plots result and attempts to extract more than
one binding constant can lead to very large errors.
Before measuring saturation curves, the student should
read additional articles or books on the subject.
2,610
2. Multiple Binding Sites on a Single Molecule
A macromolecule may often be able to bind sever-
al molecules of a second compound X. Consider the
case in which the macromolecule P binds successively
one molecule of X, then a second, and a third, up to a
total of n. We define the stepwise formation constants,
K
1
, K
2
, . . ., K
n
, as follows:
Remember that these are reversible reactions even
though unidirectional arrows are used. The general
expression for the ith stepwise formation constant is
given by Eq. 7-7.
Remember that Y is the fraction of total binding sites
saturated. The number of moles of X bound per mole
of P is nY and is obtained by summing the concentra-
(7-5a)
P + X PX
K
1
PX + X PX
2
. . .
K
2
PX
n-1
+ X PX
n
K
n
(7-6)
K
f
A
max
0.5 1.0 0
A
[x] [x]
K
f
K
f
2
A
max
A
0
Y
Y
Figure 7-3 A Scatchard plot of the same data shown in Figs.
7-1 and 7-2. This is the best of the linear plots for studying
binding.
[ ]
[ ]
[ ]
[ ]
.
X
b
P [X]
X
b
P
f f
t
n
t
K K
1
=
K
i
i
i
=

[ ]
[ ][ ]
PX
PX X
1
(7-7)
328 Chapter 7. How Macromolecules Associate
P
COO
COO

P
COOH
COOH
P
COOH
COO

P
COO
COOH

(A)
(B)
K* K*
K* K*
P PH PH
2
(7-14)
tions [PX] + 2[PX
2
] + . . . and dividing the sum of all
the forms of P:
The summations are over all of the integral values
of i from 1 to n. Now, by expressing each concentration,
[PX
i
], in terms of the concentrations [X] and [P] of free
X and P, together with the stepwise formation constants,
we obtain Eq. 7-10.
A similar equation can be written for the general
case. Note that the concentration of P does not appear
in Eq. 7-10 and that Y is a function only of [X] and the
stepwise formation constants. Such equations define
the isotherms for binding of two or more molecules of
X to P. From an experimental plot of Y (or of A) vs
[X] or log [X], it is possible in favorable cases to deter-
mine the stepwise constants K
1
, K
2
, ..., K
n
. However,
this becomes quite complicated. To simplify Eq. 7-10
and the corresponding equation for the general case,
we can group the constants together and designate the
products of constants (K
1
, K
1
K
2
, K
1
K
2
K
3
, etc.) as
1
,
2
, .
. .,
n
. Our equations are now as follows:
From experimental data, it is usually easiest to first
determine the s (there are n of them), and then to
calculate from the s the stepwise constants. For
example:
For two binding sites
Y
PX PX
P PX PX
For the general case
Y PX P PX
( )
[ ] [ ]
[ ] [ ] [ ]
[ ] [ ] [ ]
n
n i
i
n
i
i
n
i
=
=
+
+ +
=
=
+
=

2
2
2
2
2
1 1
(7-9)
(7-8)
For
Y
X X
X X
For the general case
Y X X
n
n i
i
n
i
i
i
i
n
i
=
=
+
+ +
=
=
+
=

2
2
1
2
2
2
1
1 2
2
1
1
1



[ ] [ ]
[ ] [ ]
[ ] [ ]
(7-11)
(7-12)
While Eq. 7-12, known as the Adair equation,
11
might
seem to provide a complete description of the binding
process, it usually does not. In many cases, there is
more than one kind of binding site on a macromolecule
and Eq. 7-12 tells us nothing about the distribution of
the ligand X among different sites in complex PX. To
consider this problem we must examine the microscopic
binding constants.
Microscopic binding constants and statistical
effects. As discussed in Chapter 6, Section E,2, micro-
scopic binding constants represent the constants for
binding to specific individual sites. Now, consider a
straight-chain dicarboxylic acid which has two identical
binding sites for protons. If the chain connecting the
two carboxylate anions is long enough, the carboxylate
groups will be far enough apart that they do not influ-
ence each other through electrostatic interaction.
Each group will have a microscopic binding constant
(K
f
*) of 5 x 10
4
M
1
. The constant K
f
* can also be called
an intrinsic binding constant, because it is character-
istic of a carboxylate group that is free of interactions
with other groups. Intuition tells us (correctly) that,
in its binding of protons, a solution of this dicarboxy-
lic acid dianion will behave exactly like a solution of
the monovalent anion R COO

at twice the concentra-


tion. A single intrinsic binding constant suffices to
describe both binding sites. It may seem surprising
then that the stepwise formation constants (also called
stoichiometric or macroscopic formation constants)
K
1
and K
2
differ: K
1
= 10 x 10
4
M
1
and K
2
= 2.5 x 10
4
M
1
. This fact reflects the so-called statistical effect.
Either of the two carboxylate groups in the molecule
can bind a proton in the first step to give two indistin-
guishable molecules, PH:
K K K
1 1 2
2 1
= = , etc.
(7-13)
OOC

COO

K
f
* = 5 10
4
K
f
* = 5 10
4
Y
X X
X X
K K K
K K K
=
+
+ +
2
1
2
1 2
2
1
1 1 2
2
[ ] [ ]
[ ] [ ]
(7-10)
For n = 2
329 A. Describing Binding Equilibria
If we label the two forms of PH as A and B (Eq. 7-14)
and consider that each one of them is independently in
equilibrium with P through formation constant K
f
*, we
obtain Eq. 7-15 (which may be compared with Eqs. 6-75
and 6-76, which are written for dissociation constants).
This result is related to probability and arises for the
same reason that if you reach into a barrel containing
50% white balls and 50% black balls, you will pull out
one of each just twice as often as you will pull out a
pair of white or a pair of black. In the general case of n
equivalent binding sites, the microscopic formation
constants K
i
* are related
12,13
to the stepwise constants
K
i
as follows:
It is also easy to show,
14
using Eqs. 7-12 and 7-16 that
for n completely equivalent and independent binding sites
Eqs. 7-17 and 7-18 hold:
In this case the microscopic association constants are
all identical and represent a single intrinsic constant
applicable to all of the sites. In fact, Eq. 7-18 is identical
to that for association of a single proton (or other ligand)
with a single binding site, satisfying our intuitive
notion that a set of n completely independent binding
sites should behave just like a solution of an n-fold
more concentrated compound with a single binding
site. Thus, our arithmetic has led us to a conclusion
that was already obvious. However, it is rarely true
that binding sites on a single macromolecule are com-
pletely independent; there is almost always interaction
between them, and the equations that we have derived
for evaluation of stepwise and intrinsic constants
cannot be applied without modification.
Electrostatic repulsion: anticooperativity.
As we have seen, a hypothetical acid with an infinite
distance between the carboxylate groups and log K
f
* =
4.8 would have two macroscopic binding constants
K K K K
1
2
2
2 =
+
+
= =
[ ] [ ]
[ ][ ]
* *
PH
A
PH
B
P H
and
(7-15)
K
i
n i
i
K
i
=
+ ( )
*
1
(7-16)
Y
X X
X
or
Y
X
X
=
+

+
=
+
K K
n
K
n
K
K
*[ ]( *[ ])
( *[ ])
*[ ]
*[ ]
1
1
1
1 (7-18)
(7-17)
separated by the statistical distance (log 4 = 0.6).
Compare these values with the observed binding
constants for protons with the dianions of acids con-
taining 7, 4, 2, and 1 CH
2
groups given in Table 7-1.
For the longest chain (that of azelaic acid) the log K
f
values are not very different from those of the hypo-
thetical long-chain acid. However, as the groups come
closer together, the first binding constant is increased
markedly because of the additional electrostatic attrac-
tion and the second is decreased. The spread between
the two log K
f
values increases from 0.6 to as much as
2.9 as a result of interaction between the binding sites.
In malonic and succinic acids the first proton
bound can be shared by both carboxyl groups through
formation of a hydrogen bond. (See discussion in
Chapter 9, Section D.) Additional factors operate in
oxalic acid where the carboxyl groups are connected
directly and for which pK
a
values (pK
a
= log K
f
) are
4.19 and 1.23. In all of these examples the binding of
the first proton makes it harder to bind a second proton.
Such negative interaction or anticooperativity between
binding sites is very common and always leads to a
spread of the formation constants and a broadening of
the curve of Y vs log [X]. This is shown graphically on
the right side of Fig. 7-4 where the binding curves for
protons with acetate ion and with succinic acid dianion
are compared. Notice that binding of protons increases
as log [H
+
] increases, giving the curves an unfamiliar
appearance when compared with the more familiar
curve of dissociation vs pH.
Can we predict the pK
a
values in Table 7-1? With
an appropriate dielectric constant chosen Eq. 2-8 can
be applied. The difference between the two successive
log K
f
values reduced by 0.6 (the statistical factor) is a
TABLE 7-1
Binding Constants of Protons to Dianions of
Dicarboxylic Acids
a
Hypothetical 5.1 4.5
dianion with
log K* = 4.8
Azelaic 7 5.41 4.55
Adipic 4 5.41 4.42
Succinic 2 5.48 4.19
Malonic 1 5.69 2.83
a
From R. P. Bell, (1973) The Proton in Chemistry, 2nd ed., p. 96.
Cornell Univ. Press, Ithaca, New York
Acid dianion
No. of
CH
2
groups
log K
1
(pK
2
)
log K
2
(association)
(pK
1
) (dissociation)
330 Chapter 7. How Macromolecules Associate
measure of the electrostatic effect. For malonic acid
this pK
a
is 2.25 and for succinic acid it is 0.69 (Table
7-1). In 1923, N. Bjerrum proposed that the value of
pK
a
could be equated directly with the work needed
to bring the two negative charges together to a distance
representing the charge separation in the malonate
dianion.
Thus, applying Eq. 6-31, G = 5.708 pK
a
kJ mol
1
=
12.84 kJ mol
1
for malonate. Equating this with W
in Eq. 2-8 and assuming a dielectric constant of 78.5
(that of water), the distance of charge separation r is
calculated to be 0.138 nm. This is much too small.
The computation was improved by Westheimer and
Kirkwood, who assumed a dielectric constant of 2.0
within the molecule. By approximating the molecule
as an ellipsoid of revolution, they were able to make
reasonably accurate calculations of electrostatic effects
on pK
a
values.
15
Thus, for malonic acid Westheimer
and Shookhoff
16
predicted r = 0.41 nm for malonic acid
dianion. Recently more sophisticated calculations
17
have been used to predict pK
a
values for the com-
pounds in Table 7-1 and others.
18
Electrostatic theory has also been used successfully
to interpret titration curves of proteins in which the
net negative or positive charge distributed over the
surface of the protein varies continuously from high
pH to low as more protons are added.
19
Electrostatic effects can be transmitted extremely
effectively through aromatic ring systems, a fact that
explains some of the significance of heterocyclic aro-
matic systems in biochemical molecules. Consider the
Figure 7-4 Binding of protons to the
thiamin anion, the succinate dianion,
and the acetate anion. Acetate (dashed
line) binds a single proton with a normal
width binding curve. Succinate dianion
binds two protons with anticooperativity,
hence a broadening of the curve. The
thiamin anion (yellow form, see Eq. 7-19)
binds two protons with complete coop-
erativity and a steep binding curve.
log[H
+
] or pH
Thiamin
anion
12 11 10 13
pK = 11.6
1.0
0.5
0.0
Succinate
dianon
pK
1
= 4.19
Acetate

,
pK
a
= 4.76
pK
2
= 5.48
7 6 5 4 8 3 2
Y
microscopic binding constant of the phenolate anion of
pyridoxine as influenced by the state of protonation of
the ring nitrogen. These are shown in Eq. 6-75, where
pK
a
* = 4.94 and pK
d
* = 8.20 define the binding constants
for protonation of a phenolate ion when the ring nitrogen
is protonated or unprotonated, respectively. We see
that pK
a
= 3.26, even greater than that of malonic acid.
3. Cooperative Processes
Can it ever happen that interaction between
groups leads to a decrease from the statistical separa-
tion between values of the stepwise constants instead
of to an increase? At first glance, the answer seems to
be no. A decreased separation would imply that the
intrinsic binding constant for the second proton bound
is higher than that for the first, but common sense tells
us that the first proton ought to bind at the site with
the highest binding constant. However, look at the
experimental binding curve of protons with the anion
of thiamin shown in Fig. 7-4. Instead of being broad-
ened from the curve of acetate, it is just half as wide.
The explanation depends upon some rather amusing
chemistry of thiamine. Under suitable conditions, this
vitamin can be crystallized as a yellow sodium salt,
the structure of whose anion is shown in Eq. 7-19.
Weak binding of a proton to one of the nitrogens as
shown in Eq. 7-19 creates an electron deficiency at
the adjacent carbon and the S

anion adds to the C=N


group, closing the ring to an unstable tricyclic form of
thiamin. This tricyclic form can be observed in methanol
and can be crystallized. It is unstable in water because
the central ring can open, with the electrons flowing as
indicated by the small arrows to create a strongly basic
site on the same nitrogen. A second proton combines
at this basic nitrogen with a high binding constant to
form a cation.
The key to the reversed order of strength of the binding
C
H H
C C
O O
O O

r 0.4 nm
331
constants lies in the molecular rearrangements intervening
between the two binding steps.
20,21
In this particular case,
we cannot measure the successive binding constants
K
1
and K
2
directly from the titration curve because
K
2
is almost two orders of magnitude larger than K
1
.
Consequently, the binding curve shown in Fig. 7-4 is
(within the experimental error) twice as steep at the
center (the slope is 2 x 0.576) as that for acetate ion and
is accurately represented in Eq. 7-20. Comparison of
Eq. 7-20 with Eq. 7-10 shows how the latter has been
simplified because no significant concentration of the
form PX is present in the cooperative case.
This is only part of the story about the acidbase
chemistry of thiamin. For the rest, see Chapter 14,
Section D,1.
The binding of protons by the thiamin anion is an
example of a cooperative process, so named because
binding of the first proton makes binding of the second
easier. Although relatively rare among small molecules,
cooperative processes are very common and important
in biochemistry.
22,23
A cooperative binding curve is
sometimes referred to as sigmoidal because the plot
of Y against [X] (the binding isotherm) is S shaped.
The maximum possible cooperativity is observed
A. Describing Binding Equilibria
when the binding of the first ligand enhances the
affinity of all other sites so much that no species other
than P and PX
n
are present in significant concentration.
It is easy to show that, for n binding sites with such
completely cooperative binding, the saturation fraction is:
where K = (K
1
. . . K
n
)
1/ n
. The midpoint slope in the
binding curve (Y vs log [X]) is 0.576n and the change,
log [X], between Y = 0.1 and Y = 0.9 is 1.81/ n.
Equation 7-21 can be rewritten as
Taking logarithms (Eq. 7-22)
A plot of log [Y/ (1 Y)] vs log [X] is known as a Hill
plot. According to Eq. 7-22, it is linear with a slope of n.
Remember that this equation was derived for an ideal
case of completely cooperative binding at n sites.
24
However, Hill plots are often used to plot experimen-
tal data for systems in which cooperativity is incom-
plete. Thus, the experimentally measured
slope of a Hill plot (n
Hill
) is not an integer and
is usually less than n, the number of binding
sites. A comparison of n
Hill
with n is often
used as a measure of the degree of cooperativ-
ity: n
Hill
/n = 1.00 for complete cooperativity
but is less than one if cooperativity, is incom-
plete. An example of a very high degree of
cooperativity is provided by the hexameric
enzyme glutamate dehydrogenase, whose
saturation curve for substrate displays n
Hill
approaching six.
25
It is not necessary to make
a Hill plot to get n
Hill
. From the usual binding
curve of Y (or A) vs log [X] the midpoint
slope can be measured with satisfactory preci-
sion. Alternatively, the difference, log [X],
between 0.1 and 0.9 saturation can be evaluat-
ed and n
Hill
calculated from Eq. 7-24.
Binding curves sometimes show more than
one step; in such cases Hill plots are not linear
and no simple measure of cooperativity can be
defined.
(7-19)
Y
X
X
=
+
K
K
2 2
1
2 2
[ ]
[ ] (7-20)
Y
X
X
=
+
K
n n
K
n n
[ ]
[ ] 1 (7-21)
Y Y X ( ) [ ] 1 = K
n n
(7-22)
log [ ( )] log log [ ] Y Y X 1 = + n K n
(7-23)
N
N
N N
S
H
3
C
CH
3
CH
2
CH
2
OH
N
N
N N
S
CH
3
H
3
C
OH
N
N
N N
S
CH
3
H
3
C
OH
H
H
N
N
N H
2
N
S
CH
3
H

+
H
3
C
OH
H
Yellow anion of
thiamin (vitamin B
1
)
+
Cation
(colorless)
+
weak
binding
H
+
strong
binding
Tricylic
unstable form
A strongly basic
site is created here
by ring opening
(7-24)
n
Hill
=
midpoint slope
0.576
1.81
log[X]
=
332 Chapter 7. How Macromolecules Associate
A second example of cooperativity is provided by
the reversible denaturation of coiled peptide chains.
Some proteins can be brought to a pH of 4 by addition
of acid but without protonation of buried groups with
intrinsic pK
a
values greater than four. When a little
more acid is added, some less basic group is protonated,
permitting the protein to unfold and to expose the more
basic hidden groups. Thus, cooperative proton binding
is observed. As in the case of thiamin the cooperativity
depends upon the occurrence of a conformational
change in the molecule linked to protonation of a
particular group.
The reversible transformation between an helix
and a random coil conformation is also cooperative.
In this case, once a helix is started, additional turns
form rapidly and the molecule is completely converted
into the helix. Likewise, once it unfolds it tends to
unfold completely. Melting of DNA (Chapter 5) or,
indeed, of any crystal is cooperative.
8
The stacking of
nucleotides alongside a template polynucleotide can
also be cooperative. For example, the binding of an
adenylate residue to two strands of polyuridylic acid
leads to cooperative formation of a triple-helical
complex (Chapter 5, Section C,4). Here the stacking
interactions make helix growth energetically easier
than initiation of new helical regions.
26
B. Complementarity and the Packing of
Macromolecules
Because the forces acting between them are weak,
two molecules will cling together tightly only if there
is a close fit between their surfaces. For a firm bond to
be formed many atoms must be in contact and the two
molecular surfaces must be complementary one to the
other. If a knob, such as a CH
3
group, is present on
one surface, there must be an appropriate hollow in
the complementary surface. A positive charge in one
surface is likely to be opposite a negative charge in the
other. A proton donor group can form a hydrogen
bond only if it is opposite a group with unshared
electrons; nonpolar (hydrophobic) groups must be
opposite each other if hydrophobic interaction is to
occur. An important principle is that two molecules with
complementary surfaces tend to join together and interact,
whereas molecules without complementary surfaces do not
interact. Watson called this selective stickiness.
27
Selective stickiness permits the self-assembly of bio-
logical macromolecules having surfaces of comple-
mentary shape into fibers, tubes, membranes, and
polyhedra. It also provides the means for specific
pairing of purine and pyrimidine bases during the
replication of DNA and during the synthesis of RNA
and of proteins.
Complementarity of surfaces is equally important
to the chemical reactions of cells. Each of these reactions
is catalyzed by an enzyme, which contains reactive
chemical groupings in the right places and in the right
orientations to interact with and promote a chemical
change in another molecule, the substrate. Specific
catalysis is one of the most basic characteristics of
living things. Enzymatic catalysis provides the basis
not only for the reactions of metabolism but also for
the movement of muscle fibers, the flowing of the cyto-
plasm in the ameba, and virtually all other biological
responses. To understand these phenomena requires
an examination of the structures of the macromole-
cules involved and of the ways in which they can fit
together.
Just as the amino acids, sugars, and nucleotides
are the building blocks for formation of proteins,
polysaccharides, and nucleic acids, these three kinds
of macromolecule are the units from which larger
subcellular structures are assembled. Fibers, micro-
tubules, virus coats, and small symmetric groups of
subunits in oligomeric proteins all result from the
packing of macromolecules in well-defined ways,
something that is often called quaternary structure.
1. Rings and Helices
Consider first the aggregation of identical protein
subunits. While many protein molecules are nearly
spherical, they are nevertheless asymmetric. In the
drawings that follow the asymmetry is exaggerated,
but the principles illustrated are valid. One easily
observed lesson from nature is that even though living
things are made up of asymmetric materials, a great
deal of symmetry is evident.
28
At the molecular level
the symmetry of crystalline arrays of atoms or mole-
cules is described mathematically by the elements of
symmetry present in space groups (p. 133). There
are 230 of these but only 65 accommodate asymmetric
objects (Chapter 3).
29
Two of the natural ways for
identical asymmetric subunits to interact lead to rings
and helices, respectively.
Molecules with cyclic symmetry. Consider a
subunit (protomer) of the shape shown in Eq. 7-25
and containing a region a that is complementary to the
surface j on another part of the same molecule. Two
such protomers will tend to stick together to form a
dimer, region a of one protomer sticking to region j of
the other. The dimer will still contain a free region a
at one end and a region j at the other which are not
involved in bonding. Other protomers can stick to
these free ends. In some instances long chains can be
formed. However, if the geometry is just right, a third
subunit can fit in to form a closed ring (a trimer).
Depending on the geometry of the subunits, the ring
can be even smaller (a dimer) or it can be larger (a
tetramer, pentamer, etc.). The bonding involved is
333
between two different regions (a and j) of a subunit
and is sometimes described as heterologous.
30
To
obtain a closed ring of subunits, the angle between
the bonding groups a and j must be correct or the ring
cannot be completed.
A ring formed using exclusively heterologous
interactions possesses cyclic symmetry. The trimer
in Eq. 7-25 has a threefold axis: Each subunit can be
superimposed on the next by rotation through 360/ 3.
The oligomer is said to have C
3
symmetry. Many real
proteins, including all of those with 3, 5, or another
uneven number of identical protomers, appear to be
formed of subunits arranged with cyclic symmetry.
An example is the cholera toxin from Vibrio cholerae,
which forms a pentamer with an outer ring of subunits
with C
5
symmetry (Fig. 7-5).
Now consider the quantitative aspects of heterolo-
gous interactions with ring formation. Let K
f
be the
formation constant and G the Gibbs energy change
for the reaction of the j end of protomer P with the a end
of a second protomer to form the dimer P
2
(Eq. 7-25).
In the second step (Eq. 7-25) a third protomer
combines. It forms two newaj interactions. If we assume
for this step that G
f
is 2 G, K
f
will be equal to K
2
.
The overall association constant for formation of a
trimer from three protomers will be given by Eq. 7-26.
3 P P
3
K
f
= K
3
G
f
= 3 G
This will be true only if G for formation of both new
aj bonds in the trimers is exactly the same as that for
formation of the aj bond in the dimer. The reader may
wish to criticize this assumption
30a,b,c
and to suggest
conditions that might lead to overestimation or under-
estimation of K
f
for the trimer as calculated previously.
Now consider a hypothetical example: Protomer P
is continuously synthesized by a cell. At the same time
some subunits are degraded to a nonaggregating form
via a second metabolic reaction. The two reactions are
balanced so that [P] is always present at a steady state
B. Complementarity and the Packing of Macromolecules
value of 10
5
M. Suppose that a value for a single aj
interaction of K = 10
4
(and G = 22.8 kJ mol
1
) governs
aggregation to form dimers and trimeric rings. What
concentration of dimers and of trimer rings will be
present in the cell in equilibrium with the 10
5
concen-
tration of P. Using Eq. 7-25 we see that the concentra-
tion of dimers [P
2
] is 10
4
x (10
5
)
2
= 10
6
M. (Note that
the amount of material in this concentration of dimer
is equivalent to 2 x 10
6
M of the monomer units.) The
concentration of rings [P
3
] is (10
4
)
3
x (10
5
)
3
= 10
3
M,
equivalent to 3 x 10
3
M of the monomer units. Thus,
of the total P present in the cell (10
5
+ 0.2 x 10
5
+ 300 x
10
5
M), 99.6% is associated with trimers, 0.33% is still
monomers, and only 0.07% exists as dimers. Thus, the
formation of two heterologous bonds simultaneously to
complete a ring imparts a high degree of cooperativity
to the association reaction of Eq. 7-25. We will find in
a cell mostly either rings or monomer but little dimer.
Figure 7-5 Stereoscopic view
of the B
5
pentamer of cholera
toxin B. The pentamer, known
as choleragenoid, has a central
hole of ~ 1.5 nm diameter into
which a helix from the A subunit
is inserted. As viewed here, the
front surface of the pentamer has
binding sites for the oligosaccha-
ride chains of ganglioside GM
1
,
which serves as the toxin receptor.
The back side binds the A sub-
unit. See also Box 11-A. From
Zhang et al.
31
a
j
P
Step 1
K
f
= K G
f
= G
a
j
a
j
Step 2
K
f
K
2
G
f
2G
P
2
a
j
a
j
a
j
P
3
(7-25)
(7-26)
334 Chapter 7. How Macromolecules Associate
Now consider what will happen to the little rings
within the cell if the process that removes P to a non-
associating form suddenly becomes more active so that
[P] falls to 10
6
M. If K is still 10
4
, what will be the
percentages of P, P
2
, and P
3
at equilibrium? Here we
note a characteristic of cooperative processes: A higher
than first power dependence on a concentration.
Helical structures. If the angle at the interface aj
is slightly different, instead of a closed ring, we obtain
a helix as shown in Fig. 7-6A. The helix may have an
integral number of subunits per turn or it may have a
nonintegral number, as in the figure. The same type of
heterologous interaction aj is involved in joining each
subunit to the preceding one, but in addition other
interactions occur. If the surfaces involved in these
additional interactions are complementary and the
geometry is correct, groups from two different parts
of the molecule (e.g., b and k) may fit together to form
another heterologous bond. Still a third heterologous
interaction cl may be formed between two other parts
of the subunit surfaces. If interactions aj, bk, and cl are
strong (i.e., if the surfaces are highly complementary
over large areas), extremely strong microtubular struc-
tures may be formed, such as those in the flagella of
eukaryotic organisms (Fig. 1-8). If the interactions are
weaker, labile microfilaments and microtubules, such
as are often observed to form and dissociate within
cells, may arise.
The geometry of subunits within a helix is often
advantageously displayed by imagining that the sur-
face of the structure can be unfolded to give a radial
projection (Fig. 7-6B). Here subunits corresponding to
those in the helix in Fig. 7-6A are laid out on a plane
obtained by slitting the cylinder representing the sur-
face of the helix and laying it out flat. In the example
shown, the number of subunits per turn is about 4.8
but it can be an integral number. The interactions bk
between subunits along the direction of the fiber axis
may sometimes be stronger than those (aj) between
adjacent subunits around the spiral. In such cases
the microtubule becomes frayed at the ends through
breaking of the aj interactions. This phenomenon can
be observed under the electron microscope for the
microtubules from flagella of eukaryotic organisms.
Figures 7-7 to 7-10 show four helical structures from the
molecular domain. They are a filamentous bacterio-
phage, a plant virus, a bacterial pilus, and an actin
microfibril. Each is composed largely of a single kind
of protomer. A larger and more complex helical struc-
ture, the microtubule, is shown in Fig. 7-34.
Filamentous bacteriophages. Bacteriophages of
the Ff family include the fd, f1, and M13 strains.
31a,32 36
Phage M13 is widely used in cloning genes and for
many other purposes (Chapter 26). The genome is a
circular, single-stranded DNA of ~ 6400 nucleotides
which is held in an elongated double-stranded form
by a helical sheath of about 2700 subunits of a 50-
residue protein. The rod is about 6 nm in diameter
and 880 nm long and it is capped by two specialized
proteins at one end and a different pair of proteins at
the other end. The five coat proteins are encoded by
five of the 11 genes present in these little viruses.
37,38
Each coat subunit in the Ff viruses is coiled into an
-helical rod of 7 nm length. These are arranged in the
virus in a right-handed helical pattern with a pitch of
1.5 nm and with 4.4 subunits per turn (Fig. 7-7). The
protein rods are inclined to the helix axis and extend
inward. This arrangement permits a knobs-in-holes
hydrophobic bonding between subunits. The helix of
pitch 1.5 nm is the primary or one-start helix. How-
ever, in every regular helical structure we can also
trace a two-start helix, a three-start helix, etc. In this
instance the five-start helix is easiest to see.
The protein coat of these viruses provides an
elongated cylindrical cavity to protect the circular,
single-stranded DNA molecule that is the genome.
Although there are two antiparallel strands of DNA,
a regular base-paired structure is impossible and the
DNA is probably not present in a highly ordered
form.
38a
There are about 2.4 nucleotides in the DNA
per protein subunit. However, there are related viruses
with ratios as low as one nucleotide per protein sub-
unit and containing more highly extended DNA.
34,39
A rod-shaped plant virus. The tobacco mosaic
virus (Figs. 5-41, 7-8) is a 300-nm-long rod constructed
from 2140 identical wedge-shaped subunits whose
detailed molecular structure is known.
40
Each 158-
residue subunit contains five helices and a small
sheet. A single strand of RNA containing 6395 nucle-
(A)
c
k
a
a
j
j
b
pitch
l
(B)
k
b
c
a
l j
j
j
j
j
a
a
a
Figure 7-6 (A) Heterologous bonding of subunits to form a
helix. (B) Radial projection of subunits arranged as in helix
A. Different bonding regions of the subunit are designated
a, b, c, j, k, and l.
335 B. Complementarity and the Packing of Macromolecules
Figure 7-7 Structure of the virus fd
protein sheath. (A) Left. A single coat
subunit, with its N terminus towards
the top, as if moved from the left side of
the sheath. The dark circles represent
charged atoms of Asp, Glu, and Lys side
chains. The backbone of the protein is
a C

diagram. The positively charged


atoms near the C terminus line the inner
surface of the sheath neutralizing the
negative charge of the DNA core. Right.
Each subunit is represented by a helical
tube through successive C

atoms.
Three nearest neighbors, indexed as 0, 6, and 11, are indicated. The axial slab shown represents ~1% of the total length of the
virion. From Marvin.
31a
(B) A 2.0 nm section through the virus coat with the helices shown as curved cylinders. The view is
down the axis from the N-terminal ends of the rods. The rods extend upward and outward. The rods with indices 0 to 4
start at the same level, forming a five-start helical array. The rods with more negative indices start at lower levels and are
therefore further out when they are cut in this section. (C) The same view but with wire models of the atomic structure of
the rods. From Marvin et al.
32
otides (~ 3 per protein subunit) lies
coiled in a groove where it interacts
with side chains from two of the heli-
ces (Fig. 7-8B).
4144a
The virus is as-
sembled by the binding of a region of
the RNA 800 1000 nucleotides from
the 3' end to a two-turn helix of sub-
units that appears to form spontane-
ously. Additional subunits then add at
each end, binding to the RNA as well
as the adjacent protein subunits.
42,44,45
A relative with a very similar struc-
ture is cucumber green mottle mosaic
virus.
46
Bacterial pili. The adhesion pili,
or fimbriae,
48
of bacteria are also helical
arrays of subunits. The P pili of E. coli
are encoded by a cluster of 11 genes in
the pap (pilus associated with pyelo-
nephritis) cluster. They are needed
to allow the bacteria to colonize the
human urinary tract. The bulk of the
~1-m-long pilus is made up of about
1000 subunits of a 185-residue protein
encoded by gene PapA. They form a
right-handed helix of ~ 7 nm diameter
with 3.28 subunits per turn and a
pitch of ~ 2.5 nm (Fig. 7-9A).
49 51
The
rod is anchored to the bacterial outer
membrane by a protein encoded by
gene PapH, while subunits encoded
by PapE and PapF fasten the adhesin
protein (PapG) to the tips of the
pili.
49,52,53,53a
The adhesin binds to the
Gal1 4Gal ends of glycolipids in
B
C
A
336 Chapter 7. How Macromolecules Associate
the kidney.
51,54
The PapE, F, and
G subunits form a thin ~ 2.5-nm-
thick by 15 nm fibrillum which
is attached by an adapter protein
encoded by gene PapK. A special
chaperonin (PapD gene) is also
required for pilus assembly in E.
coli
53a
as well as other bacteria.
55
Another E. coli pilus adheres to
mannose oligosaccharides.
53b
Similar pili of Neisseria gonno-
rheae are used by that bacterium.
The three-dimensional structure of
the 158-residue pilin subunit is that
of a globular subunit with an 8.5 nm
-helical spine at one end.
56,57
A
proposed model of the intact pilus
shaft is shown in Fig. 7-9B,C. Notice
the similarity of the packing of the
-helical spines in the center to the
packing arrangement in the bacterio-
phage coat in Fig. 7-7. Similar features
may be present in the P pilus rod
shown in Fig. 7-9A. However, there
is uncertainty about the packing
arrangement. The E. coli type 1 pilus
subunits contain immunoglobulin
folds that are completed by donation
of an N-terminal strand from a
neighboring subunit.
53a
In thin
fimbriae of Salmonella extended,
parallel helices may be formed
(see Fig. 2-17)
53e,53f
Other types of pili are also well-
known.
53c,d
F pili or conjugative pili
are essential for sexual transfer of
DNA between bacterial cells (Chapter
26). F
+
strains of E. coli form hollow
pili of 8.5 nm diameter with a 2.0-nm
central hole.
58,59
Their 90-residue
subunits apparently form rotationally
symmetric pentamers which stack to
form the pili.
59
These pili are essen-
tial to establishing the initial contact
between conjugating bacterial cells.
The thin filaments of muscle.
An essential component of skeletal
muscle (discussed further in Chapter
19) is filamentous actin (F-actin). It
is composed of 375-residue globular
subunits of a single type and with a
highly conserved sequence.
60,61
It is
found not only in muscle but also in
other cells where it is a component
of the cytoskeleton. The actin micro-
filament has the geometry of a left-
C
B
C
A
Figure 7-8 (A) Electron micrograph of the rod-shaped particles of tobacco
mosaic virus. Omikron, Photo Researchers. See also Butler and Klug.
42
(B) A stereoscopic computer graphics image of a segment of the 300 nm long
tobacco mosaic virus. The diameter of the rod is 18 nm, the pitch of the helix is
2.3 nm, and there are 16 1\ 3 subunits per turn. The coat is formed from ~ 2140
identical 17.5-kDa subunits. The 6395-nucleotide genomic RNA is represented
by the dark chain exposed at the top of the segment. The resolution is 0.4 nm.
From Namba, Caspar, and Stubbs.
47
(C) A MolScript ribbon drawing of two
stacked subunits. From Wang and Stubbs.
46
337
~1 m
d 7 nm
2.5 nm pitch
PapH
Pap A rod
Pap G
Pap F
2.5 nm
15 nm length
Pap E
Pap K
Figure 7-9 (A) Schematic diagram of a bacterial P pilus. The ~1-m-long helical rod is anchored to the outer cell membrane
by protein Pap H. The adhesion Pap G binds to galactosyl glycolipids of the host. (B, C) The structure of pilus fiber from
Neisseria gonnorrheae modeled from the atomic structure of the 54-residue pilin subunit. The exact structure of the fiber is
uncertain, but the model generated here by trying various possible helical packings matches the dimensions obtained from
fiber diffraction patterns and electron microscopic images. (B) Cross section. (C) Stereoscopic view. The experimental dimensions
of 4.1-nm pitch and 6.0-nm diameters are shown by the transparent ring in (B). From Parge et al.
57
B. Complementarity and the Packing of Macromolecules
handed one-start or primary helix with a pitch of
only 0.54 nm and with approximately two subunits
per turn (Fig. 7-10).
62,63
It can also be described as a right-handed two-start
helix in which two chains of subunits coil around one
another with a long pitch (Fig. 7-10).
B
C
A
2. Oligomers with Twofold (Dyad) Axes
Paired interactions. If two subunits are held
together with interactions aj and are related by a two-
fold axis of rotation as shown in Fig. 7-11, we obtain an
isologous dimer. Each point such as a in one subunit
is related to the same point in the other subunit by
reflection through the axis of rotation. In the center,
along the twofold axis, points c and c are directly
opposite the same points in the other subunit. Figure 7-11
338 Chapter 7. How Macromolecules Associate
is drawn with a hole in the center so that groups c and
c do not actually touch, and it is the paired interactions
such as aj of groups not adjacent to the axis that con-
tribute most to the bonding. However, a real protein
dimer may or may not have such a hole. The pair of
identical interactions in an isologous dimer may be
referred to as a single isologous bond. Such a bond
always contains the paired interactions between
complementary groups (aj) and has pairs of identical
groups along the axis. However, because they are
identical those groups usually cannot interact in a
specific complementary manner.
Isologous bonding is very important in oligomeric
enzymes, and it has been suggested that isologous
interactions evolved early. Initially there may not have
been much complementarity in the bonding but two
hydrophobic spots on the surface of the subunits
came together in a nonspecific association.
64
Later in
evolution the more specific paired interactions could
have been added.
Dihedral symmetry. Isologous dimers can serve
as subunits in the formation of larger closed oligomers
and helices; for example, an isologous pair of the sort
shown in Fig. 7-11A can be flipped over onto the top of
another similar pair as shown in Figs. 7-11B and 7-11C.
Again, if the proper complementary surfaces exist,
bonds can form as shown (bk in Fig. 7-11B and bk and
cl in Fig. 7-11C). Both structures in Figs. 7-11B and 7-11C
possess dihedral (D
2
) symmetry.
65
In addition to the
twofold axis of rotation lying perpendicular to the two
rings, there are two other twofold axes of rotation as
Figure 7-10 (A) Model of the
F-actin helix composed of eight
monomeric subunits. The model
was constructed from the known
structure of the actin monomer
with bound ADP using X-ray data
from oriented gels of fibrous actin
to deduce the helical arrangement
of subunits. The main interactions
appear to be along the two-start
helix. See also Holmes et al.
62
(B) Ribbon drawing of an actin
monomer with the four domains
labeled. Courtesy of Ivan Rayment.
3
2
4
1
B A
339
In 1968, a tiny cylindrical particle, which appeared
to be a stack of 11-nm rings, was observed by electron
microscopy of an extract of erythrocytes.
a,b
Later, a
similar particle was found in both the nucleus and
the cytoplasm of other cells of many organisms. The
particles were soon recognized as a new type of
protein-hydrolyzing enzyme, a large 700-kDa particle
consisting of 2030 subunits of several different types
which came to be known as the multicatalytic pro-
tease or 20S proteasome.
c,d
Electron microscopy
and X-ray diffraction showed that the particle is
formed from four stacked rings, each of which con-
sists of seven subunits whose molecular masses
range from 21 to 31 kDa.
e j
Proteasomes are strikingly similar in architecture,
though not in peptide sequences, to another particle
found in both bacteria and eukaryotes: a molecular
chaperone or chaperonin. The chaperonins, of
which there are several types, protect proteins while
they fold or undergo translocation within cells.
k
One of the best studied members is the E. coli protein
GroEL, which is also composed of double rings of
14 subunits with seven-fold rotational symmetry
and with two of these assemblies associated back-
to-back with dihedral symmetry.
l
The dimensions
of GroEL and 20S proteasomes are nearly the same.
However, GroEL has only two rings of ~ 60-kDa
subunits, more than twice the size of proteosomal
subunits. The accompanying sketch illustrates this
fact and also the basic structural similarity of 20S
proteosomes with GroEL. The pairs of the pro-
teosome, correspond to single subunits of the chap-
eronin, but these subunits have three distinct
domainsapical, intermediate, and equatorial
BOX 7-A LIFE AND DEATH FOR PROTEINS: CHAPERONINS AND PROTEASOMES
(labeled A, I, and E , respectively, in the drawing).
m
After a protein, whether correctly, incorrectly, or
only partially folded, enters a cavity in GroEL, a
second protein GroES of smaller size (~10 kDa)
but with seven-fold symmetry binds to one end of
the chaperonin.
p
Seven molecules of ATP also bind
to sites on the GroEL ring to which GroES binds (the
cis ring). The binding of the ATP and GroES evi-
dently induces a major conformational change in
the GroEL subunits
l,m,n,q
which causes the binding
cavity to expand to over twice the original volume.
This change (see drawing) also causes hydrophobic
surfaces of the cavity to become buried and hydro-
philic side chains to be exposed. The cavity surface
was initially largely hydrophobic and able to bind
many proteins nonspecifically, but upon expansion
it becomes hydrophilic and less likely to bind. This
releases the encased protein to complete its folding
or to partially unfold and refold without interference
from other proteins.
While a protein is adjusting its folding in the cis
compartment another protein molecule may become
trapped in the trans compartment. After some time
the bound ATP molecules are hydrolyzed. As in the
contraction of muscle, which is discussed in Chapter
19, the loss of inorganic phosphate (P
i
) and ADP
from the active site can be accompanied by move-
ment. In the chapenonin this involves a conforma-
tional switch so that the ES heptamer is released and
the conformation of the trans ring of EL is switched
to that of the initial cis ring and vice versa. The new
cis ring is ready to receive an ES cap and the new
trans ring can release the folded protein.
r
A variety
of experimental approaches are being used in an
B. Complementarity and the Packing of Macromolecules
A
I
E E
I
A
E
I
A A
I
E
13.7 nm


1.3 nm
11.3 nm
20S Proteasome
2.7 nm
14.8 nm

GroEL Chaperonin
14.6 nm
4.5 nm 1.0 nm
Expanded GroEL-GroES
complex with enclosed
folding polypeptide
o
Catalytic sites Hydrophobic binding
patches
From Weissman, Sigler and Horwich
g
. Illustrations by K. Sutliff.
Right: From Mayhew and Hartl
o
340 Chapter 7. How Macromolecules Associate
BOX 7-A LIFE AND DEATH FOR PROTEINS: CHAPERONINS AND PROTEASOMES (continued)
effort to further understand the action of GroEL.
s x
The chaperonin may function repeatedly before a
protein becomes properly folded.
t
While chaperonins assist proteins to fold cor-
rectly proteasomes destroy unfolded chains by
partial hydrolysis, cutting the chains into a random
assortment of pieces from 3 to 30 residues in length
with an average length of ~ 8 residues.
y
Proteasomes
destroy not only unfolded and improperly folded
proteins but also proteins marked for destruction by
the ubiquitin system described in Box 10-C. It has
been hard to locate true proteosomes in most bacteria.
However, they do contain protease particles with
similar characteristics
z bb
and archaeons, such as
Thermoplasma acidophilum, have proteasomes similar
to those of eukaryotes.
cc
The Thermoplasma proteasome contains only two
kinds of subunits, and , which have similar amino
acid sequences. These form
7
and
7
rings which
associate in
7

7
pairs with two of these double rings
stacked back-to-back with dihedral D7 symmetry:

7
. The crystal structure has been determined
for this 20S proteasome from T. acidophilum
g,dd
and
for the corresponding proteasome from yeast (Sac-
charomyces cerevisiae).
f,ee
The accompanying drawings
illustrate top and side views of the T. acidophilum
proteasome. The particle contains three internal
cavities. The outer two are formed between the
7
and
7
rings and the inner is formed between the
two
7
rings. A channel only 1.3 nm in diameter
permits the entrance of peptide chains into the
compartments.
The active sites of the enzymes
ff
are located in
the subunits in the central cavity.
dd
While the
yeast and human proteasomes are similar to those
of Thermoplasma, the subunits consist of seven
different protein-hydrolyzing enzymes whose cata-
lytic activities and mechanisms are considered in
Chapter 12. There are also seven different sub-
units, all of whose sequences are known.
i,gg
To make
the story more complex, additional subunits, some
of which catalyze ATP hydrolysis, form a 600- to
700-kDa cap which adds to one or both ends of a 20S
proteosome to give a larger 26S proteasome.
b,d,e,w
These larger proteasomes carry out an ATP-depen-
dent cleavage of proteins selected for degradation
by the ubiquitin system (Box 10-C; Chapter 12).
Some of the short peptide segments formed by pro-
teasomes may leave cells and participate in intercel-
lular communication. For example, pieces of antigenic
peptides are used by cells of the immune system for
antigen presentation (Chapter 31),
hh
an important
process by which the immune system recognizes
which cells are self and which are foreign or malig-
nant and must be killed.
The structure of the caps on the 26S proteasome
ends is complex. At least 20 different regulatory
subunits have been identified.
ii,jj
(A) Electron micrograph of 26S
proteasomes from Xenopus oocytes
negatively stained with 2% uranyl
acetate. (B) Image of the 26S
proteasome (left) and a 20S protea-
some with only one end cap. These
views were obtained by correlation
averaging of 527 individual images
of the 26S proteasome and 395
images of the single-ended form.
From Peters et al.
e
Courtesy of
Wolfgang Baumeister.
100 nm
10 nm
B
A
341
BOX 7-A (continued)
(A) Top view of the 20S protea-
some as an -carbon plot showing
the seven-fold symmetry. The
subunits are in front of the sub-
units. (B) Side view showing the
proteasome cut open along its
seven-fold axis. From Lwe et al.
dd
Courtesy of Robert Huber.
a
Harris, J. R. (1968) Biochim. Biophys. Acta. 150, 534537
b
Peters, J.-M. (1994) Trends Biochem. Sci. 19, 377382
c
Bosch, G., Baumeister, W., and Essen, L.-O. (2000) J. Mol. Biol.
301, 1925
d
Ferrell, K., Wilkinson, C. R. M., Dubiel, W., and Gordon, C.
(2000) Trends Biochem. Sci. 25, 8388
e
Peters, J.-M., Cejka, Z., Harris, J. R., Kleinschmidt, J. A., and
Baumeister, W. (1993) J. Mol. Biol. 234, 932937
f
Groll, M., Ditzel, L., Lwe, J., Stock, D., Bochtler, M., Bartunik,
H. D., and Huber, R. (1997) Nature (London) 386, 463471
g
Weissman, J. S., Sigler, P. B., and Horwich, A. L. (1995) Science
268, 523524
h
DeMartino, G. N., and Slaughter, C. A. (1999) J. Biol. Chem. 274,
2212322126
i
Gerards, W. L. H., de Jong, W. W., Bloemendal, H., and Boelens,
W. (1998) J. Mol. Biol. 275, 113121
j
Schmidtke, G., Schmidt, M., and Kloetzel, P.-M. (1997) J. Mol.
Biol. 268, 95106
k
Netzer, W. J., and Hartl, F. U. (1998) Trends Biochem. Sci. 23,
6873
l
Sigler, P. B., Xu, Z., Rye, H. S., Burston, S. G., Fenton, W. A., and
Horwich, A. L. (1998) Ann. Rev. Biochem. 67, 581608
m
Kawata, Y., Kawagoe, M., Hongo, K., Miyazaki, T., Higurashi,
T., Mizobata, T., and Nagai, J. (1999) Biochemistry 38, 1573115740
n
Betancourt, M. R., and Thirumalai, D. (1999) J. Mol. Biol. 287,
627644
o
Mayhew, M., and Hartl, F. U. (1996) Science 271, 161162
p
Hunt, J. F., Weaver, A. J., Landry, S. J., Gierasch, L., and
Deisenhofer, J. (1996) Nature (London) 379, 3745
q
Cliff, M. J., Kad, N. M., Hay, N., Lund, P. A., Webb, M. R.,
Burston, S. G., and Clarke, A. R. (1999) J. Mol. Biol. 293, 667684
r
Rye, H. S., Burston, S. G., Fenton, W. A., Beechem, J. M., Xu, Z.,
Sigler, P. B., and Horwich, A. L. (1997) Nature (London) 388,
792798
s
Chatellier, J., Buckle, A. M., and Fersht, A. R. (1999) J. Mol. Biol.
292, 163172
t
Nieba-Axmann, S. E., Ottiger, M., Wthrich, K., and Plckthun,
A. (1997) J. Mol. Biol. 271, 803818
u
Gervasoni, P., Gehrig, P., and Plckthun, A. (1998) J. Mol. Biol.
275, 663675
v
Torella, C., Mattingly, J. R., Jr., Artigues, A., Iriarte, A., and
Martinez-Carrion, M. (1998) J. Biol. Chem. 273, 39153925
w
Horwich, A. L., Weber-Ban, E. U., and Finley, D. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1103311040
x
Buckle, A. M., Zahn, R., and Fersht, A. R. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 35713575
y
Kisselev, A. F., Akopian, T. N., and Goldberg, A. L. (1998) J. Biol.
Chem. 273, 19821989
z
Kessel, M., Maurizi, M. R., Kim, B., Kocsis, E., Trus, B. L., Singh,
S. K., and Steven, A. C. (1995) J. Mol. Biol. 250, 587594
aa
Shin, D. H., Lee, C. S., Chung, C. H., and Suh, S. W. (1996) J.
Mol. Biol. 262, 71 76
bb
Tamura, T., Tamura, N., Cejka, Z., Hegerl, R., Lottspeich, F., and
Baumeister, W. (1996) Science 274, 13851389
cc
Maupin-Furlow, J. A., and Ferry, J. G. (1995) J. Biol. Chem. 270,
2861728622
dd
Lwe, J., Stock, D., Jap, B., Zwickl, P., Baumeister, W., and
Huber, R. (1995) Science 268, 533539
ee
Stuart, D. I., and Jones, E. Y. (1997) Nature (London) 386, 437
438
ff
Voges, D., Zwickl, P., and Baumeister, W. (1999) Ann. Rev.
Biochem. 68, 10151068
gg
Schmidt, M., and Kloetzel, P.-M. (1997) FASEB J. 11, 12351243
hh
Goldberg, A. L., and Rock, K. L. (1992) Nature (London) 357,
375379
ii
Adams, G. M., Falke, S., Goldberg, A. L., Slaughter, C. A.,
DeMartino, G. N., and Gogol, E. P. (1997) J. Mol. Biol. 273,
646657
jj
Knowlton, J. R., Johnston, S. C., Whitby, F. G., Realini, C.,
Zhang, Z., Rechsteiner, M., and Hill, C. P. (1997) Nature
(London) 390, 639643
B. Complementarity and the Packing of Macromolecules
A
B
342 Chapter 7. How Macromolecules Associate
Figure 7-12 A ribbon representation of the ornithine decar-
boxylase dodecamer. Six dimers of the 730-residue subunits
are related by C
6
crystallographic symmetry. MolScript
drawing from Momany et al.
66
Courtesy of Marvin Hackert.
indicated in the drawings. Again,
the interactions are paired and iso-
logous; of many possible contacts
two bk interactions and two cl inter-
actions are marked for each pair of
subunits in Fig. 7-11. There are a
total of six pairs of these interac-
tions, one between each combina-
tion of two subunits. This may be a
little more difficult to see in Fig. 7-11B
than in Fig. 7-11C because in the
former the subunits are arranged in
a more or less square configuration.
Nevertheless, a pair of interactions
between the left-hand subunit in
the top ring and the subunit in the
lower ring at the right does exist,
even if it is only electrostatic and at
a distance. An example of a tetra-
meric enzyme with perfect dihedral
symmetry of the type shown in Fig.
7-11B is lactate dehydrogenase
(Chapter 15). The plant agglutinin
concanavalin A has a quaternary
structure resembling that in Fig. 7-11C.
Square arrays of four subunits
can be formed using either heterolo-
gous or isologous interactions. Both types of bonding
can occur in larger aggregates. For example, two
trimers such as that shown in Eq. 7-25 can associate to
a hexamer having dihedral (D
3
) symmetry; a heterolo-
gous square tetramer can dimerize to give a dihedral
(D
4
) octamer.
65
The enzymes ornithine decarboxylase
(Fig. 7-12)
66
and glutamine synthetase (Chapter 24)
67
each consist of double rings of six subunits each. The
upper ring is flipped over onto the lower giving dihe-
dral symmetry (D
6
) with one 6-fold axis and six 2-fold
axes at right angles to it.
Oligomers with cubic symmetry (polyhedra).
Symmetrical arrangements containing more than one
axis of rotation of order higher than 2-fold are said to
have cubic symmetry. The tetrahedron is the simplest
example. It contains four 3-fold axes which pass through
the vertices and the centers of the faces and three 2-fold
axes which pass through the midpoints of the six edges.
Since protein subunits are always asymmetric, a tetrameric
protein cannot possess cubic symmetry. As we have already
seen, tetrameric enzymes have dihedral symmetry.
However, a heterologous trimer with 3-fold symmetry
can form a face of a tetrahedron containing a total of
12 asymmetric subunits. Twenty-four subunits can
interact to form a cube. Three 4-fold axes pass through
the centers of the faces, four 3-fold axes pass through
the vertices, and six 2-fold axes pass through the edges
(see Figs. 7-13 and 16-3).
The largest structure of cubic symmetry that can
c
c
c'
j j
a
a
j'
b
b
k
a'
j
j
a
a b
k
k j' a'
c
c
b
k l
l
k
b
j
j
a
a
Figure 7-11 (A) Isologous bonding between pairs of subunits; (B) an isologous
square arrangement of subunits; (C) an apparently tetrahedral arrangement of
subunits. Note the three twofold axes.
B
C
A
343
Figure 7-13 (A) MolScript
ribbon drawing of a subunit of
the iron oxide storage proteins
L-ferritin from amphibian red
cells. This 4-helix bundle is
represented by cylinders of 1.3
nm diameter in the oligomer.
(B) Helices A and C of the mono-
mer are on the outer surfaces of
the oligomer and helices B and
D are on the inner surface. The
oligomer consists of a shell of 24
subunits and is viewed down a
4-fold axis illustrating its 423 (cubic)
symmetry. The molecule is
illustrated further in Fig. 16-3.
From Trikha et al.
74
Courtesy of
Elizabeth Theil.
B. Complementarity and the Packing of Macromolecules
B A
Figure 7-14 (A) Schematic drawing illustrating an icosahedrally symmetric structure with sixty identical asymmetric sub-
units all in equivalent positions. The 5-fold axes are located at the vertices of the icosohedron and the 2-fold and 3-fold axes
can readily be seen. (B) Ribbon drawing of the 195-residue polypeptide chain of the coat subunit of satellite tobacco necrosis
virus. The protein folds into an inwardly projecting N-terminal segment and a -jellyroll domain. The packing of this
subunit in the virus particle is shown schematically in (C). The symmetry axes drawn next to the subunit diagram (B) can be
used to position it in the structure. Contacts between subunits are labeled D, T, and P (dimer, trimer, pentamer). Diagrams
courtesy of Drs. Strandberg, Liljas, and Harrison.
68
(D) The distribution of RNA helical segments in a hemisphere of a virion
of a similar small virus, the satellite tobacco mosaic virus. The virion viewed down a 3-fold axis from the virus exterior. The
helical axes of the RNA segments are along icosahedral edges. From Larson et al.
75
5
2
3
5
P
D
T
C D
A
B
344 Chapter 7. How Macromolecules Associate
be made is the icosahedron, a regular solid with 20
triangular faces. Sixty subunits, or some multiple of
60, are required and at each vertex they form a heterol-
ogous pentamer. As with the tetrahedron, each face
contains a heterologous trimer, while isologous bonds
across the edges form dimers (Fig. 7-14C). Many
viruses consist of roughly spherical protein shells
(coats) containing DNA or RNA inside.
68 70
As with
the filamentous viruses, the protein coats consist of
many identical subunits, a fact that can be rationalized
in terms of economy from the genetic viewpoint. Only
one gene is needed to specify the structure of a large number
of subunits.
70,71
Under the electron microscope the viruses
often have an icosahedral appearance (Figs. 5-41A, 7-14),
and chemical studies show that the number of the
most abundant subunits is usually a multiple of 60.
An example is the tiny satellite tobacco necrosis
virus,
72
diameter ~ 18 nm, whose coat contains just
60 subunits of a 195-residue protein. Its genome is a
1239-nucleotide molecule of RNA. The structure of
the coat has been determined to 0.25-nm resolution.
73
Many virus coats have 180 subunits or a number
that is some other multiple of 60. However, in these
coats the subunits cannot all be in identical environ-
ments. Two cases may be distinguished. If all of the
subunits have identical amino acid sequences they
probably exist in more than one distinct conformation
that permit them to pack efficiently. (Next section)
Alternatively, two or more subunits of differing sequence
and structure may associate to form 60 larger subunits
that do pack with icosahedral symmetry. For example,
the polioviruses (diameter 25 nm) contain three major
coat proteins (, , and or VP1, VP2, and VP3). These
are formed by cleavage of a large precursor protein
into at least four pieces.
76,77
The three largest pieces of
~ 33-, 30-, and 25-kDa mass (306, 272, and 238 residues,
respectively) aggregate as ()
60
. Sixty copies of a
fourth subunit of 60 residues are found within the shell.
Related picorna viruses such as
human rhinoviruses (Fig. 7-15),
69,78,79
foot-and-mouth disease virus, parvo-
virus,
80
and Mengo virus
81
have
similar architectures. The small
(diameter 25 nm) single-stranded
DNA bacteriophages such as X174
also have three different coat pro-
teins, one of which forms small
hollow spikes at the vertices of the
icosahedral shell (Fig. 5-41A).
82
Asymmetry and quasi-equiva-
lence in oligomers. It is natural to
think about association of subunits
in symmetric ways. Consequently
the observation of square, pentagonal,
and hexagonal arrangements of sub-
units directly with the electron micro-
scope led to a ready acceptance of
the idea that protomers tend to
associate symmetrically. However,
consider the predicament of the two
molecules shown in Fig. 7-16. They
might get together to form an isolo-
gous dimer if it were not for the fact
that their noses are in the way.
Despite the obvious steric hindrance,
an isologous dimer can be formed
in this case if one subunit is able to
undergo a small change in confor-
mation (Fig. 7-16). In the resulting
dimer the two subunits are only
quasi-equivalent.
Unsymmetrical dimerization of
proteins appears to be a common
phenomenon that is often observed
in protein crystals. For example, the
Figure 7-15 (A) Schematic diagram of the icosahedral shell of a human
rhinovirus showing the arrangement of the three subunits VP1, VP2, and
VP3, each present as 60 copies. (B) Stereoscopic view of an image of the
virus decorated by the binding of two immunoglobulinlike domains of the
intercellular adhesion molecule ICAM-1, a natural receptor for the virus.
Part of this receptor binds into a groove or canyon, which in marked in (A)
by the dark bands. From Olson et al.
78
Courtesy of Michael Rossmann.
B
A
50 nm
345
enzymes malic dehydrogenase and glyceraldehyde
phosphate dehydrogenase (Chapter 15) are both tetra-
mers of approximate dihedral symmetry but X-ray
crystallography revealed distinct asymmetries
83,84
which include a weaker binding of the coenzyme
NAD
+
in one subunit. This may simply reflect differ-
ences in environment within the crystal lattice. How-
ever, negative cooperativity in coenzyme binds has also
been revealed by kinetic experiments.
85
The polypeptide hormone insulin is a small protein
made up of two chains (designated A and B) which are
held together by disulfide bridges (Fig. 7-17A). Figure
7-17B is a sketch of the structure as revealed by X-ray
crystallography,
86,87
with only the backbone of the
peptide chains and a few side chains shown. In the
drawing, the B chain lies behind the A chain. Beginning
with the N-terminal Phe 1 of the B chain the peptide
backbone makes a broad curve, and then falls into an
helix of three turns lying more or less in the center of
the molecule. After a sharp turn, it continues upward
on the left side of the drawing in a nearly completely
extended structure. The A chain has an overall U
shape with two roughly helical portions. The U shape
is partly maintained by a disulfide bridge running
between two parts of the A chain. Two disulfide bridges
hold the A and B chains together, and hydrophobic
bonding of internal side chain groups helps to stabilize
the molecule.
Insulin in solution dimerizes readily, the subunits
occupying quasi-equivalent positions. Figure 7-17C
shows some details of the bonding between the sub-
units in the insulin dimer with a view from the outside
of the molecule down the 2-fold axis (marked by the X
in the center of the Phe 25 ring in the right-hand chain)
through the dimer. The C-terminal ends of the B chains
are seen in an extended conformation. The two anti-
parallel chains form a structure with two pairs of
hydrogen bonds. If there were perfect isologous bond-
ing, the two pairs would be entirely equivalent and
symmetrically related one to the other. A straight line
drawn from a position in one chain and passing through
the twofold axis (X) would also pass approximately
through the corresponding position in the other chain.
However, there are many deviations from perfect
symmetry, the most striking of which is at the center
where the Phe 25 from the right-hand chain projects
upward and to the left. If the symmetry were perfect
the corresponding side chain from the left-hand chain
would project upward and to the right and the two
phenylalanines would collide, exactly as do the noses
in Fig. 7-16. In insulin one phenylalanine side chain
has been flipped back out of the way.
Under proper conditions, three insulin dimers
associate to form a hexamer of approximate dihedral
(D
3
) symmetry that is stabilized by the presence of
two zinc ions. Figure 7-17D is a crude sketch of the
hexamer showing the three dimers, the 3-fold axis of
symmetry, and the two pseudo 2-fold axes, one passing
between the two subunits of the dimer and the other
between two adjacent dimers. Figure 7-18 is a stereo-
scopic ribbon diagram of the atomic structure, with the
A chains omitted, as obtained by X-ray diffraction.
87
The structure has also been obtained by NMR spectro-
scopy.
88
Note that each of the two zinc atoms lies on
the threefold axis and is bound by three imidazole
rings from histidines B-10. The significance of the zinc
binding is uncertain but these hexamers readily form
rhombohedral crystals, even within the pancreatic cells
that synthesize insulin. The structure illustrates a
feature that is common to many oligomers of circular
or dihedral symmetry. A central channel is often
quite open and protruding side chain groups, such as
the imidazole groups in insulin, form handy nests into
which ions or molecules regulating activity of proteins
can fit. Conformational differences in insulin are in-
duced by the binding of phenol. In Fig. 7-18A the
C-terminal ends of the chains are extended but in the
phenol complex (B) they have coiled to extend the
helices.
Quasi-equivalence in virus coats. A large
number of icosahedral viruses have coats consisting
of 180 identical subunits. For example, the small
RNA-containing bacteriophage MS 2 consists of an
eicosahedral shell of 180 copies of a 129-residue pro-
tein that encloses one molecule of a 3569-residue
RNA.
89
The virus also contains a single molecule of a
44-kDa protein, the A protein, which binds the virus
to a bacterial pilus to initiate infection. Related bacte-
riophages GA, fr, f2, and Q
90,91
have a similar
B. Complementarity and the Packing of Macromolecules
1 2
1 2
Figure 7-16 Nonsymmetric bonding in a
dimer. (A) Two molecules which cannot
dimerize because of a bad fit at the center.
(B) A solution: Molecule 1 has refolded its
peptide chain a little, changing shape enough
to fit to molecule 2.
B A
346 Chapter 7. How Macromolecules Associate
3-fold axis
3.5 nm
5.0 nm
Local 2-fold
axis through
dimer
Dimer-dimer
axis
Zn
Zn
Figure 7-17 The structure of insulin. (A) The amino acid
sequence of the A and B chains linked by disulfide bridges. (B)
Sketch showing the backbone structure of the insulin molecule
as revealed by X-ray analysis. The A and B chains have been
labeled. Positions and orientations of aromatic side chains are
also shown. (C) View of the paired N-terminal ends of the B
chains in the insulin dimer. View is approximately down the
pseudo-twofold axis toward the center of the hexamer. (D)
Schematic drawing showing packing of six insulin molecules in
the zinc-stabilized hexamer.
Gly Ile Val Glu Gln Cys
Cys Tyr Ser
Ile
S S
Cys Ser Leu Tyr Gln Leu
Gln
Ala Leu
Gln
Val
Leu
Tyr
Leu
Val
Cys Gly Ser His
S
Cys Leu His Gln Asn Val Phe
1
1
Gly
Gln
Arg
S S
Asn 21
Cys
Tyr
Asn
Gly
Phe
Phe
Tyr Thr Pro Lys Ala 30
S
A.
B.
B
C
A
D
architecture. Many RNA-containing viruses of plants
also have 180 subunits in their coats.
68
Much studied
are the tomato bushy stunt virus (diameter ~ 33 nm,
40-kDa subunits),
68
and the related southern bean
mosaic virus.
92
The human wart virus (diameter
~ 56 nm) contains 420 subunits, seven times the number
in a regular icosahedron. Adenoviruses (diameter
~100 nm) have 1500 subunits, 25 times more than the
60 in a regular icosahedron.
93,94
Caspar and Klug
95
proposed a theory of quasi-equivalence of subunits
according to which the distances between the centers
of subunits are preserved in a family of icosadeltahedra
containing subunits in multiples of 20. However, the
angles must vary somewhat from those in a regular
icosahedron (compare with geodesic shells in which
the angles are constant but the distances are not all the
same). The resulting polyhedra contain hexamers as
well as pentamers at vertices; for example, the shells
of the 180-subunit viruses contain clusters of subunits
forming 12 pentamers and 20 hexamers. There are
347
Figure 7-18 Stereoscopic MolScript ribbon drawings of the B chains (A chains
omitted) of (A) hexameric 2-zinc pig insulin. (B) A phenol complex of the same
protein. Within each dimer the B chains are shaded differently. The Zn
2+
ions
are represented by white spheres and the coordinating histidine side chains are
shown. Six noncovalently bound phenol molecules can be seen, as can several
conformational differences. From Whittingham et al.
87
Courtesy of Peter C. E.
Moody.
also 60 trimers (on the faces) and 90
dimers (across the edges) (Fig. 7-19).
Such structures can be formed only
for certain values of T where the
number of subunits is 60T and
there are 12 pentamers (pentons)
and 10 (T-1) hexamers hexons. T
can assume values of h
2
+ hk + k
2
where h and k may be positive
integers or zero. Some allowed T
values are 1, 3, 4, 7, 9, 13, 25.
68,70,96,97
The subunits in virus coats
with T greater than one are not all
in equivalent positions. For example,
the three subunits labeled A, B,
and C in Fig. 7-19 are each slightly
differently positioned with respect
to neighboring subunits. Since
virus coats are usually tightly
packed the subunits must assume
more than one conformation. One
kind of conformational change that
allows quasi-equivalence of sub-
units is observed in the tomato
bushy stunt virus. Two structural
domains are connected by a hinge
which allows an outer protruding
domain to move slightly to pre-
serve good isologous interactions
with a corresponding domain in
another subunit.
68
The southern bean mosaic
virus has an eight-stranded anti-
parallel -barrel structure closely
similar to that of the major domain
of the bushy stunt viruses but
lacking the second hinged domain.
The problem of quasi-equivalence
is resolved by the presence of an N-
terminal extension that binds onto
a subunit across the quasi-six-fold
axis to give a set of three subunits
(labeled C in Fig. 7-19) that associate
with true three-fold symmetry and
another set (B) with a slightly differ-
ent conformation fitting between
them.
68,92
The subunits A, which
have a third conformation, fit to-
gether around the five-fold axis in
true cyclic symmetry.
A surprising finding is that the
polyoma virus coat, which was
expected to contain 420 (7 x 60)
subunits, apparently contains only
360. The result is that the hexavalent
morphological unit is a pentamer
and that quasi-eqivalence appears
B. Complementarity and the Packing of Macromolecules
B
A
A
B
C
Figure 7-19 Schematic
icosahedrally symmetric
structure with 180 sub-
units. The quasi-equivalent
units A, B, and C are
necessarily somewhat
differently positioned with
respect to their neighbors
and must therefore assume
different conformations in
order to fit together tightly.
From Harrison.
68
348 Chapter 7. How Macromolecules Associate
to be violated.
98,99
Flexible arms tie the pentamers
together.
Quasi-equivalence of subunits also provides the
supercoil in bacterial flagella (Chapter 19) and accounts
for some interesting aspects of the structure of tobacco
mosaic virus. The protein subunits of the virus can
exist either as a helix with 16.3 subunits per turn (Fig.
7-8) or as a flat ring of 17 subunits.
100
A very small
conformational difference is involved. These rings
dimerize but do not form larger aggregates. What is
surprising is that the dimeric rings do not have dihe-
dral symmetry, all of the subunits in the dimeric disk
being oriented in one direction but with two different
conformations. The disk may serve as an intermediate
in virus assembly. The inner portions of the quasi-
equivalent disk subunits have a jawlike appearance
as if awaiting the incorporation of RNA. As the RNA
becomes bound, the disks could dislocate to a lock-
washer conformation to initiate and to propagate
growth of the helical virus particle.
100,44a
However,
there is uncertainty about this interpretation.
45,101
Some enzymes, such as yeast hexokinase and
creatine kinase (Chapter 12), associate in extremely
asymmetric ways.
102
A dimer is formed by means of
heterologous interactions but steric hindrance prevents
the unsatisfied sets of interacting groups from joining
with additional monomers to form higher polymers.
As Galloway pointed out, many biological structures
are not completely ordered but nevertheless possess
well-defined and functionally important local relation-
ships.
103
Regulatory subunits and multienzyme complexes.
Proteins are often organized into large complexes,
sometimes for the purpose of regulating metabolism.
An example is aspartate carbamoyltransferase
which catalyzes the first step in the synthesis of the
pyrimidine rings of DNA and RNA (Chapter 25). The
310-kDa enzyme from E. coli can be dissociated into
two 100-kDa trimers, referred to as catalytic subunits,
and three 34-kDa dimers, the regulatory subunits
which alter their conformations in response to changes
in the ATP, UTP, and CTP concentrations.
104 107
The
molecule is roughly triangular in shape
47,108
with a
thickness of 9.2 nm and a length of the triangular side
of 10.5 nm (Fig. 7-20). The symmetry is 3:2, i.e., it is
dihedral with one 3-fold axis of rotation and three 2-
fold axes. The two trimers of catalytic subunits lie
face-to-face with the dimeric regulatory subunits fitting
between them into the grooves around the edges of the
trimers (Fig. 7-20). The dimers are not aligned exactly
parallel with the 3-fold axis, but to avoid eclipsing, the
upper half of the array is rotated around the 3-fold
axis with respect to the lower half. In the center is an
aqueous cavity of dimensions ~ 2.5 x 5.0 x 5.0 nm. The
active sites of the enzyme are inside this cavity which is
reached through six ~1.5-nm opening around the sides.
Many other oligomeric enzymes and other complex
assemblies of more than one kind of protein subunit are
known. For example, the 2-oxoacid dehydrogenases
are huge 2000- to 4000-kDa complexes containing three
different proteins with different enzymatic activities in
a cubic array (Fig. 15-14). The filaments of striated
muscle (Chapter 19), antibodies and complement of
blood (Chapter 31), and the tailed bacteriophages
(Box 7-C ) all have complex molecular architectures.
H8
H2
H10
H7
H11
H4
H9
H1
H2
H3
Zinc domain
Carbamoylphosphate domain
H3
Allosteric domain
H12
H6
H5
H1
Figure 7-20 (A) Subunit assembly of two C
3
catalytic
trimers (green) and three R
2
regulatory dimers around the
periphery in aspartate carbamoyltransferase. After Krause
et al.
109
Courtesy of William N. Lipscomb. The aspartate-
and carbamoylphosphate-binding domains of the catalytic
subunits are labeled Asp and CP, respectivley, while the zinc
and allosteric domains of the regulatory subunits are labeled
Allo and Zn, respectively. (B) Ribbon drawing of a single
pair of regulatory (left) and catalytic (right) subunits with
the structural domains labeled. MolScript drawing from
Thomas et al.
110
Asp
CP
Zn
Allo
B
A
349
C. Cooperative Changes in Conformation
A substrate will bind better to some conformations
of a protein than it will to others. This simple fact,
together with the tendency for protein monomers to
associate into clusters, allows for cooperative changes
in conformation within oligomeric proteins. These
changes provide the basis for important aspects of the
regulation of enzymes and of metabolism. They im-
part cooperativity to the binding of small molecules
such as that of oxygen to hemoglobin and of substrates
and regulating molecules to enzymes. Many of the
most fundamental and seemingly mysterious properties
of living things are linked directly to cooperative changes
within the fibrils, membranes, and other structures of
the cell.
In 1965 a simple, appealing mathematical descrip-
tion of cooperative phenomena was suggested by
Monod, Wyman, and Changeux
30,110a,110b
and focused
new attention on the phenomenon. They suggested
that conformational changes in protein subunits,
which could be associated with altered binding charac-
teristics, occur cooperatively within an oligomer. For
example, binding of phenol to hexameric 2-zinc insulin
(Fig. 7-18) could induce all six individual subunits to
change their conformation together, preserving the D
6
symmetry of the complex. (In fact, it is more complex
than this.
111
) The four subunits of hemoglobin could
likewise change their conformation and affinity for O
2
synchronously. This is very nearly true and is of major
physiological significance.
Consider an equilibrium (Eqs. 7-27 and 7-28)
between protein molecules in two different conforma-
tions A and B (T and R in the MWC terminology) and
containing a single binding site for molecule X. In the
MonodWymanChangeux (MWC) model the con-
formations are designated T (tense) and R (relaxed)
but in the interest of providing a more general treat-
ment the terminology used in this book is that of
Koshland et al.
13,112 115
K
t
= [B] / [A]
If the equilibrium constant K
t
is approximately 1, the
two conformers have equal energies, but if K
t
< 1, A
is more stable than B.
1. Unequal Binding of Substrate and
Induced Fit
Assume that conformer B binds X more strongly
than does conformer A (as is suggested by the shapes
of the binding sites in Eq. 7-27). The intrinsic binding
constants to the A and B conformers K
AX
and K
BX
(or
K
T
and K
R
) are defined by Eq. 7-29:
K
AX
= [AX] / [A] [X]
K
BX
= [BX] / [B] [X])
The entire set of equilibria for this system are shown in
Eq. 7-30. Note that the constant relating
BX to AX is not independent of the other three constants
but is given by the expression K
t
K
BX
/K
AX
. Now con-
sider the following situation. Suppose that A predom-
inates in the absence of X but that X binds more tightly
to B than to A. There will be largely either free A or BX
in the equilibrium mixture with smaller amounts of
AX and B. An interesting kinetic question arises. By
which of the two possible pathways from A to BX (Eq.
7-30) will the reaction take place? The first possibility,
assumed in the MWC model, is that X binds only to
preformed B, which is present in a small amount in
equilibrium with A. The second possibility is that X
can bind to A but that AX is then rapidly converted to BX.
We could say that X induces a conformational change that
leads to a better fit. This is the basis for the induced fit
theory of Koshland. Bear in mind that the equilibrium
constants can give us the equilibrium concentrations
of all four forms in Eq. 7-30. However, rates of reaction
are often important in metabolism and we cannot say
a priori which of the two pathways will be followed.
If K
BX
/K
AX
is very large, an insignificant amount
of AX will be present at equilibrium. In such a case
there is no way experimentally to determine K
AX
. The
two constants K
t
and K
BX
are sufficient to describe the
equilibria but an induced fit mechanism may still hold.
Now consider the association of A and B to form
oligomers in which the intrinsic binding constants K
AX
and K
BX
have the same values as in the monomers.
Since more enzymes apparently exist as isologous
dimers than as any other oligomeric form,
116
it is
appropriate to consider the behavior of such dimers
in some detail. Monod et al. emphasized that both
(7-27)
K
t
X
A (or T) conformer
X
B (or R) conformer
(7-28)
(7-29)
K
t
A B
AX BX
X
X
K
BX
K
t
K
BX
/K
AX
K
AX
(7-30)
C. Cooperative Changes in Conformation
350 Chapter 7. How Macromolecules Associate
2
2
2 2
2 2 2
2 2 2 2
1
2
2 2
2 2
2 2
Y based on dimer
AX BX A X A X ABX
AXB ABX B X B X
A AX B BX A A X
A X AB ABX AXB
ABX B
( )
[ ] [ ] [ ] [ ] [ ]
[ ] [ ] [ ] [ ]
( [ ] [ ] [ ] [ ] ) [ ] [ ]
[ ] [ ] [ ] [ ]
[ ] [ ]
=
+ + + +
+ + + +
+ + + + +
+ + + +
+ + + [[ ] [ ] B X B X
2 2 2
+
(7-35)
conformers A and B (T and R) can associate to form
isologous dimers in which symmetry is preserved (Eq.
7-31).
On the other hand, association of B and A would lead
to an unsymmetric dimer in which bonding between
subunits might be poor:
In the MWC treatment, the assumption is made
that the mixed dimer AB can be neglected entirely.
However, a more general treatment requires that we
consider all dimeric forms. The formation constants
K
AA
, K
BB
, and K
AB
are defined as follows
13,113
(Eqs. 7-32
to 7-34; note the statistical factor of 2 relating K
AB
to
the association constant K
f
):
2. Binding Equilibria for a Dimerizing Protein
All of the equilibria of Eqs. 7-28 through 7-34
involved in formation of dimers A
2
, AB, and B
2
and in
the binding of one or two molecules of X per dimer are
depicted in Fig. 7-21. Above each arrow the microscopic
constant associated with that step is shown multiplied
by an appropriate statistical factor. The fractional
saturation Y is given by Eq. 7-35. Each of the nine
terms in the numerator gives the concentration of
bound X represented by one of the nine forms contain-
ing X in Fig. 7-21. The 14 terms in the denominator
represent the concentration of protein in each form
including those containing no bound X. Protein con-
centrations are given in terms of the molecular mass
of the dimer; hence, some of the terms in the denomi-
nator are multiplied by 1/ 2.
All of the terms in both the numerator and the
denominator of Eq. 7-35 can be related back to [X],
using the microscopic constants from Fig. 7-21 to give
an equation (comparable to Eq. 7-8) which presents Y
in terms of [X], K
AX
and K
BX
, K
t
, and the interaction
constants K
AA
, K
AB
, and K
BB
. Since the equation is too
complex to grasp immediately, let us consider several
specific cases in which it can be simplified.
The Monod-Wyman-Changeux (MWC) model.
If both K
AA
and K
BB
are large enough, there will be no
dissociation into monomers. The transition between
conformation A and conformation B can occur cooper-
atively within the dimer or higher oligomer, and the
mathematical relationships shown in Fig. 7-21 are still
appropriate. One further restriction is needed to describe
the MWC model. Only symmetric dimers are allowed.
That is, K
AA
and K
BB
>> K
AB
(see Eq. 7-31), and only
those equilibria indicated with green arrows in Fig. 7-21
need be considered.
30
In the absence of ligand X, the
ratio [B
2
] / [A
2
] is a constant, 1/ L in the MWC termi-
nology (Eq. 7-36; see also Eq. 7-31).
Both of the association constants K
AA
and K
BB
and the
transformation constant K
t
affect the position of the
equilibrium. Thus, a low ratio of [B
2
] to [A
2
] could
result if K
BB
and K
AA
were similar but K
t
was small.
If K
t
were ~ 1 a low ratio could still arise because K
AA
>
(7-31)
K = 1/L (in MWC terminology)
A A B B
Mixed AB dimer
which associates weakly
A B
2
2
A
A
AA
= K
[ ]
[ ]
2
2
2
2
2
B
B
B
A
BB
t
= = K
K
[ ]
[ ]
[ ]
[ ]
2
2
AB
A B
AB
A
f AB
t
= = = K K
K
[ ]
[ ][ ]
[ ]
[ ]
(7-32)
(7-33)
(7-34)
2A A
2
2B B
2
A + B AB
[ ]
[ ]
B
A
BB
AA
t
2
2
2
1
= =
L
K
K
K
(7-36)
351
For an oligomer with n subunits Monod et al. assumed
that all sites in either conformer are independent and
equivalent. The equation for Y (based on Eq. 7-17) is
We assume initially that B
2
binds X more strongly
than does A
2
. Hence, if the equilibrium in Eq. 7-36
favors B
2
strongly (L is small), the addition of X to the
system will not shift the equilibrium between the two
conformations and binding will be noncooperative
(Eq. 7-39 will reduce to Eq. 7-18). However, if the
equilibrium favors A
2
(L is large), addition of X will
shift the equilibrium in favor of B
2
(which binds X
more tightly). Furthermore, since the expression for Y
(Eq. 7-39) contains a term in K
2
BX
[X]
2
in the numerator,
binding will tend to be cooperative. In the extreme
case that L is large and K
AX
~ 0, most of the terms in
Eq. 7-39 drop out and it approaches the equation
previously given for completely cooperative binding
(Eq. 7-21) with K = K
BX
2
L. With other values of K
AX
,
K
BX
, and L incomplete cooperativity is observed.
112
Further development of the MWC theory as it applies
to enzyme kinetics is given in Chapter 9, Section B.
The induced fit model. In this model, only A
2
,
ABX, and B
2
X
2
are considered (heavy arrows in Fig.
7-15).
13,114
The expression for 2Y is:
The constants used here are defined by Eqs. 7-8 through
7-10 and differ from those of Koshland, who sometimes
arbitrarily set K
AA
= 1 and redefined K
BB
as an interac-
tion constant equal to K
BB
/ K
AA
. Although this simplifies
the algebra it is appropriate only for completely asso-
ciated systems and might prove confusing.
When K
AB
is small (no mixed dimer) Eq. 7-16
also simplifies to Eq. 7-45 for completely cooperative
binding with the value K given by Eq. 7-17. On the
other hand, if K
AB
is large compared to K
AA
and K
BB
,
anticooperativity (negative cooperativity) will be
observed. The saturation curve will contain two sepa-
rate steps just as in the binding of protons by succinate
dianion (Fig. 7-5).
K
BB
, i.e., because the subunits are associated more
tightly in A
2
than in B
2
. For this case Eq. 7-35 simplifies
to Eq. 7-37.
Substituting from Eq. 7-36 into Eq. 7-37 we obtain (Eq.
7-38):
+
A
K
AA
A
2
X
2 K
AX
X
A
2
X
X
A
2
X
2
X
2 K
BX
K
t
K
AB
/K
AA
AXB
X
ABX
2
X
X
K
AX
/2
X
ABX AB X
K
AX
X
K
BX
B
2
+
B
K
BB
B
2
X B
2
X
2
X
2 K
BX
X X X
X
K
BX
/2
K
t
2 K
AB
L
X
AX
X
K
AX
X
K
BX
BX
X
K
BX
K
t
K
BB
/2 K
AB
X
X
X
K
AX
K
BX
X
X
Figure 7-21 Possible forms of a dimerizing protein existing
in two conformations with a single binding site per protomer
for X. Green arrows indicate equilibria considered by MWC.
Solid arrows indicate equilibria considered by Koshland et
al.
13,114
Heavy gray arrows are for the simplest induced fit
model with no dissociation of the dimer. Note that all equilibria
are regarded as reversible (despite the unidirectional arrows).
K
AX
and K
BX
are assumed the same for subunits in monomeric
and dimeric forms.
2
2 2
2 2
2 2
2
2 2 2 2 2 2
2 2 2 2 2 2 2 2
2 2
2 2 2
Y
A X A X B X B X
[A ] + [A X] + [A X ] + [B ] + [B X] + [B X ]
X X
X X
X
AA AX AA AX
BB BX t BB BX t
AA AA AX AA
=
+ + +
=
+
+ +
+ +
[ ] [ ] [ ] [ ]
[ ] [ ]
[ ] [ ]
[ ]
K K K K
K K K K K K
K K K K KK K K
K K K K K K
AX BB t
BB BX t BB BX t
X
X X
2 2 2
2 2 2 2
2
[ ]
[ ] [ ]
+
+ +
(7-37)
C. Cooperative Changes in Conformation
Y for dimer
X X X X
X X
AX AX BX BX
AX BX
( )
[ ]( [ ]) [ ]( [ ])
( [ ]) ( [ ])
=
+ + +
+ + +
L K K K K
L K K
1 1
1 1
2 2
(7-38)
Y
X X X X
X X
AX AX BX BX
AX BX
=
+ + +
+ + +

L K K K K
L K K
n n
n n
[ ]( [ ]) [ ]( [ ])
( [ ]) ( [ ])
1 1
1 1
1 1
(7-39)
2
2
2 2
1 2
2 2
2 2 2
2 2
2 2
Y
ABX B X
A ABX B X
X X
X X
BX t
AB
AA
BX t
BB
AA
BX t
AB
AA
BX t
BB
AA
=
+
+ +
=
+
+ +
[ ] [ ]
[ ] [ ] [ ]
[ ] ( ) [ ]
[ ] ( ) [ ]
K K
K
K
K K
K
K
K K
K
K
K K
K
K
(7-40)
352 Chapter 7. How Macromolecules Associate
Since Gibbs energies are additive, the formation constant
will be the product of formation constants representing
the individual interactions; thus, K
ajAA
represents the
formation constant of a dimer in which only the aj pair
of bonds is formed.
In the isologous tetrahedron (Fig. 7-5) the third set
of paired interactions cl must be taken into account.
(However, the third interaction constant will not be an
association constant of the type represented by K
ajAA
and K
bkAA
but a dimensionless number.) On the other
hand, the heterologous square has only a single inter-
action constant.
Now consider the binding of one molecule of X to
the isologous tetramer with a conformational change
in one subunit (Eq. 7-44). We see that one pair of aj
interactions and one pair of bk interactions have been
altered. The equilibrium constant for the binding of X
to the tetramer will be (Eq. 7-45) in which the 4 is a
These
subunits
face in
opposite
directions
Each interface contains
a pair of aj interactions
aj
bk
Isologous square
k
a
aj
bk
k
a
Heterologous square
Each interface contains
a pair of aj interactions
b
j
j
b
j a
j
a
a
j a j
Figure 7-22 Comparison of the interactions in isologous
(dihedral) and heterologous (cyclic) square configurations of
subunits.
G G G
K K K
aj bk
aj bk
f AA AA
f AA AA
= +
=
2 2
2 2
(7-43)
A
3
BX
X
A
4
+ X
(7-44)
K
K K
K K
K K
aj bk
aj bk
= 4
AB AB
AA AA
BX t
(7-45)
One conformational state dissociated. It may
happen that A
2
is a dimer but that B
2
dissociates into
monomers because K
BB
is very small. In such a case
binding of X leads to dissociation of the protein. A
well-known example is provided by hemoglobin of the
lamprey which is a dimer and which dissociates to a
monomer upon binding of oxygen.
117
Equation 7-11
simplifies to Eq. 7-42. The reader may wish to consider
whether the weakly cooperative binding of oxygen by
lamprey hemoglobin is predicted by this equation.
Look again at the expression for L, the constant
determining the relative amounts of a protein in con-
formations A and B in the absence of ligand. From
Eq. 7-36 we see that a large value of L (conformer A
favored) can result either because K
t
is very small or
because K
BB
<< K
AA
. Thus, if K
t
~ 1 and L is large, the
subunits must associate much more weakly in B
2
than
in A
2
and the chances are that binding of X will disso-
ciate the molecule as in the case of lamprey hemoglobin.
On the other hand, if K
t
is very small, implying that
the molecule is held in conformation A because of
some intrinsically more stable folding pattern in that
conformation, K
BB
might exceed K
AA
very much; if K
AA
were low enough A
2
could be completely dissociated.
Binding of ligand would lead to association and to
cooperative binding. This can be verified by writing
down the appropriate terms from Eq. 7-35.
3. Higher Oligomers
Mathematical treatment of binding curves for
oligomers containing more than two subunits is com-
plex, but the algebra is straightforward. A computer
can be programmed to do necessary calculations.
Avoid picking an equation from the literature and
assuming that it will be satisfactory. Consider the two
tetrameric structures in Fig. 7-22. In the isologous
square (also shown in Fig. 7-11) separate contributions
to the free energy of binding can be assigned to the
individual pairs of interactions aj and bk.
Thus, following Cornish-Bowden and Koshland
114
for assembly of the tetramer (Eq. 7-43):
K K K
K
K
K
L
= =
BX t
BB
AA
BX
2 2
2
(7-41)
2
2
1
2
Y
BX
A BX
=
+
[ ]
[ ] [ ] (7-42)
353
statistical factor arising from the fact that there are four
different ways in which to form A
3
BX. When a second
molecule of X is added three geometrical arrangements
are possible:
Each one can be formed in two ways. It is a simple
matter to write down the microscopic constants for
addition of the second molecule of X as the sum of
three terms. Because values of the constants for aj and
bk differ, it will be clear that the three ways of adding
the second molecule of X are not equally probable.
Thus, the oligomer will show preferred orders of
loading with ligand X.
Two different geometries for the heterologous
tetramer are possible in form A
2
B
2
X
2
. Again, the dif-
ferent arrangements need not be equally probable and
the relative distribution of each will be determined by
the specific values of the interaction constants. In the
heterologous tetramer A
4
only one type of an interac-
tion is present between subunits. However, as soon
as a single molecule of X is bound and one subunit of
conformation B is present, two kinds of aj interactions
exist. (One in which group a is present in conformation
A and the other in which it is present in conformation
B.) Since these interactions always occur in equal
numbers they can be lumped together.
While the foregoing may seem like an unnecessarily
long exercise, it should provide a basic approach which
can be applied to specific problems. However, remem-
ber that mathematical models require simplification.
Real proteins often have more than two stable confor-
mations.
117
The entire outside surface of a protein is
made up of potential binding sites for a number of
different molecules, both small and large. Filling of
almost any of these sites can affect the functioning of
a protein.
D. The Oxygen-Carrying Proteins
1. Myoglobin and Hemoglobin
The most studied example of a conformational
change in a multisubunit protein induced by binding
of a small molecule is provided by the cooperative
binding of oxygen to hemoglobin.
118 120
Mammalian
hemoglobin is an
2

2
tetramer of ~ 16-kDa subunits,
each containing 140 150 residues. Within each sub-
unit the peptide chain folds in a characteristic largely
-helical pattern around a single large flat iron-containing
ring structure called heme (Fig. 7-23). The folding is
essentially the same in all hemoglobins, both in the
and subunits and in the monomeric muscle oxygen
storage protein, myoglobin. Amino acid residues are
customerily designated by their position in one of the
eight helices A H. The imidazole group of histidine
F-8 is coordinated with the iron in the center of the
heme on the proximal side. The other side of the
iron atom (the distal side) is the site of binding of
a single molecule of O
2
.
Although the folding of the peptide chain is al-
most the same in both subunits, and almost identical
to that of myoglobin,
118,121,122
there are numerous
differences in the amino acid sequence. If it were not
for these differences, hemoglobin would be a highly
symmetric molecule with the bonding pattern indicated
in Fig. 7-5 with three 2-fold axes of rotation. In fact,
hemoglobin has one true axis of rotation and two
pseudo-twofold axes. There are two sets of true isolo-
gous interactions (those between the two subunits
and between the two subunits) and two pairs of
unsymmetrical interactions (between and subunits).
The nearly symmetric orientation of different portions
of the peptide backbone is clearly seen in the beautiful
drawings of Geis.
119
The contact region involved in one pair of interac-
tions in hemoglobin (
1

1
) is more extensive than the
other. There is close contact between 34 different amino
acid side chains and 110 atoms lie within 0.4 nm of
each other.
118
Hydrophobic bonding is the principal
force holding the two subunits together, and only a
few reciprocal contacts of the type found in a true isolo-
gous bond remain. The second contact designated

2
involves only 19 residues and a total of 80 atoms.
Because this interaction is weaker, hemoglobin disso-
ciates relatively easily into dimers held together
by the
1

1
contacts and motion occurs along the
1

2
contacts during oxygenation. The truly isologous
interactions (i.e., and ) are weak because the
identical protomers hardly touch each other.
The binding of oxygen. Curves of percentage
oxygenation (Y) vs the partial pressure of O
2
are given
in Fig. 7-24 and illustrate the high degree of cooperat-
ivity. Depending upon conditions, values of n
Hill
(Eq.
7-24) may be as high as 3. As a result of this cooperat-
ivity the hemoglobin, in the capillaries of the lungs at a
partial pressure of oxygen of ~ 100 mm of mercury, is
nearly saturated with oxygen. However, when the red
cells pass through the capillaries of tissues in which
oxygen is utilized the partial pressure of oxygen falls
to about 5 mm of mercury. The cooperativity means
that the oxygen is more completely unloaded than it
would be if all four heme groups acted independently.
Deoxyhemoglobin has a low affinity for O
2
, but
the observed cooperativity in binding implies that in
the fully oxygenated state the O
2
is held with a high
X binds on the back side
X
2
X
X
X
2
X X
2
X
D. The Oxygen-Carrying Proteins
354 Chapter 7. How Macromolecules Associate
affinity. The monomeric myoglobin also has a high
affinity for oxygen, as does the abnormal hemoglobin H,
which is made up of four subunits. The latter also
completely lacks cooperativity in binding.
123
These
results can be interpreted according to the MWC model
to indicate that deoxyhemoglobin exists in the T (A)
conformation, whereas oxyhemoglobin is in the R (B)
conformation. Myoglobin stays in the R conformation
in both states of oxygenation as do the separated and
chains of hemoglobin. The subunits of hemoglobin
H also appear to be frozen in the R conformation, even
though the quaternary structure is similar to that of
deoxyhemoglobin.
123,124
Oxygenation curves of hemoglobin are often fitted
with the Adair equation (Eq. 7-12). Thus, at pH 7.4
under the conditions given in Table 7-2, Imai
125
found
for the successive formation constants K
1
= 0.004, K
2
=
0.009, K
3
= 0.002 and K
4
= 0.95 in units of mm Hg
1
.
From the definition of a formation constant the oxygen
pressure PO
2
required for 50 % oxygenation in the first
step will be at PO
2
= 1/ K
f
or log PO
2
= log (1/.004) = 2.4.
This is a high oxygen pressure, far to the right side of
the oxygenation curve in Fig. 7-24A. However, log K
4
is about 0.02, well to the left on the oxygenation curve.
From these formation constants we can say that after
three of the subunits have become oxygenated the
affinity of the remaining subunit has increased about
300-fold when the concentration of the effector 2,3-
bisphosphoglycerate is present at the normal physio-
logical concentraton (see Section 4).
125 127
However, we must ask what uncertainties are
present in the data used to obtain these constants. To
extract four successive binding constants from a curve
like that in Fig. 7-24A is extremely difficult.
129,130
This
fact has encouraged the widespread use of the simpler
MWC model.
30a,127a
When the same data were treated
by Imai
131
using the MWC model it was found that L =
2.8 x 10
6
and c = K
f (T)
/ K
f (R)
= 0.0038. Changes in
Figure 7-23 Folding pattern of the hemoglobin monomers. The pattern shown is for the chain of human hemoglobin.
Some of the differences between this and the chain and myoglobin are indicated. Evolutionarily conserved residues are
indicated by boxes, highly conserved, invariant. Other markings show substitutions observed in some abnormal
human hemoglobins. Conserved residues are numbered according to their location in one of the helices A H, while mutant
hemoglobins are indicated by the position of the substitution in the entire and chain.
HC-2 Tyr
F-4 Leu
C-4 Thr
CD-1 Phe
F8 His
355
enthalpy, entropy, and Gibbs energy are given in Table 7-2. Kinetic
data
131a
as well as O
2
-binding measurements with single crystals
132
are
partially consistent with the MWC model.
133
However, the discovery
of a third quaternary structure of hemoglobin, similar to the R state but
distinct from it,
134136a
emphasizes the complexity of this allosteric
molecule.
Hemoglobin tetramers tend to stay tightly associated but some
dissociation of oxyhemoglobin into dimers does occur (K
f
= 7 x 10
5
M).
137,138
Deoxyhemoglobin is about 40,000 times more tightly associated. All of
P
e
r
c
e
n
t

o
x
y
g
e
n
a
t
i
o
n
2 1 0 1 2
log pO
2
(mm Hg)
Hemoglobin free of
biphosphoglycerate
Myoglobin
Hemoglobin in
whole blood
0 0.4 0.8
0.0
0.2
0.4
0.6
0.8
1.0
1.0
log pO
2
(mm Hg)
Y
Figure 7-24 Cooperative binding of oxygen by hemoglobins. (A) Binding
curve for myoglobin (noncooperative) and for hemoglobin in the absence
and presence (in whole blood) of 2,3-bisphosphoglycerate. Oxygen affinity
is decreased by bisphosphoglycerate. (B) Saturation curve for hemoglobin
(erythrocruorin) of Arenicola, a spiny annelid worm. The molecule contains
192 subunits and 96 hemes. It shows very strong cooperativity with n
Hill
6.
From (A) Benesch and Benesch,
123
and (B) Waxman.
128
D. The Oxygen-Carrying Proteins
B
A
the equilibria involved are strongly
affected by pH and by the presence
of salts such as NaCl.
126,139
This is
in part a reflection of the strong
role of ionic interactions in holding
together the subunits in the T state
as is discussed in the following
sections.
127
Structural changes accompa-
nying oxygen binding. Perutz and
associates, using X-ray crystallogra-
phy, found small but real differences
in the conformation of the subunits
of deoxy- and oxy- hemoglobin.
140,141
More striking is the fact that upon
oxygenation, both and subunits
undergo substantial amounts of
rotation, the net result being that the
hemes of the two subunits move
about 0.07 nm closer together in the
oxy form than in the deoxy form.
Within the
1

1
contacts (Fig. 7-25)
little change is seen. On the other
hand, contact
1

2
, the allosteric
interface, is altered drastically. As
Perutz expressed it, there is a jump
in the dovetailing of the CD region
of the subunit relative to the FG
region of the subunit. The hydro-
gen-bonding pattern is also changed.
A major difference is seen in the
hydrogen-bonded salt bridges
present at the ends of the molecules
of deoxyhemoglobin. The NH
3
+
group of Lys H-10 in each subunit
is hydrogen bonded to the carboxyl
group of the C-terminal arginine of
the opposite chain. The guani-
dinium group of each C-terminal
arginine is hydrogen bonded to the
carboxyl group of Asp H-9 in the
opposite chain. It is also hydro-
gen bonded to an inorganic anion
(phosphate or Cl

), which in turn is
hydrogen-bonded to the amino
group of Val 1 of the opposite
chain
142
forming a pair of isologous
interactions.
At the other end of the molecule,
the C-terminal group of His 146 of
each chain binds to the amino
group of Lys C-6 of the chain,
while the imidazole side chain binds
to Asp FG-1 of the same chain
(Fig. 7-25). These salt bridges
appear to provide extra stability to
T T (O
2
)
4
51 1 154 4 5.0 1.7 7.5
R R (O
2
)
4
62 2 146 6 19 2 2.0 x 10
3
T R 70 7 111 25 37 10 3.6 x 10
7
(unoxygenated)
T (O
2
)
4
R (O
2
)
4
19 2.1 x 10
3
(oxygenated)
Parameters for MWC model
L = (3.6 x 10
7
)
1
= 2.8 x 10
6
c = K
f (T)
/K
f (R)
= 0.0038
TABLE 7-2
Thermodynamic Functions for Oxygenation of Hemoglobin
a
a
From Imai, K. (1979) J. Mol. Biol. 133, 233 247. The measurements were made at pH
7.4 in the presence of 0.1 M chloride ion and 2 mM 2,3-bisphosphoglycerate to mimic
physiological conditions. The values of H, S, and G given are per mole of heme,
i.e., per monomer unit. They must be multiplied by 4 to correspond to the reactions as
shown for the tetramer.
Reaction
H
(kJ mol
-1
)
S
(JK
-1
mol
-1
)
G
(kJ mol
-1
) K
f
356 Chapter 7. How Macromolecules Associate
Binding site for 2,3-bisphosphoglycerate
in deoxyhemoglobin

1
contact changes
little upon oxygenation
But a large change
occurs in the
1

2
contact
Salt bridge between
C-terminal COO

of chain and C-6 Lys


NH
3
of chain
Upon oxygenation this
salt bridge is broken and
the proton of His HC-3
dissociates (Bohr effect)
Tyrosines HC-2 present in
pocket in deoxyhemoglobin
come out in oxyhemoglobin

E
F
H
Figure 7-25 (A) Structural changes
occurring upon oxygenation of
hemoglobin. After Dickerson
144
and
Perutz.
143
(B) Rotation at the con-
tact
1

2
causes a jump in the dove-
tailing of the CD region of relative
to the FG region of and a switch
of hydrogen bonds as shown.
143
(C) Some details of the salt bridges.
C
O
O
H
N H
H
N
N H
H
O
O
Lys 40

O H O
Asp 94
Tyr 145
Val 98
O
C
O
N
H
H
N

2
Asp 126

2
Val 1

2
Lys 127
H
+
H
H

+
N
N
N
+
H
C O
O

O H O
Val 93
Try 140
1 Arg 141
C
O
Val 34
H
H
H
Cl
H
+

+
2 FG corner
1
1 FG corner 2
His 146
B
C
A
357 D. The Oxygen-Carrying Proteins
deoxyhemoglobin and account for the high value of
the constant L. In deoxyhemoglobin the side chain of
the highly conserved Tyr HC-2 lies tucked into a pocket
between the H and F helices and is hydrogen bonded
to the main chain carbonyl of residue FG-5 (Figs. 7-23
and 7-25). Upon oxygenation this tyrosine in each
subunit is released from its pocket; the salt bridges at
the ends of the molecules are broken and the subunit
shifts into the new bonding pattern characteristic of
oxyhemoglobin.
143,144
Cooperativity in O
2
binding is
absent or greatly decreased in mutant hemoglobins
with substitutions in the residues involved in these
salt bridges
145 147
or in residues lying in the
1

2
interface.
148,149
How does the binding of O
2
to the iron of heme
trigger the conformational change in hemoglobin? An
enormous amount of effort by many people has been
expended in trying to answer this question. As is
pointed out in Chapter 16, the iron atom in deoxyhe-
moglobin lies a little outside the plane of the heme rings.
When oxygenation occurs the iron atom moves toward
the oxygen and into the plane of the heme.
150,151
This
movement probably amounts to only about 0.05 nm.
Nevertheless, this small displacement evidently induces
the other structural changes that are observed. The
iron pulls the side chain of histidine F-8 with it and
moves helix F which is also hydrogen bonded to this
imidazole ring. Because of the tight packing of the
various groups this motion cannot occur freely but is
accompanied by a movement of the F helix by 0.1 nm
across the heme plane. These movements may induce
additional structural changes in the irregularly folded
FG corners that allow the subunits to shift to the new
stable position of the R state. All four subunits appear
to change conformation together. This conformational
change must also cause the affinity for oxygen of any
unoxygenated subunits to rise dramatically, presum-
ably by shifting the iron atoms into the planes of the
heme rings. This ensures the cooperative loading of
the protein by O
2
.
While there is no doubt that the iron atom moves
upon oxygenation, it is not obvious that this will lead
to the observed cooperativity. Oxygenated heme has
some of the characteristics of an Fe(III)peroxide anion
complex.
152
The iron atom acquires an increased
positive charge upon oxygenation by donating an
electron for bond formation.
This may transmit an electronic effect through either
His F-8 or the heme ring to the nearby
1

2
interface
and also affect the subunit interactions. The reaction
of various heme proteins with oxygen is discussed
further in Chapter 16.
(7-46)
Fe
2+
(deoxy) + O
2
Fe
3+
O
2

The Bohr effect and allosteric regulators. The


breaking of the salt bridges at the ends of the hemo-
globin molecule upon oxygenation has another impor-
tant result. The pK
a
values of the N-terminal valines
of the subunits and of His HC-3 of the subunits are
abnormally high in the deoxy form because they are
tied up in the salt bridges. In the oxy form in which
the groups are free, the pK
a
values are lower. If hemo-
globin is held at a constant pH of 7, these protons disso-
ciate upon oxygenation. This Bohr effect, described
in 1904,
153 156
is important because acidification of hemo-
globin stabilizes the deoxy form. In capillaries in which
oxygen pressure is low and in which carbon dioxide
and lactic acid may have accumulated, the lowering of
the pH causes oxyhemoglobin to release oxygen more
efficiently. These effects are also strongly dependent
on the presence of chloride ions.
139,142,143,157,158
Just as the conformational equilibria in hemoglobin
can be shifted by attachment of oxygen to the heme
groups, so the binding of certain other molecules at
different sites can also affect the conformation. Such
compounds are called allosteric effectors or regulators
because they bind at a site other than the active site.
They are considered in more detail in Chapter 9. An
important allosteric effector for human hemoglobin is
2,3-bisphosphoglycerate, a compound found in
human red blood cells in a high concentration approxi-
mately equimolar with that of hemoglobin. One mole-
cule of bisphosphoglycerate binds to a hemoglobin
tetramer in the deoxy form with K
f
= 1.4 x 10
5
but
has only half this affinity for oxyhemoglobin.
159
X-ray
crystallography shows that bisphosphoglycerate binds
between the two chains of deoxyhemoglobin directly
on the twofold axis (Fig. 7-26).
159
Because of the presence
of 2,3-bisphosphoglycerate in erythrocytes the affinity
of oxygen for hemoglobin in whole blood is less than
that for isolated hemoglobin
160,161
(Fig. 7-24). This is
important because it allows a larger fraction of the
oxygen carried to be unloaded from red corpuscles in
body tissues. The bisphosphoglycerate level of red cells
varies with physiological conditions, e.g., people living
at higher elevations have a higher concentration.
161
It has been suggested that artificial manipulation of
the level of this regulatory substance in erythrocytes
may be of clinical usefulness for disorders in oxygen
transport.
P
O
O O
O
C
O
O
H
O
H
H
P
O
O
O

2,3-Bisphosphoglycerate
358 Chapter 7. How Macromolecules Associate
in the CO
2
-rich respiring tissues. Hemoglobin carries
a significant fraction of CO
2
to the lungs, and there the
oxygenation of hemoglobin facilitates the dissociation
of CO
2
from the carbamino groups. Hemoglobin is
also one of the major pH buffers of blood.
Carbon monoxide, cyanide, and nitric oxide.
A danger to hemoglobin and other heme proteins is
posed by competing ligands such as CO, CN

, and
NO. All of these are present within organisms and
both CO and NO act as hormones. Hemoglobin and
myoglobin are partially protected from carbon monox-
ide by the design of the binding site for O
2
. The distal
imidazole of histidine E7 hydrogen bonds to O
2
but
not to the nonpolar CO. The site also accommodates
the geometry of the bound O
2
better than that of CO.
165
Bound CO can be released from hemes by the action of
light. Using X-ray diffraction
166,166a
and X-ray absorp-
tion measurements
167
at cryogenic temperatures, it has
been possible to observe the motions of both the released
CO and the heme in myoglobin, motions which may
shed light on the normal oxygen transport cycle. Co-
operativity in the binding of CO to hemoglobins has
been studied extensively,
158,168,169
as has binding to
model heme compounds.
170
Cyanide ions bind most
tightly and also cooperatively
171 173
to the oxidized
Fe
3+
form, which is called methemoglobin.
Nitric oxide is a reactive, paramagnetic gaseous
free radical which is formed in the human body and
in other organisms by an enzymatic oxidation of
L-arginine (Eq. 18-65). Since about 1980, NO has been
recognized as a hormone with a broad range of effects
Not all species contain 2,3-bisphosphoglycerate
in their erythrocytes. In birds and turtles its function
appears to be served by inositol pentaphosphate. In
crocodiles the site between the two chains that binds
organic phosphates in other species has been modified
so that it binds bicarbonate ion, HCO
3

, specifically.
This ion, which accumulates in tissues as crocodiles
lie under water, acts as an allosteric regulator in these
animals.
162,163
It allows the animals to more completely
utilize the O
2
from the hemoglobin and to remain under
water longer. The Bohr effect, which was considered
in the preceding section, can be viewed as resulting
from the action of protons as allosteric effectors that
bind to the amino and imidazole groups of the salt
linkages. Carbon dioxide also acts as a physiological
effector in mammalian blood by combining reversibly
with NH
2
-terminal groups of the and subunits to
formcarbamino ( NH COO

) groups (Eq. 7-47).


119,164
It is the N-terminal amino groups rather than lysyl
side chain groups that undergo this reaction. Because
of their relatively low pK
a
values there is a significant
fraction of unprotonated NH
2
groups at the pH of
blood. The affinity for CO
2
is highest in deoxygenated
hemoglobin. Consequently, unloading of O
2
is facilitated
N
H
C
O
O

+ H
+
CO
2
NH
2
+ Protein
(7-47)
His
143
FHelix
EHelix
AHelix
Residue 6
His 2
BPG
1.0 nm
NH
3
+

Lys
82
Figure 7-26 The allosteric
effects of 2,3-bisphosphogly-
cerate (BPG) bound to the
chains of human deoxyhemo-
globin. The phosphate groups
of the BPG form salt bridges
with valines 1 and histidines 2
and 143 of both chains and
with lysine 82 of one chain.
This binding pulls the A helix
and residue 6 toward the E
helix. From Arnone.
159
Val 1
359
2
1
1
(Chapters 11, 18). It binds to the iron of heme groups
in either the Fe
2+
or Fe
3+
form and also reacts with
thiol groups of proteins and small molecules to form
S-nitrosothiols (R S N= O).
174 176
It reacts with the
heme iron of myoglobin and hemoglobin and, by
transfer of one electron, can oxidize the iron of hemo-
globin to the Fe
3+
methemoglobin with formation of
the nitroxyl ion NO

.
177,178
This reaction may be a
major cause of methemoglobin formation.
One of the major effects of NO is to induce the
relaxation of smooth muscle of blood vessels, an im-
portant factor in the regulation of blood pressure.
Hemoglobin can carry NO both on its heme and on the
thiol group of cysteine 93.
174,175
The affinity for NO is
high in the T state and low in the R state. This allows
hemoglobin to carry NO from the lungs to tissues, where
it can be released and participate in the regulation of
blood pressure.
174,179
A cytoplasmic hemoglobin of the clam Lucina
pectinata has evolved to carry oxygen to symbiotic
chemoautotrophic bacteria located within cells of
the hosts gills. It is also readily oxidized to the Fe
3+
methemoglobin form which binds sulfide ions
extremely tightly
180 182
and is thought to transport
sulfide to the bacteria.
2. Abnormal Human Hemoglobins
Many alterations in the structure of hemoglobin
have arisen by mutations in the human population.
It is estimated that one person in 20 carries a mutation
that will cause a hemoglobin disorder in a homozy-
gote.
183
There are also many unrecognized and harm-
less substitutions of one amino acid for another. How-
ever, substitutions near the heme group often adversely
affect the binding of oxygen and substitutions in the
interfaces between subunits may decrease the coop-
erative interaction between subunits.
184
One of the
most common and serious abnormal hemoglobins is
hemoglobin S, which is present in individuals suffering
from sickle cell disease (see Box 7-B). In Hb S, glutamic
acid 6 of the chain is replaced by valine. Replacement
of the same amino acid by lysine leads to Hb C
185
and
is associated with a mild disease condition . A few of
the many other substitutions that have been studied
are indicated in Fig. 7-23. The locations of the defects
in the hemoglobin structure have been established
with the aid of protein fingerprinting (Fig. 7-27).
A group of serious defects are represented by the
hemoglobins M. Only heterozygotic individuals
survive. Their blood is dark because in Hb M the iron
in half of the subunits is oxidized irreversibly to the
ferric state. The resulting methemoglobin is present in
normal blood to the extent of about 1%. While normal
methemoglobin is reduced by a methemoglobin
reductase system (Box 15-H), methemoglobins M
cannot be reduced. All of the five hemoglobins M result
from substitutions near the heme group. In four of
them, one of the heme-linked histidines (either F-8 or
E-7) of either the or the subunits is substituted by
tyrosine. In the fifth, valine 67 of the chains is substi-
tuted by glutamate. The two hemoglobins M that carry
substitutions in the subunits (M
Boston
and M
Iwate
) are
frozen in the T (deoxy) conformation and therefore have
low oxygen affinities and lack cooperativity.
D. The Oxygen-Carrying Proteins
Figure 7-27 Fingerprints of human hemoglobins. The
denatured hemoglobin was digested with trypsin and the 28
resulting peptides were separated on a sheet of paper by
electrophoresis in one direction (horizontal in the figures;
anode to the left) and by chromatography in the other direc-
tion (vertical in the figure). The peptides were visualized by
spraying with ninhydrin or with specific reagents for histi-
dine or tyrosine residues. Since trypsin cuts only next to
lysine, which occurs infrequently, the petide pattern provides
a fingerprint, characteristic for any pure protein. (A) The
fingerprint of normal adult hemoglobin A. (B) Fingerprint of
hemoglobin S (sickle cell hemoglobin). One histidine-con-
taining peptide (1) is missing and a new one (2) is present.
This altered peptide contains the first eight residues of the N-
terminal chain of the subunit of the protein. From H. Leh-
mann and R. G. Huntsman, Mans Haemoglobin.
187
B
A
360 Chapter 7. How Macromolecules Associate
BOX 7-B SICKLE CELL DISEASE, MALARIA, AND BLOOD SUBSTITUTES
Many persons, especially if they are of west
African descent, suffer from the crippling and often
lethal sickle cell disease.
a,b
In 1949, Pauling, Itano,
and associates discovered that hemoglobin from
such individuals migrated unusually rapidly upon
electrophoresis.
c
Later, Ingram devised the method
of protein fingerprinting illustrated in Fig. 7-27 and
applied it to hemoglobin.
d
He split the hemoglobin
molecule into 15 tryptic peptides which he separated
by electrophoresis and chromatography. From these
experiments the abnormality in sickle cell hemoglobin
(hemoglobin S; Hb S) was located at position 6 in
the chain (see Fig. 7-23). The glutamic acid present
in this position in hemoglobin A was replaced by
valine in Hb S. This was the first instance in which
a genetic disease was traced directly to the presence
of a single amino acid substitution in a specific
protein. The DNA of the normal gene for the globin
chain has since been sequenced and found to have
the glutamic acid codon GAG at position 6. A single
base change to GTG (see Table 5-5) causes the sickle
cell mutation. Persons homozygous for this altered
gene have sickle cell disease, while the much more
numerous heterozygotes have, at most, minor prob-
lems.
When HbS is deoxygenated it tends to crystal-
lize in red blood cells, which contain 33% by weight
hemoglobin. The crystallization (actually gel forma-
tion) distorts the cells into a sickle shape and these
distorted corpuscles are easily destroyed, leading to
anemia. The introduction of the hydrophobic valine
residue in Hb S at position 6 near the end of the
molecule helps form a new bonding domain by
which the hemoglobin tetramers associate to form
long semicrystalline microfilamentous arrays.
b,e g
Why is there such a high incidence of the sickle
cell gene, estimated to be present in three million
Americans? The occurrence and spread of the gene
in Africa was apparently the result of a balance
between its harmful effects and a beneficial effect
under circumstances existing there. The malaria
parasite, the greatest killer of all time, lives in red
blood cells during part of its life cycle (see Fig. 1-9).
Left: Normal erythrocytes, Biophoto Associates, Photo Researchers.
Right: Sickled erythrocytes, Nigel Calder.
Red cells that contain Hb S as well as
Hb A are apparently less suitable than
cells containing only Hb A for growth
of the malaria organism. Thus, het-
erozygotic carriers of the sickle cell
gene survived epidemics of malaria
but at the price of seeing one-fourth
of their offspring die of sickle cell
disease.
What is the outlook for the many
(50,000 in the United States alone)
sufferers of sickle cell disease today? Careful medi-
cal care, including blood transfusion, can prolong
life greatly
h
and intense efforts are under way to
find drugs that will prevent Hb S from crystallizing.
i
The problem arises from a hydrophobic interaction
of valine B6 with phenylalanine B85 and leucine B88
of another molecule in the filaments of Hb S. The
latter two residues are on the outside surface of
helix F (see Fig. 7-23). It is difficult to modify one
of these residues chemically but various alterations
at the nearby N-termini of the chains do inhibit
sickling. Cyanate does so by specifically carbamoy-
lating these amino groups. However, although it
was tested in humans, cyanate is too toxic for use.
j
Another approach employs an aldehyde that will
form Schiff bases (Eq. 13-4)
k
with the same amino
groups.
k,l
A third approach is to use an acylating
reagent. For example, methylacetyl phosphate
m
acetylates the same Lys 82 amino groups that react
with bisphosphoglycerate and with cyanate.
Aspirin (2-acetoxybenzoic acid) is also a mild
acetylating reagent and two-headed aspirins such
Chain C
C
O
NH
2
C
H
H
3
C
H
H
3
C
Chain C
C
O
N
C
H
H
3
C
H
H
3
C
C
O
N

Cyanate
C
NH
2
O
H
H
3
C
C
O
P
O CH
3
O O
O

361
BOX 7-B (continued)
as the following react specifically to crosslink the
hemoglobin chains.
erythrocyte the hemoglobin tetramers tend to disso-
ciate to dimers, losing cooperativity and escaping
through kidneys. Suitable crosslinking helps to
solve this problem.
x,y,z
Both and chains can be
produced from cloned genes and reassembled to
form hemoglobin.
aa,bb
This will probably allow
genetic engineering to form more stable but suitably
cooperative hemoglobins that can be used to avoid
hazards of transmission of viruses by transfusion.
These compounds bind into the bisphospho-
glycerate binding site (Fig. 7-26) and prevent the
chains from spreading apart as far as they normally
do in the deoxy (T) state. Since it is only the latter
that crystallizes, the compounds have a powerful
antisickling action.
n,o
Various other crosslinking
reagents have been developed and more than one
could be used together.
p r
New drugs that serve as
allosteric modifiers in the same fashion as bisphos-
phoglycerate may also be useful.
s
A fouth approach to treatment of sickle cell
disease is gene therapy. This might allow patients
to produce, in addition to Hb S, an engineered
hemoglobin with compensating mutations that
would mix with the Hb S and prevent gelling.
t,u,v
This is impractical at present but there is another
approach. Persons with sickle cell disease some-
times also have the disorder of hereditary persis-
tence of fetal hemoglobin. They continue to make
Hb F into adulthood. Great amelioration of sickle
cell disease is observed in patients with 20 25 % Hb
F.
t
Hydroxyurea stimulates a greater production of
Hb F and in patients with hereditary persistence of
Hb F hydroxyurea may raise its level in erythocytes
to ~ 50% of the total hemoglobin.
w
Crosslinking of the alpha chains of normal
deoxyhemoglobin through lysines 99 yields a hemo-
globin with normal oxygen-binding behavior and
an increased stability.
o
It makes a practical emer-
gency blood substitute, whereas unmodified hemo-
globin is unsatisfactory.
v
Unless encapsulated in an
a
Weatherall, D. J., Clegg, J. B., Higgs, D. R., and Wood, W. G.
(1995) in The Metabolic and Molecular Bases of Inherited Disease,
7th ed., Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S., and
Valle, D., eds), pp. 34173484, McGraw-Hill, New York
b
Harrington, D. J., Adachi, K., and Royer, W. E., Jr. (1998) J. Biol.
Chem. 273, 3269032696
c
Strasser, B. J. (1999) Science 286, 14881490
d
Ingram, V. M. (1957) Nature (London) 180, 326328
e
Cretegny, I., and Edelstein, S. J. (1993) J. Mol. Biol. 230, 733738
f
Padlan, E. A., and Love, W. E. (1985) J. Biol. Chem. 260, 82728279
g
Cao, Z., and Ferrone, F. A. (1996) J. Mol. Biol. 256, 219222
h
Acquaye, C., Wilchek, M., and Gorecki, M. (1981) Trends
Biochem. Sci. 6, 146149
i
Klotz, I. M., Haney, D. N., and King, L. C. (1981) Science 213,
724731
j
Harkness, D. R. (1976) Trends Biochem. Sci. 1, 73
k
Acharya, A. S., Sussman, L. G., and Manning, J. M. (1983) J.
Biol. Chem. 258, 22962302
l
San George, R. C., and Hoberman, H. D. (1986) J. Biol. Chem.
261, 68116821
m
Ueno, H., Pospischil, M. A., and Manning, J. M. (1989) J. Biol.
Chem. 264, 1234412351
n
Walder, J. A., Walder, R. Y., and Arnone, A. (1980) J. Mol. Biol.
141, 195216
o
Chatterjee, R., Welty, E. V., Walder, R. Y., Pruitt, S. L., Rogers, P.
H., Arnone, A., and Walder, J. A. (1986) J. Biol. Chem. 261, 9929
9937
p
Benesch, R. E., and Kwong, S. (1988) Biochem. Biophys. Res.
Commun. 156, 914
q
Kluger, R., Wodzinska, J., Jones, R. T., Head, C., Fujita, T. S., and
Shih, D. T. (1992) Biochemistry 31, 75517559
r
Jones, R. T., Shih, D. T., Fujita, T. S., Song, Y., Xiao, H., Head, C.,
and Kluger, R. (1996) J. Biol. Chem. 271, 675680
s
Abraham, D. J., Wireko, F. C., Randad, R. S., Poyart, C., Kister,
J., Bohn, B., Liard, J.-F., and Kunert, M. P. (1992) Biochemistry 31,
91419149
t
McCune, S. L., Reilly, M. P., Chomo, M. J., Asakura, T., and
Townes, T. M. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 98529856
u
Cole-Strauss, A., Yoon, K., Xiang, Y., Byrne, B. C., Rice, M. C.,
Gryn, J., Holloman, W. K., and Kmiec, E. B. (1996) Science 273,
13861388
v
May, C., Rivella, S., Callegari, J., Heller, G., Gaensler, K. M. L.,
Luzzatto, L., and Sadelain, M. (2000) Nature (London) 406, 8286
w
Eaton, W. A., and Hofrichter, J. (1995) Science 268, 11421143
x
Snyder, S. R., Welty, E. V., Walder, R. Y., Williams, L. A., and
Walder, J. A. (1987) Proc. Natl. Acad. Sci. U.S.A. 84, 72807284
y
Dick, L. A., Heibel, G., Moore, E. G., and Spiro, T. G. (1999)
Biochemistry 38, 64066410
z
Manjula, B. N., Malavalli, A., Smith, P. K., Chan, N.-L., Arnone,
A., Friedman, J. M., and Acharya, A. S. (2000) J. Biol. Chem. 275,
55275534
aa
Yamaguchi, T., Pang, J., Reddy, K. S., Witkowska, H. E., Surrey,
S., and Adachi, K. (1996) J. Biol. Chem. 271, 2667726683
bb
Jeong, S. T., Ho, N. T., Hendrich, M. P., and Ho, C. (1999)
Biochemistry 38, 1343313442
Br
Br
COO
O
C O
O
O
Br
OOC Br

Lys 82 (
1
)NH2 H
2
NLys 82(
2
)
Br
Br
COO
OH

2
Lys 82 (
1
)
N
N
Lys 82(
2
)
O
O
H
H
D. The Oxygen-Carrying Proteins
362 Chapter 7. How Macromolecules Associate
In hemoglobins Rainier and Nancy the usually
invariant C-terminal tyrosine 145 of the chains is
substituted by cysteine and by aspartate, respectively.
Oxygen affinity is high and cooperativity is lacking.
186
Hemoglobin Kansas, in which the
102
asparagine is
substituted by threonine, also lacks cooperativity and
has a very low oxygen affinity, while hemoglobin Rich-
mond, in which the same amino acid is substituted by
lysine, functions normally. In hemoglobin Creteil the
89 serine is replaced by asparagine with the result that
the adjacent C-terminal peptide carrying tyrosine 145
becomes disordered. In hemoglobin Hiroshima the
C-terminal histidine in the chain is replaced by aspar-
tic acid. This histidine is one that donates a Bohr proton
and in the mutant hemoglobin the oxygen affinity is
increased 3-fold and the Bohr effect is halved.
188
In
hemoglobin Suresnes the C-terminal arginines 141 of
the chains are replaced by histidine with loss of one
of the anion-binding sites mentioned in Section 3.
189
3. Comparative Biochemistry of Hemoglobin
Even within human beings there are several hemo-
globins. In addition to myoglobin, a brain protein neuro-
globin,
189a
and adult hemoglobin A (Hb A,
2

2
), there is
a minor hemoglobin A
2
(
2

2
). Prior to birth the blood
contains fetal hemoglobin, also called hemoglobin F
(Hb F,
2

2
). In the presence of 2,3-bisphosphoglycerate
Hb F has a 6-fold higher oxygen affinity than Hb A as
befits its role in obtaining oxygen from the mothers
blood.
190 192
Hemoglobin F disappears a few months
after birth and is replaced by Hb A. Each of the hemo-
globins differs from the others in amino acid sequence.
In other species the amino acid composition of
hemoglobins varies more, as do the interactions
between subunits. Hemoglobins and myoglobins are
found throughout the animal kingdom and even in
plants.
192a
The leghemoglobins
193195
are formed in
root nodules of legumes and are involved in nitrogen
fixation by symbiotic bacteria. Other hemoglobins
apparently function in the roots of plants.
196197
Hemoglobins or myoglobins are found in some cyano-
bacteria
198
and in many other bacteria.
197,199
The globin
fold of the polypeptide is recognizable in all of these.
200
The quaternary structure of hemoglobin also varies.
Myoglobin is a monomer, as is the leghemoglobin. Hemo-
globin of the sea lamprey dissociates to monomers upon
oxygenation.
201,201a
The clam Scapharca inaequivalvis has
a dimeric hemoglobin that binds O
2
cooperatively even
though the interactions between subunits are very differ-
ent from those in mammalian hemoglobins.
202204
Hemoglobin of the nematode Ascaris is an octamer.
205,206
It has puzzling properties, including a very high affinity
for O
2
and a slow dissociation rate. The distal His E-7
is replaced by glutamine, which has a hydrogen-
bonding ability closely similar to that of histidine.
Earthworms,
207
polychaete worms,
208,209
and
leeches
210
have enormous hemoglobin molecules
consisting of as many as 144 globin chains arranged into
12 dodecamers and held together by 3642 linker chains.
These hemoglobins are often called erythrocruorins.
In a few families of polychaetes chloroheme (Fig. 16-5)
substitutes for heme and the proteins are called
chlorocruorins.
208
What is common to all of the hemoglobins? The
same folding pattern of the peptide chain is always
present. The protein is always wrapped around the
heme group in an identical or very similar manner. In
spite of this striking conservation of overall structure,
when animal hemoglobins are compared, there are only
ten residues that are highly conserved. They are indicated
in Fig. 7-23 by the boxes. The two glycines (or alanine)
at B-6 and E-8 are conserved because the close contact
between the B and E helices does not permit a larger
side chain. Proline C-2 helps the molecule turn a corner.
Four of the other conserved residues are directly asso-
ciated with the heme group. Histidine E-7 and His F-8
are the heme-linked histidines. Tyrosine HC-2, as
previously mentioned, plays a role in the cooperativity
of oxygen binding. Only Lys H-9 is on the outside of
the molecule. The reasons for its conservation are
unclear.
211
When sequences from a broader range of
organisms were determined five residues (see Fig. 7-23)
were found to be highly conserved; only two are com-
pletely conserved. These are His F-8 and Phe CD-1,
which binds the heme noncovalently.
212
Hemoglobins
are not the only biological oxygen carriers. The two-
iron hemerythrins (Fig. 16-20) are used by a few
phyla of marine invertebrates, while the copper con-
taining hemocyanins (Chapter 16, Section D,4) are
used by many molluscs and arthropods.
E. Self-Assembly of Macromolecular
Structures
While it is easy to visualize the assembly of oligo-
meric proteins, it is not as easy to imagine how complex
objects such as eukaryotic cilia (Fig. 1-8) or the sarco-
meres of muscle (Fig. 19-6) are formed. However, study
of the assembly of bacteriophage particles and other
small biological objects has led to the concepts of self-
assembly and assembly pathways, concepts that are
now applied to every aspect of the architecture of cells.
1. Bacteriophages
A remarkable example of self-assembly is that of
the T-even phage (Box 7-C).
213 215
From genetic analysis
(Chapter 26) at least 22 genes are known to be required
for formation of the heads, 21 genes for the tails, and 7
genes for the tail fibers. Many of these genes encode
363
BOX 7-C THE T-EVEN BACTERIOPHAGES
Among the most remarkable objects made
visible by the electron microscope are the T-even
bacteriophage (T2, T4, and T6) which attack E. coli.
a e
While it is not often evident how a virus gains access
to a cell, these molecular syringes literally inject
their DNA through a hole dissolved in the cell wall
of the host bacterium. The viruses, of length ~ 200
nm and mass ~ 225 x 10
6
Da each, contain 130 x 10
6
Da of DNA in a 100 x 70 nm head of elongated
icosahedral shape. The head surface appears to
be formed from ~ 840 copies of a 45-kDa protein
known as gp23 (gene product 23; it is encoded by
gene 23; see map in Fig. 26-2). These protein mole-
cules are arranged as 140 hexamers (hexons) and
together with ~ 55 copies of protein gp24, arranged
as 11 pentamers (pentons), make up the bulk of the
shell.
f
The head also contains at least nine other
proteins, including three internal, basic proteins
that enter the bacterium along with the DNA. Addi-
tional proteins form the neck, collar, and whiskers.
The phage tail, which fastens to the collar via a
connector protein,
g
contains an internal tube with
a 2.5-nm hole, barely wide enough to accommodate
the flow of the DNA molecule into the bacterium.
The tube is made up of 144 subunits of gp19. The 8
x 10
6
-Da sheath that surrounds the tail tube is made
up of 144 subunits of gp18, each of mass 55 kDa,
arranged in the form of 24 rings of six subunits
each.
h,i
The sheath has contractile properties. After
Drawing courtesy of F. Eiserling and the
American Society for Microbiology.
a
200 nm
Hexon
Penton
Neck
Internal tube
Sheath
Baseplate
Adhesin
Tail fibers
Whiskers
Collar
Head
the virus has become properly attached to the host it
shortens from ~ 80 to ~ 30 nm, forcing the inner tube
through a hole etched in the wall of the bacterium.
At the end of the tail is a baseplate, a hexagonal
structure bearing six short pins, each a trimer of a
55 kDa zinc metalloprotein.
j
One of the ten proteins
known to be present in the baseplate is the enzyme T4
lysozyme (Chapter 12). The baseplate also contains
six molecules of the coenzyme 7,8-dihydropteroyl-
hexaglutamate (Chapter 15, Section D).
Six elongated, jointed tail fibers are attached
to the baseplate. The proximal segment of each
fiber is a trimer of the 1140-kDa protein gp34. A
globular domain attaches it to the baseplate. The
distal segment is composed of three subunits of
the 109-kDa gp37, three subunits of the 23-kDa
gp36, and a single copy of the 30-kDa gp35.
k
The
C-terminal ~140 residues of the 1026-residue gp37
are the specific adhesin that binds to a lipopolysac-
charide of E. coli type B cells or to the outer mem-
brane protein OmpC (Chapter 8).
l
Among the
smaller molecules present in the virus are the poly-
amines putrescine and spermidine (Chapter 24),
which neutralize about 30% of the basic groups of
the DNA.
How is infection by a T-even virus initiated?
Binding of the tail fibers to specific receptor sites on
the bacterial surface triggers a sequence of confor-
mational changes in the fibers, baseplate, and
Bacteriophage T4. 240,000
Micrograph courtesy of Tom Moninger.
E. Self-Assembly of Macromolecular Structures
200 nm
364 Chapter 7. How Macromolecules Associate
sequences of proteins that are incorporated into the
mature virus, but several specify enzymes needed in
the assembly process. Several mutant strains of the
viruses are able to promote synthesis of all but one of
the structural proteins of the virion. Proteins accumu-
lating within these defective bacterial hosts have no
tendency to aggregate spontaneously. However, when
the missing protein (synthesized by bacteria infected
with another strain of virus) is added complete virus
particles are formed rapidly. Investigations resulting
from this and other observations have led to the con-
clusion that during assembly each different protein is
added to the growing aggregate in a strictly specified se-
quence or assembly pathway. The addition of each protein
creates a binding site for the next protein. In some
cases the protein that binds is an enzyme that cuts off
a piece from the growing assembly of subunits and
thereby creates a site for the next protein to bind.
Before considering this complex process further,
lets look at the assembly of simpler filamentous bacte-
riophages (Fig. 7-7) and bacterial pili (Fig. 7-9). The
filamentous bacteriophages are put together from
hydrophobic protein subunits and DNA. After their
synthesis the protein subunits are stored within the
cytoplasmic membrane of the infected bacteria.
216,217
These small, largely -helical rods can easily fit within
the membrane and remain there until a DNA molecule
also enters the membrane.
218,219
Two additional pro-
teins (gene I proteins) also enter the membrane. One
has a 348-residue length, while the second is a 107-
residue protein formed by translational initiation at
a later point in the DNA sequence of the gene. These
proteins help to create an assembly site at a place where
the inner and outer membrane of the host bacterium
are close together.
220
It isnt clear how the process is
initiated, but it is likely that each subunit of the viral
coat contains a nucleotide binding site that interacts
with the DNA. Adjacent sides of the subunits are
hydrophobic and interact with other subunits to spon-
taneously coat the DNA. As the rod is assembled, the
hydrophobic groups become buried. It is postulated
that the remaining side chain groups on the outer surface
of the virus are hydrophilic and that the formation of
this hydrophilic rod provides a driving force for auto-
matic extrusion of the phage from the membrane.
216
Bacterial pili appear to be extruded in a similar
manner. They arise rapidly and may possibly be
retracted again into the bacterial membrane. The P
pilus in Fig. 7-9A is made up of subunits PapA, G, F,
E, and K which must be assembled in the correct
sequence. A chaperonin PapD is also required as is
an usher protein, PapC,
50
and also the disulfide
exchange protein DsbA (Chapter 10). DsbA helps
PapD to form the correct disulfide bridges as it folds
and PapD binds and protects the various pilus sub-
units as they accumulate in the periplasmic space of
the host. The usher protein displaces the chaperonin
PapD and escorts the subunits into the membrane
where the extrusion occurs.
50,55
Because eicosahedra are regular geometric solids
and the faces can be made up of hexons and pentons
of identical subunits, it might seem that self-assembly
of eicosahedral viruses would occur easily. However,
the subunits usually must be able to assume three or
more different conformations and the shells can easily
be assembled incorrectly. Several stategies are em-
BOX 7-C THE T-EVEN BACTERIOPHAGES (continued)
sheath. The lysozyme is released from the baseplate
and etches a hole in the bacterial cell wall. Contrac-
tion of the sheath is initiated at the baseplate and
continues to the upper end of the sheath. The tail
tube is forced into the bacterium and the DNA
rapidly flows through the narrow hole into the host
cell.
During contraction the subunits of the sheath
undergo a remarkable rearrangement into a struc-
ture containing 12 larger rings of 12 subunits each.
h
Thus, a kind of mutual intercalation of subunits
occurs. In its unidirectional and irreversible nature
the shortening of the phage tail differs from the
contraction of muscle. The protein subunits of the
sheath seem to be in an unstable high energy state
when the tail sheath of the phage is assembled. The
stored energy remains available for later contraction.
a
Mathews, C. K., Kutter, E. M., Mosig, G., and Berget, P. B., eds.
(1983) Bacteriophage T
4
, Am. Soc. Microbiology, Washington, D.C.
b
Wood, W. B., and Edgar, R. S. (1967) Sci. Am. 217(Jul), 6074
c
Mathews, C. K. (1971) Bacteriophage Biochemistry, Van Nostrand-
Reinhold, Princeton, New Jersey
d
Voyles, B. A. (1993) The Biology of Viruses, Mosby, St. Louis,
Missouri
e
Tikhonenko, A. S. (1970) Ultrastructure of Bacterial Viruses,
Plenum, New York
f
Branton, D., and Klug, A. (1975) J. Mol. Biol. 92, 559565
g
Cerritelli, M. E., and Studier, F. W. (1996) J. Mol. Biol. 258,
299307
h
Moody, M. F., and Makowski, L. (1981) J. Mol. Biol. 150,
217244
i
Mller, D. J., Engel, A., Carrascosa, J. L., and Vlez, M. (1997)
EMBO J. 16, 25472553
j
Zorzopulos, J., and Kozloff, L. M. (1978) J. Biol. Chem. 253,
55435547
k
Cerritelli, M. E., Wall, J. S., Simon, M. N., Conway, J. F., and
Steven, A. C. (1996) J. Mol. Biol. 260, 767780
l
Ttart, F., Repoila, F., Monod, C., and Krisch, H. M. (1996) J.
Mol. Biol. 258, 726 731
365 E. Self-Assembly of Macromolecular Structures
ployed to avert this problem.
221,222
Some viruses
assemble an empty shell into which the DNA flows,
but many others first form an internal scaffolding or
assembly core around which the shell is assembled.
An external scaffold may also be needed.
223
The RNA
virus MS2 forms its T = 3 capsid by using the RNA
molecule as the assembly core.
224
Other viruses may
have one or more core proteins which dissociate from
the completed shell or are removed by the action of
proteases. This is a feature of the small X phage (Fig.
7-28),
225,226,230
the tailed phages,
227,228
and double-
stranded RNA viruses including human reoviruses.
229
Bacteriophage PRD1, another virus of E. coli and
Salmonella typhimurium, has a membrane inside the
capsid apparently playing a role in assembly.
232
A general concept that seems to hold in all cases
is one of local rule. Several conformers of a virus
subunit may equilibrate within a cell. However, they
Figure 7-28 (A) Stereoscop-
ic view of the X174 114 S
mature virion viewed down a
twofold axis after a cryoelec-
tron microscopy reconstruc-
tion. From McKenna et al.
225
(B) Morphogenesis of X174
(based on a report of Hayashi
et al.
231
). Proteins A and C are
required for DNA synthesis.
Drawing from McKenna et
al.
225
Courtesy of Michael G.
Rossmann.
B
A
F
9S
B
D
6S
G
H
B
D
Stage III
DNA
synthesis
A
C
J
DNA packaging
12S
Protein
F
G
Copies/particle
5
5
Protein
F
G
H
B
D
Copies/particle
60
60
12
60 (internal scaffold)
240 (external scaffold)
108S Prohead 132S
Protein
F
G
J
H
D
Copies/particle
60
60
60
12
240
114S Mature virion
Protein
F
G
J
H
Copies/particle
60
60
60
12
F capsid protein
G capsid protein
H pilot protein
J DNA packaging protein
B scaffolding protein
D tetrameric scaffolding unit
Key
15 nm
366 Chapter 7. How Macromolecules Associate
can associate only through surfaces that are complementary.
A conformer that allows pentons to form cannot as-
semble into a hexon and only certain combinations
of other conformers can give rise to hexons, etc.
233
If
there is only one conformation and the shape is right a
T=1 shell will be formed. If there are three conformers
a T= 3 shell may arise. Another generalization is that
in most cases the procapsid or prohead that is formed
initially is fragile. Subunits may still be undergoing
conformational changes. However, a final conforma-
tional alteration, which may include chain cleavage
by a protease, usually occurs. This often expands the
overall dimensions of the capsid and creates new
intersubunit interactions which greatly strengthen the
mature capsid.
234
Scaffolding proteins are then removed
and DNA or RNA enters the capsid, again in a precise
sequence. There are many variations and the detail
that is known about virus assembly is far too great to
describe here.
Figure 7-29 illustrates the assembly pathway for
the very small X174, a T=1 virus. The major capsid
protein F is a 426-residue eight-stranded -barrel. The
175-residue G protein forms pentameric spikes while
60 copies of the internal scaffolding protein and 240
copies of the external scaffolding protein D and 12
copies of the pilot protein are required to form the
prohead. The single-stranded DNA enters along with
60 copies of a DNA packaging protein J.
Assembly of the tailed bacteriophages (Box 7-C)
is even more complex. The genome of the viruses is
large. The 166-kb circular dsDNA of phage T4 contains
~ 250 genes, many of which encode proteins of the
virion or enzymes or chaperonins needed in assembly.
The assembly pathway for the bacteriophage heads
requires at least 22 gene products.
235
Seven of these
form the assembly core which serves as a scaffolding
around which the 840 copies of gp23 and 55 copies of
gp24 (see Box 7-C) are added to give the elongated
icosahedral prohead I. Most of the internal proteins
are then dissolved by proteases, one of which is the
phage-specified gp21. A protease also cuts a piece
from each molecule of gp23 to form the smaller gp23*,
the major protein of the mature prohead II. This
cleavage also triggers the conformational change leading
to head expansion. The empty proheads are now filled
with DNA in a process which is assisted by another
series of catalytic proteins.
The T4 phage tail is assembled in a separate se-
quence. Six copies of each of three different proteins
form a hub with hexagonal symmetry (Fig. 7-29).
In another assembly sequence, seven different proteins
form wedge-shaped pieces, six of which are then joined
to the hub to form the hexagonal baseplate. Two more
proteins then add to the surface of the base plate and
activate it for the growth of the internal tail tube. Only
after assembly of the internal tube begins does the sheath
Figure 7-29 Assembly se-
quence for bacteriophage T4
with details for the tail. The
numbers refer to the genes in
the T4 chromosome map (Fig.
26-2). A P after the number
indicates that the protein gene
product is incorporated into
the phage tail. Other numbers
indicate gene products that are
thought to have essential cata-
lytic functions in the assembly
process. Adapted from King
and Mykolajewycz
236
and
Kikuchi and King.
214
5P
27P
51
28
26
29P
Hub
6 copies
9P
12P
48
54
19P
Inner
tube
19P
18P
10P
11P
7P
8P
6P
53P
25P
Wedge
Sheath
15P
Connector
Head
Collar
Whisker
2 45 nm
Sheath
Neck
110 nm
80 nm Assembly of
head and neck
Tail
fiber
156
69nm
69nm
37P
36P
34P
57
35
57.38
Tail fiber
assembly
367 E. Self-Assembly of Macromolecular Structures
EGF-like
241 244
~ 45 Ca
2+
SH2
238,245 247
~ 100 Phosphotyrosine
Structure
248 253
SH3
239,246,254
~ 60 Polyproline, PXXP
Structure
255 257
PTB
238,258,259
Proline-rich sequence
PH (plekstrin homology)
260,261
Phosphotyrosine
Structure
262,263
PDZ
264 266
80 100 C-terminal XS / TXV COO

Immunoglobulin repeat ~ 100


(Fig. 2-16)
267
Kringles, blood clotting
proteins
268 270
80-85 Calcium binding
Apple, Blood clotting
Factor X
271
90 Calcium binding
WW (TrpTrp)
272
~ 38 Proline
Serine protease
273
P (Trefoil)
274
~ 50
TPR (Tetratrico peptide repeat)
247,275,276
ZBD (Zinc-binding domains)
Zinc finger (Fig. 5-37)
277
Others
277 280
TABLE 7-3
A Few Well-Known Structural Domains
Name
Length in
amino acid residues
Specific
ligands
begin to grow, and only when both of these tubular
structures have reached the correct length, is a cap
protein placed on top. The DNA-filled head is then
attached by a special connector and only then do the
tail fibers, which have been assembled separately, join
at the opposite end.
How can each step in this complex assembly
process set the stage for the next step? Apparently the
structure of each newly synthesized protein monomer
is stable only until a specific interaction with another
protein takes place. The binding energy of this inter-
action is sufficient to induce a conformational alteration
that affects a distant part of the protein surface and
generates complementarity toward a binding site on
the next protein that is to be added. Every one of the
baseplate proteins must have such self-activating
properties! Sometimes proteolytic cleavage of a sub-
unit is required. If it occurs at an appropriate point in
the sequence it provides thermodynamic drive for the
assembly process.
The induction of a change in one protein by inter-
action with another protein is a phenomenon that is
met also in the construction of micro-
tubules, ribosomes, cilia, and myo-
fibrillar assemblies of muscle. It is
basic to the assembly of the many
labile but equally real cascade sys-
tems of proteinprotein interactions
such as that involved in the clotting
of blood (Chapter 12) and signaling
at membrane surfaces.
2. Kringles and Other
Recognition Domains and
Motifs
The assemble of either transient
or long lasting complexes of pro-
teins is often dependent upon the
presence of conserved structural
domains of 30100 residues. A
similar domain may occur in many
different proteins and often two or
more times within a single protein.
The sequences within such a domain
are homologous, allowing it to be
recognized from protein or gene
sequences alone.
237,238
Domains
are often named after the protein
in which they were first discovered.
For example, EGF-like domains
resemble the 53-residue epidermal
growth factor. SH2 and SH3 do-
mains are src-homology domains,
named after Src (c-src), the protein
encoded by the src protooncogene
(Table 11-3).
239
The SH2 and SH3 domains are found
near the N terminus of this 60-kDa protein. They are
also found in many other proteins. An adapter protein
called Grb2, important in cell signaling, consists of
nothing but one SH2 domain and two SH3 domains
(Figs. 11-13, 11-14).
240
The SH2 domains bind to phos-
photyrosyl side chains of various proteins, while SH3
domains bind to a polyproline motif. Another phos-
photyrosyl binding domain, the plekstrin homology or
PH domain, is named for the protein in which it was
discovered. Kringle and apple describe the appear-
ances of the folded proteins in those domains. Struc-
tural domains often function to hold two proteins
together or to help anchor them at a membrane surface
by binding to specific protein groups, such as phos-
photyrosyl, or calcium ions. Table 7-3 lists a few well-
known folding domains and Fig. 7-30 shows three-
dimensional structures of two of them.
Recognition domains often function transiently.
For example, SH2 domains are often found in proteins
that interact with phosphotyrosyl groups of activated
cell surface receptors. The receptors become activated
368 Chapter 7. How Macromolecules Associate
by conformational alteration resulting from the binding.
The src protein is a tyrosine kinase and, when activated,
uses ATP to phosphorylate tyrosyl groups of other
proteins, and using its SH2 domains it will bind to such
groups forming and passing an intracellular message
to them.
249
F. The Cytoskeleton
The cytoplasm of eukaryotic cells contains a com-
plex network of slender rods and filaments that serve
as a kind of internal skeleton. The properties of this
cytoskeleton affect the shape and mechanical proper-
ties of cells. For example, the cytoskeleton is responsible
Figure 7-30 (A) Stereoscopic
MolScript view showing 30 super-
posed solution structures of the
SH2 / phosphopeptide complex
from protein Shc calculated from
NMR data. The N and C termini of
the protein as well as the phospho-
tyrosine (pY) and (pY + 3) residues
of the phosphopeptide are indicat-
ed. From Zhou et al.
252
Courtesy
of Stephen W. Fesik. (B) View of
the phosphotyrosine side chain of
the peptide pYEEI bound to a
high-affinity SH2 domain from the
human src tyrosine kinase called
p56
lck
. The phosphate group forms
a series of hydrogen bonds with
groups in the protein and with
water molecules (small dots) and
an ion pair with the guanidinium
group of R134. From Tong et al.
248
Courtesy of Liang Tong. (C) Struc-
ture of kringle 2 from human tissue
plasminogen activator (see Chapter
12). From de Vos et al.
269
Courtesy
of Abraham M. de Vos.
A
B
C
369
45 nm
Linker
regions
Tails
Heads
for the biconcave disc shape of erythrocytes and for
the amebas ability to rapidly interconvert gel-like and
fluid regions of the cytoplasm.
281 283
Three principal components of the cytoskeleton
are microfilaments of ~ 6 nm diameter, microtubules
of 23 25 nm diameter, and intermediate filaments
of ~10 nm diameter. A large number of associated
proteins provide for interconnections, for assembly,
and for disassembly of the cytoskeleton. Other pro-
teins act as motors that provide motion. One of these
motors is present in myosin of muscle. This protein is
not only the motor for muscular work but also forms
thick filaments of 12 16 nm diameter, which are a
major structural component of muscle (see Fig. 19-6).
1. Intermediate Filaments
In most cells the intermediate filaments provide
the scaffolding for the cytoskeleton
284 286
They may
account for only 1% of the protein in a cell but provide
up to 85% of the protein in the tough outer layers of
skin. Intermediate filament proteins are encoded by
over 50 human genes
286
which specify proteins of
various sizes, structures, and properties. However,
all of them have central 300- to 330-residue -helical
regions through which the molecules associate in
parallel pairs to form coiled-coil rods with globular
domains at the ends (Fig. 7-31). Some of these proteins,
such as the keratin of skin, are insoluble. Others, including
the nuclear lamins (Chapter 27)
287
and vimentin,
288289a
dissociate and reform filaments reversibly.
Vimentin is found in most cells and predominates
in fibroblasts and other cells of mesenchymal origin.
Desmin (55-kDa monomer) is found in both smooth and
skeletal muscle.
289b,290,290a
In the latter, it apparently
ties the contractile myofibrils to the rest of the cyto-
skeletal network and the individual myofibrils to each
other at Z disc (see Fig. 19-6). The glial filaments
from the astroglial cells of the brain are composed
mainly of a single type of 55-kDa subunits of the
glial fibrillar acidic protein but the neurofilaments
of mammalian neurons are composed of three distinct
subunits of 68-, 150-, and 200-kDa mass.
291 293
The
larger subunits have C-terminal tails that are not
required for filament formation but which can be
phosphorylated and form bridges to neighboring
neurofilaments and other cytoskeletal components
and organelles. Keratin filaments, which eventually
nearly fill the highly differentiated epidermal cells, are
also made up of several different subunits.
294
Extensions
of the keratin chains are rich in cysteine side chains
which form disulfide crosslinkages to adjacent mole-
cules to provide a network that can be dehydrated to
form hair and the tough outer layers of skin.
286
Elastin-associated microfibrils are important constituents
of elastic tissues of blood vessels, lungs, and skin.
295
A common architecture of intermediate filaments
is a staggered head-to-tail and side-by-side association
of pairs of the coiled-coil dimers into 2- to 3-nm proto-
filaments and further association of about eight
protofilaments to form the 10-nm intermediate
filaments.
286,290,296
2. Microfilaments
The most abundant microfilaments are composed
of fibrous actin (F-actin; Fig. 7-10). The thin filaments
of F-actin are also one of the two major components
of the contractile fibers of skeletal muscle. There is
actually a group of closely related actins encoded by
a multigene family. At least four vertebrate actins are
specific to various types of muscle, while two (- and
-actins) are cytosolic.
298,299
Actins are present in all
animal cells and also in fungi and plants as part of the
cytoskeleton. The microfilaments can associate to
Figure 7-31 A model for the structure of keratin microfibrils
of intermediate filaments. (A) A coiled-coil dimer, 45-nm in
length. The helical segments of the rod domains are inter-
rupted by three linker regions. The conformations of the
head and tail domains are unknown but are thought to be
flexible. (B) Probable organization of a protofilament, in-
volving staggered antiparallel rows of dimers. From Jeffrey
A. Cohlberg
297
A B Dimer Protofilament
F. The Cytoskeleton
370 Chapter 7. How Macromolecules Associate
form larger arrays and actin often exists as thicker
cables, some of which form the stress fibers seen in
cultured cells adhering to a glass surface. In the red
blood cells the spectrinactin meshwork (Fig. 8-14),
which lies directly beneath the plasma membrane,
together with the proteins that anchor it to the mem-
brane, form the cytoskeleton.
284,300,301
Its mechanical
properties appear to be responsible for the biconcave
disc shape of the cell.
The acrosomal process of some invertebrate sperm
cells is an actin cable that sometimes forms almost
instantaneously by polymerization of the actin mono-
mers and shoots out to penetrate the outer layers of the
egg during fertilization (Chapter 32). The stereocilia,
the hairs of the hair cells in the inner ear, contain
bundles of actin filaments.
302
Motion of the stereocilia
caused by sound produces changes in the membrane
potential of the cells initiating a nerve impulse. In certain
lizards each hair cell contains about 75 stereocilia of
lengths up to 30 m and diameter 0.8 m and contain-
ing more than 3000 actin filaments in a semicrystalline
array. Microvilli (Fig. 1-6) contain longitudinal arrays
of actin filaments.
In every instance, groups of microfilaments are
held together by other proteins. Stress fibers of higher
eukaryotes contain the muscle proteins tropomyosin,
-actinin, and myosin (Chapter 19), although the
latter is usually not in fibrillar form. Filamin (250-
kDa) and a 235-kDa protein are associated with actin
in platelets.
303
The high-molecular-weight synemin
crosslinks vimentin and desmin filaments,
304
while
the smaller, highly polar filaggrin provides a matrix
around the keratin filaments in the external layers of
the skin.
305
Postsynthetic modifications of cytoskeletal micro-
filaments can also occur. For example, epidermal
keratin has been found to contain lanthionine, (-gluta-
myllysine) and lysinoalanine, both presumably arising
from crosslinkages.
306
3. Microtubules
A prominent component of cytoplasm consists of
microtubules which appear under the electron micro-
scope to have a diameter of 24 2 nm and a 13 - to 15-nm
hollow core.
307 310
However, the true diameter of a
hydrated microtubule is about 30 nm and the micro-
tubule may be further surrounded by a 520 nm low
density layer of associated proteins. Microtubules are
present in the most striking form in the flagella and cilia
of eukaryotic cells (Fig. 1-8). The stable microtubules
of cilia are integral components of the machinery caus-
ing their motion (Chapter 19). Labile microtubules,
which form and then disappear, are often found in
cytoplasm in which motion is taking place, for example, in
the pseudopodia of the ameba. The mitotic spindle
consists largely of microtubules which function in the
movement of chromosomes in a dividing cell (Box 7-D
and Chapter 26).
Microtubules in the long axons of nerve cells
function as rails for the fast transport of proteins
and other materials from the cell body down the axons.
In fact, microtubules appear to be present throughout
the cytoplasm of virtually all eukaryotic cells (Fig. 7-32)
and also in spirochetes.
311
Motion in microtubular
systems depends upon motor proteins such as kinesin,
which moves bound materials toward what is known
as the negative end of the microtubule,
312
dyneins
which move toward the positive end.
310
These motor
proteins are driven by the Gibbs energy of hydrolysis
of ATP or GTP and in this respect, as well as in some
structural details (Chapter 19), resemble the muscle
protein myosin. Dynein is present in the arms of the
microtubules of cilia (Fig. 1-8) whose motion results
from the sliding of the microtubules driven by the action
of this protein (Chapter 19).
Microtubules are assembled from ~ 55-kDa tubulins,
which are mixed dimers of subunits (450 residues)
and subunits (445 residues) with 40% sequence
identity. The dimers, whose structure is shown in
Figure 7-32 Micrograph of a mouse embryo fibroblast was
obtained using indirect immunofluorescence techniques.
313
The cells were fixed with formaldehyde, dehydrated, and
treated with antibodies (formed in a rabbit) to microtubule
protein. The cells were then treated with fluorescent goat
antibodies to rabbit -globulins (see Chapter 31) and the
photograph was taken by fluorescent light emission. Cour-
tesy of Klaus Weber.
371
BOX 7-D MITOSIS, TETRAPLOID PLANTS, AND ANTICANCER DRUGS
Microtubules in cells undergoing mitosis are
the target of several important drugs. One of these
is the alkaloid colchicine which is produced by
various members of the lily family and has been used
since ancient Egyptian times for the alleviation of
the symptoms of gout.
a,b
This compound, with its tropolone ring system, binds
specifically and tightly and prevents assembly of
microtubules, including those of the mitotic spindle.
b
Colchicine forms a complex with soluble tubulin,
c
perhaps a dimeric complex of the two subunits.
d
Dividing cells treated with colchicine appear to be
blocked at metaphase (Chapter 26) and daughter
cells with a high degree of polyploidy are formed.
This has led to the widespread use of colchicine in
inducing formation of tetraploid varieties of flower-
ing plants. Similar effects upon microtubules are
produced by the antitumor agents vincristine and
vinblastine, alkaloids formed by the common plant
Vinca (periwinkle), and also by a variety of other
drugs.
e
The more recently discovered Taxol (paclitaxel)
was extracted from the bark of the Pacific yew.
f
It
stabilizes microtubules, inhibiting their disassembly.
g
Taxol also blocks mitosis and causes the cells which
fail to complete mitosis to die. Taxol has been syn-
thesized
f
and is a promising drug that is being used
in treatment of breast, ovarian, and other cancers.
f
Binding sites for the compound have been located
in tubulin subunits (Fig. 7-33).
h,i
Attempts are
being made to develop taxoids and other drugs
more effective than taxol against cancer cells.
j,k
a
Margulis, T. N. (1974) J. Am. Chem. Soc. 96, 899 902
b
Chakrabarti, G., Sengupta, S., and Bhattacharyya, B. (1996)
J. Biol. Chem. 271, 2897 2901
c
Panda, D., Daijo, J. E., Jordan, M. A., and Wilson, L. (1995)
Biochemistry 34, 9921 9929
d
Shearwin, K. E., and Timasheff, S. N. (1994) Biochemistry 33,
894 901
e
Hastie, S. B., Williams, R. C., Jr., Puett, D., and Macdonald, T. L.
(1989) J. Biol. Chem. 264, 6682 6688
f
Nicolaou, K. C., Nantermet, P. G., Ueno, H., Guy, R. K.,
Couladouros, E. A., and Sorensen, E. J. (1995) J. Am. Chem. Soc.
117, 624 633
Laser scanning confocal micrograph of chromosomes
at metaphase. Courtesy of Tom Moninger
g
Derry, W. B., Wilson, L., and Jordan, M. A. (1995) Biochemistry
34, 2203 2211
h
Rao, S., He, L., Chakravarty, S., Ojima, I., Orr, G. A., and
Horwitz, S. B. (1999) J. Biol. Chem. 274, 3799037994
i
Makowski, L. (1995) Nature (London) 375, 361 362
j
Nicolaou, K. C., Guy, R. K., and Potier, P. (1996) Sci. Am.
274(Jun), 94 98
k
Kowalski, R. J., Giannakakou, P., and Hamel, E. (1997) J. Biol.
Chem. 272, 2534 2541
l
Kwa, M. S. G., Veenstra, J. G., Van Dijk, M., and Roos, M. H.
(1995) J. Mol. Biol. 246, 500 510
Another group of drugs that bind to microtu-
bules are benzimidazole and related compounds.
These have been used widely to treat infection by
parasitic nematodes in both humans and animals.
Unfortunately resistance has developed rapidly.
In a nematode that infects sheep a single tyrosine
to phenylalanine mutation at position 200 in the
-tubulin subunit confers resistance.
l
O
O
O
N
H
C
O
CH
3
H
O
O
Colchicine
H
3
C
H
3
C
H
3
C
CH
3
OH
O
O
O
O
CH
3
O
H
CH
3
OH O O
H
3
C
O
H
3
C
O
CH
3
CH
3 O
OH
NH
O
Taxol (paclitaxel)
F. The Cytoskeleton
372 Chapter 7. How Macromolecules Associate
Fig. 7-33
314 316
are thought to be packed into an imper-
fect helix as indicated in Fig. 7-34. The structure can
also be regarded as an array of longitudinal protofila-
ments. Naturally formed microtubules usually have
precisely 13 protofilaments and a discontinuity in the
helical stacking of subunits as shown in Fig. 7-34. When
grown in a laboratory the microtubules usually have
14 protofilaments
317
and rarely 10 or 16 protofilaments
with regular helical packing.
318
Microtubules of some
moths and also of male germ cells of Drosophila have
16-protofilament microtubules without a discontinuity,
an architecture that is specified by the geometry of a
specific -tubulin isoform.
319
Each tubulin dimer binds one molecule of GTP
strongly in the subunit and a second molecule of
GTP or GDP more loosely in the subunit. In this
respect, tubulin resembles actin, whose subunits are
about the same size. However, there is little sequence
similarity. Labile microtubules of cytoplasm can be
formed or disassembled very rapidly. GTP is essential
for the fast growth of these microtubules and is hydro-
lyzed to GDP in the process.
320
However, nonhydro-
lyzable analogs of GTP, such as the one containing the
linkage P CH
2
P between the terminal and central
phosphorus atoms of the GTP, also support polymer-
ization.
321
Since microtubules have a distinct polarity,
the two ends have different tubulin surfaces exposed,
and polymerization and depolymerization can occur
at different rates at the two ends. As a consequence,
microtubules often grow at one end and disassemble
at the other. Such treadmilling may be important in
movement of chromosomes in neuronal migration
322
and in fast axonal transport of macromolecules (Chapter
30).
323
During mitosis the minus ends of the microtu-
bules are believed to be tightly anchored at the centro-
some while subunit exchanges can occur at the plus
ends
323,324
where the subunits are exposed. Using a
phage display system (see Fig. 3-16) it could be shown
that the N termini of the subunits are exposed at
the minus ends.
325
Kinesin can bind to the sub-
units all along the microtubule.
326
Microtubules are
formed by growth from microtubule nucleation sites
in microtubule organizing centers found in centro-
somes, spindle poles, and other locations.
327
Several
proteins, including -tubulin, are required.
317,328,329
A
proposed assembly pathway is illustrated in Fig. 7-34.
Isolated microtubules always contain small amounts
of larger ~ 300-kDa microtubule-associated proteins
(MAPS).
330
These elongated molecules may in part lie
in the grooves between the tubulin subunits and in part
be extended outward to form a low-density layer around
the tubule.
283,309
Nerve cells that contain stable microtu-
bules have associated stabilizing proteins.
331
A family
of proteins formed by differential splicing of mRNA are
known as tau. The tau proteins are prominent compo-
nents of the cytoskeleton of neurons. They not only
interact with microtubules but also undergo reversible
phosphorylation. Hyperphosphorylated tau is the
primary component of the paired helical filaments found
in the brains of persons with Alzheimer disease.
330
Figure 7-33 Stereoscopic
ribbon diagram of the tubulin
dimer with -tubulin with
bound GTP at the top and
-tubulin with bound GDP
at the bottom. The -tubulin
subunit also contains a bound
molecule of taxotere (see Box
7-D) which is labeled TAX.
This model is based upon
electron crystallography of
zinc-induced tubulin sheets
at 0.37-nm resolution and is
thought to approximate
closely the packing of the
tubulin monomers in microtu-
bules.
315
The arrow at the left
points toward the plus end of
the microtubule. Courtesy of
Kenneth H. Downing.
373 F. The Cytoskeleton
Figure 7-34 Growth of a microtubule from a -tubulin ring
complex (TURC). The helical -tubulin rings are formed in
the microtubule organizing centers which, in animal cells,
are the centrosomes. Thirteen -tubulin subunits are shown
in a hypothetical array formed together with a base of other
molecules of unknown structure. The microtubule grows by
addition of successive layers of /-tubulin dimers, each a
split ring of 13 dimers with the -tubulin subunits toward
the base, the negative end, and the -tubulin subunits
toward the growing positive end. After Zheng et al.
317
TuRC
13
Tubulin dimer with 2
molecules of bound GTP
13 -Tubulin
n
44a. Eisenstein, M., Shariv, I., Koren, G., Friesem,
A. A., and Katchalski-Katzir, E. (1997) J. Mol.
Biol. 266, 135143
45. Raghavendra, K., Kelly, J. A., Khairallah, L.,
and Schuster, T. M. (1988) Biochemistry 27,
75837588
46. Wang, H., and Stubbs, G. (1994) J. Mol. Biol.
239, 371384
47. Namba, K., Casper, D. L. D., and Stubbs, G. J.
(1985) Science 227, 773776
48. Eisenstein, B. I. (1987) Escherichia coli and
Salmonella typhimurium, Am. Soc.
Microbiology, Washington, D.C., FC
Niedhardt, ed. (pp. 8490)
49. Bullitt, E., and Makowski, L. (1995) Nature
(London) 373, 164167
50. Bullitt, E., Jones, C. H., Striker, R., Soto, G.,
Jacob-Dubuisson, F., Pinkner, J., Wick, M. J.,
Makowski, L., and Hultgren, S. J. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1289012895
51. Bullitt, E., and Makowski, L. (1998) Biophys. J.
74, 623632
52. Gong, M., and Makowski, L. (1992) J. Mol.
Biol. 228, 735742
53. Kuehn, M. J., Ogg, D. J., Kihlberg, J., Slonim,
L. N., Flemmer, K., Bergfors, T., and Hultgren,
S. J. (1993) Science 262, 12341241
53a. Sauer, F. G., Ftterer, K., Pinkner, J. S.,
Dodson, K. W., Hultgren, S. J., and Waksman,
G. (1999) Science 285, 10581061
53b. Choudhury, D., Thompson, A., Stojanoff, V.,
Langermann, S., Pinkner, J., Hultgren, S. J.,
and Knight, S. D. (1999) Science 285, 1061
1065
53c. Skerker, J. M., and Shapiro, L. (2000) EMBO J.
19, 32233234
53d. Hazes, B., Sastry, P. A., Hayakawa, K., Read,
R. J., and Irvin, R. T. (2000) J. Mol. Biol. 299,
10051017
53e. Collinson, S. K., Parker, J. M. R., Hodges, R. S.,
and Kay, W. W. (1999) J. Mol. Biol. 290, 741756
53f. Murphy, F. V., IV, Sweet, R. M., and Churchill,
M. E. A. (1999) EMBO J. 18, 66106618
54. Hanson, M. S., and Brinton, C. C., Jr. (1988)
Nature (London) 332, 265268
55. St. Geme, J. W., III, Pinkner, J. S., Krasan, G. P.,
Heuser, J., and Bullitt, E. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 1191311918
56. Parge, H. E., Bernstein, S. L., Deal, C. D.,
McRee, D. E., Christensen, D., Capozza, M.
A., Kays, B. W., Fieser, T. M., Draper, D., So,
M., Getzoff, E. D., and Tainer, J. A. (1990) J.
Biol. Chem. 265, 22782285
1. Williams, D. H., Searle, M. S., Mackay, J. P.,
Gerhard, U., and Maplestone, R. A. (1993)
Proc. Natl. Acad. Sci. U.S.A. 90, 11721178
2. Weber, G. (1992) Protein Interactions, Chapman
and Hall, London
3. Scatchard, G. (1949) Ann. N.Y. Acad. Sci. 51,
660672
4. Zierler, K. (1989) Trends Biochem. Sci. 14, 314317
5. Munson, P. J., Rodbard, D., and Klotz, I. M.
(1983) Science 220, 979981
6. Deranleau, D. A. (1969) J. Am. Chem. Soc. 91,
40444049
7. Dowd, J. E., and Riggs, D. S. (1965) J. Biol.
Chem. 240, 863869
8. Feldman, H. A. (1983) J. Biol. Chem. 258,
1286512867
8a. Dunford, H. B. (1984) J. Chem. Educ. 61, 129132
8b. Klotz, I. M. (1997) Ligand-Receptor Energetics,
Wiley, New York
9. Conners, K. A. (1987) Binding Constants,
Wiley, New York
10. Motulsky, H. J., and Ransnas, L. A. (1987)
FASEB J. 1, 365374
11. Schejter, A., and Margoliash, E. (1985) Trends
Biochem. Sci. 10, 490492
12. Steinhardt, J., and Reynolds, J. A. (1969)
Multiple Equilibria in Proteins, Academic Press,
New York
13. Koshland, D. E., Jr. (1970) in The Enzymes, 3rd
ed., Vol. 1 (Boyer, P. D., ed), pp. 341396,
Academic Press, New York
14. Tanford, C. (1961) Physical Chemistry of
Macromolecules, Wiley, New York (pp. 533534)
15. Edsall, J. T., and Wyman, J. (1958) Biophysical
Chemistry, Academic Press, New York
16. Westheimer, F. H., and Shookhoff, M. W.
(1939) J. Am. Chem. Soc. 61, 555560
17. Honig, B., and Nicholls, A. (1995) Science 268,
11441149
18. Rajasekaran, E., Jayaram, B., and Honig, B.
(1994) J. Am. Chem. Soc. 116, 82388240
19. Tanford, C. (1961) Physical Chemistry of
Macromolecules, Wiley, New York (Chapter 8)
20. Maier, G. D., and Metzler, D. E. (1957) J. Am.
Chem. Soc. 79, 43864391
21. Metzler, D. E. (1960) in The Enzymes, 2nd ed.,
Vol. 2 (Boyer, P. D., Lardy, H., and Myrback,
K., eds), pp. 295337, Academic Press, New
York
22. Applequist, J. (1977) J. Chem. Educ. 54, 417419
23. Hill, T. (1962) Introduction to Statistical
Thermodynamics, AddisonWesley, Reading,
Massachusetts (pp. 235241)
24. Weiss, J. N. (1997) FASEB J. 11, 835841
25. Wang, X.-G., and Engel, P. C. (1995)
Biochemistry 34, 1141711422
26. Damle, V. N. (1972) Biopolymers 11, 1789816
27. Watson, J. D. (1976) Molecular Biology of the
Gene, 3rd ed., Benjamin, Menlo Park,
California (p. 98)
28. Bernal, J. D. (1967) J. Mol. Biol. 24, 379390
29. Blundell, T. L., and Johnson, L. N. (1976) Protein
Crystallography, Academic Press, New York
30. Monod, J., Wyman, J., and Changeux, J. D.
(1965) J. Mol. Biol. 12, 88118
30a. Dill, K. A. (1997) J. Biol. Chem. 272, 701704
30b. Boresch, S., and Karplus, M. (1995) J. Mol.
Biol. 254, 801807
30c. Tamura, A., and Privalov, P. L. (1997) J. Mol.
Biol. 273, 10481060
31. Zhang, R.-G., Westbrook, M. L., Westbrook, E.
M., Scott, D. L., Otwinowski, Z., Maulik, P. R.,
Reed, R. A., and Shipley, G. G. (1995) J. Mol.
Biol. 251, 550562
31a. Marvin, D. A. (1998) Current Opinion in
Structural Biology 8, 150158
32. Marvin, D. A., Hale, R. D., Nave, C., and
Citterich, M. H. (1994) J. Mol. Biol. 235, 260286
33. Glucksman, M. J., Bhattacharjee, S., and
Makowski, L. (1992) J. Mol. Biol. 226, 455470
34. Day, L. A., Marzec, C. J., Reisberg, S. A., and
Casadevall, A. (1988) Ann. Rev. Biophys.
Biophys. Chem. 17, 509539
35. Williams, K. A., Glibowicka, M., Li, Z., Li, H.,
Khan, A. R., Chen, Y. M. Y., Wang, J., Marvin,
D. A., and Deber, C. M. (1995) J. Mol. Biol. 252,
614
36. Overman, S. A., Tsuboi, M., and Thomas, G.
J., Jr. (1996) J. Mol. Biol. 259, 331336
37. Makowski, L. (1992) J. Mol. Biol. 228, 885892
38. Rakonjac, J., and Model, P. (1998) J. Mol. Biol.
282, 2541
38a. Welsh, L. C., Marvin, D. A., and Perham, R.
N. (1998) J. Mol. Biol. 284, 12651271
39. Liu, D. J., and Day, L. A. (1994) Science 265,
671674
40. Bhyravbhatla, B., Watowich, S. J., and Caspar,
D. L. D. (1998) Biophys. J. 74, 604615
41. Fraenkel-Conrat, H. (1964) Sci. Am. 211(Oct),
4754
42. Butler, P. J. G., and Klug, A. (1978) Sci. Am.
239(Nov), 6269
43. Bloomer, A. C., Champness, J. N., Bricogne,
G., Staden, R., and Klug, A. (1978) Nature
(London) 276, 362368
44. Holmes, K. C. (1980) Trends Biochem. Sci. 5, 47
References
374 Chapter 7. How Macromolecules Associate
References
57. Parge, H. E., Forest, K. T., Hickey, M. J.,
Christensen, D. A., Getzoff, E. D., and Tainer,
J. A. (1995) Nature (London) 378, 3238
58. Folkhard, W., Leonard, K. R., Malsey, S.,
Marvin, D. A., Dubochet, J., Engel, A.,
Achtman, M., and Helmuth, R. (1979)
J. Mol. Biol. 130, 145160
59. Paiva, W. D., Grossman, T., and Silverman, P.
M. (1992) J. Biol. Chem. 267, 2619126197
60. Rubenstein, P. A. (1990) BioEssays 12, 309315
61. Venkatesh, B., Tay, B. H., Elgar, G., and
Brenner, S. (1996) J. Mol. Biol. 259, 655665
62. Holmes, K. C., Popp, D., Gebhard, W., and
Kabsch, W. (1990) Nature (London) 347, 4449
63. Lorenz, M., Poole, K. J. V., Popp, D.,
Rosenbaum, G., and Holmes, K. C. (1995) J.
Mol. Biol. 246, 108119
64. Valentine, R. C. (1969) in Symmetry and
Function of the Biological Systems at the
Macromolecular Level (Engstrom, A., and
Strandberg, B., eds), p. 165, Wiley, New York
(11th Nobel Symp.)
65. Klotz, I. M., Darnall, D. W., and Langerman,
N. R. (1975) in The Proteins, 3rd ed., Vol. 1
(Neurath, H., and Hill, R. L., eds), p. 293,
Academic Press, New York
66. Momany, C., Ernst, S., Ghosh, R., Chang, N.-
L., and Hackert, M. L. (1995) J. Mol. Biol. 252,
643655
67. Almassy, R. J., Janson, C. A., Hamlin, R.,
Xuong, N.-H., and Eisenberg, D. (1986) Nature
(London) 323, 304309
68. Harrison, S. C. (1984) Trends Biochem. Sci. 9,
345351
69. Rossmann, M. G., and Johnson, J. E. (1989)
Ann. Rev. Biochem. 58, 533573
70. Johnson, J. E., and Speir, J. A. (1997) J. Mol.
Biol. 269, 665675
71. Watson, J. D. (1976) Molecular Biology of the
Gene, 3rd ed., Benjamin, Menlo Park,
California (p. 107)
72. Unge, T., Liljas, L., Strandberg, B., Vaara, I.,
Kannan, K. K., Fridborg, K., Nordman, C. E.,
and Lentz, P. J., Jr. (1980) Nature (London) 285,
373377
73. Kpper, H., Keller, W., Kurz, C., Forss, S.,
Schaller, H., Franze, R., Stohmaier, K.,
Marquardt, O., Zaslavsky, V. G., and
Hofschneider, P. H. (1981) Nature (London)
289, 555559
74. Trikha, J., Theil, E. C., and Allewell, N. M.
(1995) J. Mol. Biol. 248, 949967
75. Larson, S. B., Day, J., Greenwood, A., and
McPherson, A. (1998) J. Mol. Biol. 277, 3759
76. Hogle, J. M., Chow, M., and Filman, D. J.
(1985) Science 229, 13581365
77. Hogle, J. M., Chow, M., and Filman, D. J.
(1987) Sci. Am. 256 (Mar), 4249
78. Olson, N. H., Kolatkar, P. R., Oliveira, M. A.,
Cheng, R. H., Greve, J. M., McClelland, A.,
Baker, T. S., and Rossmann, M. G. (1993) Proc.
Natl. Acad. Sci. U.S.A. 90, 507511
79. Hadfield, A. T., Oliveira, M. A., Kim, K. H.,
Minor, I., Kremer, M. J., Heinz, B. A., Shepard,
D., Pevear, D. C., Rueckert, R. R., and Rossmann,
M. G. (1995) J. Mol. Biol. 253, 6173
80. Wu, H., and Rossmann, M. G. (1993) J. Mol.
Biol. 233, 231244
81. Luo, M., Vriend, G., Kamer, G., Minor, I.,
Arnold, E., Rossmann, M. G., Boege, U.,
Scraba, D. G., Duke, G. M., and Palmenberg,
A. C. (1987) Science 235, 182191
82. Tikhonenko, A. S. (1970) Ultrastructure of
Bacterial Viruses, Plenum, New York
83. Birktoft, J. J., Rhodes, G., and Banaszak, L. J.
(1989) Biochemistry 28, 60656081
84. Skarzynski, T., Moody, P. C. E., and Wonacott,
A. J. (1987) J. Mol. Biol. 193, 171187
85. Zimmerle, C. T., and Alter, G. M. (1993)
Biochemistry 32, 1274312748
86. Hodgkin, D. C. (1975) Nature (London) 255, 103
87. Whittingham, J. L., Chaudhuri, S., Dodson, E.
J., Moody, P. C. E., and Dodson, G. G. (1995)
Biochemistry 34, 1555315563
88. Chang, X., Jorgensen, A. M. M., Bardrum, P.,
and Led, J. J. (1997) Biochemistry 36, 94099422
89. Golmohammadi, R., Valegrd, K., Fridborg, K.,
and Liljas, L. (1993) J. Mol. Biol. 234, 620639
90. Liljas, L., Fridborg, K., Valegrd, K., Bundule,
M., and Pumpens, P. (1994) J. Mol. Biol. 244,
279290
91. Tars, K., Bundule, M., Fridborg, K., and Lijas,
L. (1997) J. Mol. Biol. 271, 759773
92. Abad-Zapatero, C., Abdel-Meguid, S. S.,
Johnson, J. E., Leslie, A. G. W., Rayment, I.,
Rossmann, M. G., Suck, D., and Tsukihara, T.
(1980) Nature (London) 286, 3339
93. Athappilly, F. K., Murali, R., Rux, J. J., Cai, Z.,
and Burnett, R. M. (1994) J. Mol. Biol. 242,
430455
94. Stewart, P. L., Chiu, C. Y., Huang, S., Muir, T.,
Zhao, Y., Chait, B., Mathias, P., and Nemerow,
G. R. (1997) EMBO J. 16, 11891198
95. Caspar, D. L. D., and Klug, A. (1962) Cold
Spring Harbor Symposia on Quantitative Biology
27, 124
96. Johnson, J. E. (1996) Proc. Natl. Acad. Sci. U.S.A.
93, 2733
97. Steven, A. C., Trus, B. L., Booy, F. P., Cheng,
N., Zlotnick, A., Caston, J. R., and Conway, J.
F. (1997) FASEB J. 11, 733742
98. Rayment, I., Baker, T. S., Caspar, D. L. D., and
Murakami, W. T. (1982) Nature (London) 295,
110115
99. Liddington, R. C., Yan, Y., Moulai, J., Sahli, R.,
Benjamin, T. L., and Harrison, S. C. (1991)
Nature (London) 354, 278284
100. Champness, J. N., Bloomer, A. C., Bricogne,
G., Butler, P. J. G., and Klug, A. (1976) Nature
(London) 259, 2024
101. Fukuda, M., and Okada, Y. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 40354038
102. McDonald, R. C., Engelman, D. M., and Steita,
T. A. (1979) J. Biol. Chem. 254, 29422943
103. Galloway, J. (1984) Trends Biochem. Sci. 9, 233
238
104. Kantrowitz, E. R., and Lipscomb, W. N. (1990)
Trends Biochem. Sci. 15, 5359
105. Zhou, B.-B., and Schachman, H. K. (1993)
Protein Sci. 2, 103112
106. Xi, X. G., De Staercke, C., Van Vliet, F.,
Triniolles, F., Jacobs, A., Stas, P. P., Ladjimi, M.
M., Simon, V., Cunin, R., and Herv, G. (1994)
J. Mol. Biol. 242, 139149
107. Fetler, L., Vachette, P., Herv, G., and Ladjimi,
M. M. (1995) Biochemistry 34, 1565415660
108. Honzatko, R. B., Crawford, J. L., Monaco, H.
L., Ladner, J. E., Edwards, B. F. D., Evans, D.
R., Warren, S. G., Wiley, D. C., Ladner, R. C.,
and Lipscomb, W. N. (1982) J. Mol. Biol. 160,
219263
109. Krause, K. L., Volz, K. W., and Lipscomb,
W. N. (1985) Proc. Natl. Acad. Sci. U.S.A. 82,
16431647
110. Thomas, A., Field, M. J., and Perahia, D.
(1996) J. Mol. Biol. 261, 490506
110a. Brunori, M. (1999) Trends Biochem. Sci. 24,
158161
110b. Di Cera, E., ed. (1998) Advances in Protein
Chemistry, Linkage Thermodynamics of
Macromolecular Interactions, Vol. 51, Academic
Press, San Diego, California
111. Bloom, C. R., Choi, W. E., Brzovic, P. S., Ha, J.
J., Huang, S.-T., Kaarsholm, N. C., and Dunn,
M. F. (1995) J. Mol. Biol. 245, 324330
112. Hammes, G. G., and Wu, C.-W. (1971) Science
172, 12051211
113. Koshland, D. E., Jr., Nmethy, G., and Filmer,
D. (1966) Biochemistry 5, 365385
114. Cornish-Bowden, A. J., and Koshland, D. E.,
Jr. (1970) J. Biol. Chem. 245, 62416250
115. Cornish-Bowden, A. J., and Koshland, D. E.,
Jr. (1971) J. Biol. Chem. 246, 30923102
116. Darnall, D. W., and Klotz, I. M. (1972) Arch.
Biochem. Biophys. 149, 114
117. Doci, Y., Sugita, Y., and Yoneyama, Y. (1973) J.
Biol. Chem. 248, 23542363
118. Perutz, M. F., Muirhead, H., Cox, J. M., and
Goaman, L. C. G. (1968) Nature (London) 219,
131139
119. Dickerson, R. E., and Geis, I. (1983)
Hemoglobin: Structure, Function, Evolution and
Pathology, Benjamin / Cummings Publ.,
Redwood City, California
120. Antonini, E., and Brunori, M. (1971)
Hemoglobin and Myoglobin in Their Reactions
with Ligands, North-Hollard, Publ.,
Amsterdam
121. Kendrew, J. C. (1961) Sci. Am. 205(Dec), 96110
122. Fermi, G., Perutz, M. F., Shaanan, B., and
Fourme, R. (1984) J. Mol. Biol. 175, 159174
123. Benesch, R., and Benesch, R. E. (1974) Science
185, 905908
124. Borgstahl, G. E. O., Rogers, P. H., and Arnone,
A. (1994) J. Mol. Biol. 236, 831843
125. Imai, K. (1979) J. Mol. Biol. 133, 233247
126. Amiconi, G., Antonini, E., Brunori, M.,
Wyman, J., and Zolla, L. (1981) J. Mol. Biol.
152, 111129
127. Flanagan, M. A., Ackers, G. K., Matthew, J. B.,
Hanania, G. I. H., and Gurd, F. R. N. (1981)
Biochemistry 20, 74397449
127a. Shibayama, N. (1999) J. Mol. Biol. 285, 1383
1388
128. Waxman, L. (1971) J. Biol. Chem. 246, 73187327
129. Doyle, M. L., Di Cera, E., and Gill, S. J. (1988)
Biochemistry 27, 820824
130. Edelstein, S. J. (1996) J. Mol. Biol. 257, 737744
131. Imai, K. (1983) J. Mol. Biol. 167, 741749
131a. Gibson, Q. H. (1999) Biochemistry 38, 5191
5199
132. Paoli, M., Liddington, R., Tame, J., Wilkinson,
A., and Dodson, G. (1996) J. Mol. Biol. 256,
775792
133. Henry, E. R., Jones, C. M., Hofrichter, J., and
Eaton, W. A. (1997) Biochemistry 36, 65116528
134. Silva, M. M., Rogers, P. H., and Arnone, A.
(1992) J. Biol. Chem. 267, 1724817256
135. Jayaraman, V., and Spiro, T. G. (1995)
Biochemistry 34, 45114515
136. Schumacher, M. A., Zheleznova, E. E.,
Poundstone, K. S., Kluger, R., Jones, R. T., and
Brennan, R. G. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 78417844
136a. Tame, J. R. H. (1999) Trends Biochem. Sci. 24,
372377
137. Manning, L. R., Jenkins, W. T., Hess, J. R.,
Vandegriff, K., Winslow, R. M., and Manning,
J. M. (1996) Protein Sci. 5, 775781
138. Holt, J. M., and Ackers, G. K. (1995) FASEB J.
9, 210218
139. Perutz, M. F., Shih, D. T.-b, and Williamson,
D. (1994) J. Mol. Biol. 239, 555560
140. Perutz, M. F., Muirhead, H., Cox, J. M.,
Goaman, L. C. G., Mathews, F. S., McGandy,
L. E., and Webb, L. E. (1968) Nature (London)
219, 2932
141. Perutz, M. (1990) Mechanisms of Cooperativity
and Allosteric Regulation in Proteins,
Cambridge Univ. Press, London
142. ODonnell, S., Mandaro, R., Schuster, T. M.,
and Arnone, A. (1979) J. Biol. Chem. 254,
1220412208
143. Perutz, M. F. (1970) Nature (London) 228, 726739
144. Dickerson, R. E. (1972) Ann. Rev. Biochem. 41,
815842
375
145. Ishimori, K., Hashimoto, M., Imai, K.,
Fushitani, K., Miyazaki, G., Morimoto, H.,
Wada, Y., and Morishima, I. (1994) Biochemis-
try 33, 25462553
146. Chao, H., Snnichsen, F. D., DeLuca, C. I.,
Sykes, B. D., and Davies, P. L. (1994) Protein
Sci. 3, 17601769
147. Kim, H.-W., Shen, T.-J., Ho, N. T., Zou, M.,
Tam, M. F., and Ho, C. (1996) Biochemistry 35,
66206627
148. Kavanaugh, J. S., Weydert, J. A., Rogers, P. H.,
and Arnone, A. (1998) Biochemistry 37, 4358
4373
149. Kiger, L., Klinger, A. L., Kwiatkowski, L. D.,
De Young, A., Doyle, M. L., Holt, J. M., Noble,
R. W., and Ackers, G. K. (1998) Biochemistry
37, 43364345
150. Hoard, J. L., and Scheidt, W. R. (1973) Proc.
Natl. Acad. Sci. U.S.A. 70, 39193922
151. Fermi, G., Perutz, M. F., and Shulman, R. G.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 61676168
152. De Baere, I., Perutz, M. F., Kiger, L., Marden,
M. C., and Poyart, C. (1994) Proc. Natl. Acad.
Sci. U.S.A. 91, 15941597
153. Bohr, C., Hasselbalch, K., and Krogh, A.
(1904) Skand. Arch. Physiol. 16, 402
154. Wyman, J., Jr. (1948) Adv. Prot. Chem. 4, 407531
155. Benesch, R. E., and Benesch, R. (1974) Adv.
Prot. Chem. 28, 211237
156. Fang, T.-Y., Zou, M., Simplaceanu, V., Ho, N.
T., and Ho, C. (1999) Biochemistry 38, 13423
13432
157. Fronticelli, C., Pechik, I., Brinigar, W. S.,
Kowalczyk, J., and Gilliland, G. L. (1994) J.
Biol. Chem. 269, 2396523969
158. Perrella, M., Ripamonti, M., and Caccia, S.
(1998) Biochemistry 37, 20172028
159. Arnone, A. (1972) Nature (London) 237, 146149
160. Benesch, R., and Benesch, R. E. (1969) Nature
(London) 221, 618622
161. Brewer, G. J., and Eaton, J. W. (1971) Science
171, 12051211
162. Perutz, M. F., Bauer, C., Gros, G., Leclercq, F.,
Vandecasserie, C., Schnek, A. G., Braunitzer,
G., Friday, A. E., and Joysey, K. A. (1981)
Nature (London) 291, 682684
163. Komiyama, N. H., Miyazaki, G., Tame, J., and
Nagai, K. (1995) Nature (London) 373, 244246
164. Fantl, W. J., Di Donato, A., Manning, J. M.,
Rogers, P. H., and Arnone, A. (1987) J. Biol.
Chem. 87, 1270012713
165. Sage, J. T., and Jee, W. (1997) J. Mol. Biol. 274,
2126
166. Teng, T.-Y., Srajer, V., and Moffat, K. (1997)
Biochemistry 36, 1208712100
166a. Ostermann, A., Waschipky, R., Parak, F. G.,
and Nienhaus, G. U. (2000) Nature (London)
404, 205208
167. Chance, M. R., Miller, L. M., Fischetti, R. F.,
Scheuring, E., Huang, W.-X., Sclavi, B., Hai,
Y., and Sullivan, M. (1996) Biochemistry 35,
90149023
168. sapay, K., Theriault, Y., Wright, P. E., and
Case, D. A. (1994) J. Mol. Biol. 244, 183197
169. Martin, K. D., and Parkhurst, L. J. (1990)
Biochemistry 29, 57185726
170. Tetreau, C., Lavalette, D., Momenteau, M.,
Fischer, J., and Weiss, R. (1994) J. Am. Chem.
Soc. 116, 1184011848
171. Huang, Y., and Ackers, G. K. (1995)
Biochemistry 34, 63166327
172. Paoli, M., Dodson, G., Liddington, R. C., and
Wilkinson, A. J. (1997) J. Mol. Biol. 271, 161167
173. Daugherty, M. A., Shea, M. A., and Ackers, G.
K. (1994) Biochemistry 33, 1034510357
174. Stamler, J. S., Jia, L., Eu, J. P., McMahon, T. J.,
Demchenko, I. T., Bonaventura, J., Gernert, K.,
and Piantadosi, C. A. (1997) Science 276,
20342037
References
175. Upmacis, R. K., Hajjar, D. P., Chait, B. T., and
Mirza, U. A. (1997) J. Am. Chem. Soc. 119,
1042410429
176. Miller, L. M., Pedraza, A. J., and Chance, M.
R. (1997) Biochemistry 36, 1219912207
177. Gow, A. J., and Stamler, J. S. (1998) Nature
(London) 391, 169173
178. Lutter, L. C., Halvorson, H. R., and Calladine,
C. R. (1996) J. Mol. Biol. 261, 620633
179. Jia, L., Bonaventura, C., Bonaventura, J., and
Stamler, J. S. (1996) Nature (London) 380, 221
226
180. Rizzi, M., Wittenberg, J. B., Coda, A., Fasano,
M., Ascenzi, P., and Bolognesi, M. (1994) J.
Mol. Biol. 244, 8699
181. Rizzi, M., Wittenberg, J. B., Coda, A., Ascenzi,
P., and Bolognesi, M. (1996) J. Mol. Biol. 258,
15
182. Nguyen, B. D., Zhao, X., Vyas, K., La Mar, G.
N., Lile, R. A., Brucker, E. A., Phillips, G. N.,
Jr., Olson, J. S., and Wittenberg, J. B. (1998) J.
Biol. Chem. 273, 95179526
183. Weatherall, D. J., Clegg, J. B., Higgs, D. R.,
and Wood, W. G. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 34173484, McGraw-
Hill, New York
184. Bank, A., Mears, J. G., and Ramirez, F. (1980)
Science 207, 486493
185. Hirsch, R. E., Juszczak, L. J., Fataliev, N. A.,
Friedman, J. M., and Nagel, R. L. (1999) J. Biol.
Chem. 274, 1377713782
186. Arnone, A., Thillet, J., and Rosa, J. (1981) J.
Biol. Chem. 256, 85458552
187. Lehmann, H., and Huntsman, R. G. (1966)
Mans Haemoglobins, North-Holland Publ. Co.,
Amsterdam (p. 246)
188. Olson, J. S., Gibson, Q. H., Nagel, R. L., and
Hamilton, H. B. (1972) J. Biol. Chem. 247,
74857493
189. Poyart, C., Bursaux, E., Arnone, A.,
Bonaventura, J., and Bonaventura, C. (1980)
J. Biol. Chem. 255, 94659473
189a. Burmester, T., Weich, B., Reinhardt, S., and
Hankeln, T. (2000) Nature 407, 520523
190. Tomita, S. (1981) J. Biol. Chem. 256, 94959500
191. Giardina, B., Scatena, R., Clementi, M. E.,
Cerroni, L., Nuutinen, M., Brix, O., Sletten, S.
N., Castagnola, M., and Cond, S. G. (1993) J.
Mol. Biol. 229, 512516
192. Clementi, M. E., Scatena, R., Mordente, A.,
Cond, S. G., Castagnola, M., and Giardina, B.
(1996) J. Mol. Biol. 255, 229234
192a. Burr, A. H. J., Hunt, P., Wagar, D. R., Dewilde,
S., Blaxter, M. L., Vanfleteren, J. R., and
Moens, L. (2000) J. Biol. Chem. 275, 48104815
193. Lee, H. C., Wittenberg, J. B., and Peisach, J.
(1993) Biochemistry 32, 1150011506
194. Harutyunyan, E. H., Safonova, T. N.,
Kuranova, I. P., Popov, A. N., Teplyakov, A. V.,
Obmolova, G. V., Rusakov, A. A., Vainshtein,
B. K., Dodson, G. G., Wilson, J. C., and Perutz,
M. F. (1995) J. Mol. Biol. 251, 104115
195. Hargrove, M. S., Barry, J. K., Brucker, E. A.,
Berry, M. B., Phillips, G. N., Jr., Olson, J. S.,
Arredondo-Peter, R., Dean, J. M., Klucas, R.
V., and Sarath, G. (1997) J. Mol. Biol. 266,
10321042
196. Trevaskis, B., Watts, R. A., Andersson, C. R.,
Llewellyn, D. J., Hargrove, M. S., Olson, J. S.,
Dennis, E. S., and Peacock, W. J. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 1223012234
196a. Hargrove, M. S., Brucker, E. A., Stec, B.,
Sarath, G., Arredondo-Peter, R., Klucas, R. V.,
Olson, J. S., and Phillips, G. N., Jr. (2000)
Structure 8, 10051014
197. Hardison, R. C. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 56755679
198. Potts, M., Angeloni, S. V., Ebel, R. E., and
Bassam, D. (1992) Science 256, 16901692
199. Yeh, S.-R., Couture, M., Ouellet, Y., Guertin,
M., and Rousseau, D. L. (2000) J. Biol. Chem.
275, 16791684
200. Aronson, H.-E. G., Royer, W. E., Jr., and
Hendrickson, W. A. (1994) Protein Sci. 3,
17061711
201. Honzatko, R. B., Hendrickson, W. A., and
Love, W. E. (1985) J. Mol. Biol. 184, 147164
201a. Qiu, Y., Maillett, D. H., Knapp, J., Olson, J. S.,
and Riggs, A. F. (2000) J. Biol. Chem. 275,
1351713528
202. Condon, P. J., and Royer, W. E. J. (1994) J. Biol.
Chem. 269, 2525925267
203. Royer, W. E., Jr., Heard, K. S., Harrington, D.
J., and Chiancone, E. (1995) J. Mol. Biol. 253,
168186
204. Mozzarelli, A., Bettati, S., Rivetti, C., Rossi, G.
L., Colotti, G., and Chiancone, E. (1996) J. Biol.
Chem. 271, 36273632
205. Yang, J., Kloek, A. P., Goldberg, D. E., and
Mathews, F. S. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 42244228
206. Huang, S., Huang, J., Kloek, A. P., Goldberg,
D. E., and Friedman, J. M. (1996) J. Biol. Chem.
271, 958962
207. Lamy, J., Kuchumov, A., Taveau, J.-C.,
Vinogradov, S. N., and Lamy, J. N. (2000) J.
Mol. Biol. 298, 633647
208. de Haas, F., Taveau, J.-C., Boisset, N.,
Lambert, O., Vinogradov, S. N., and Lamy, J.
N. (1996) J. Mol. Biol. 255, 140153
209. de Haas, F., Zal, F., You, V., Lallier, F.,
Toulmond, A., and Lamy, J. N. (1996) J. Mol.
Biol. 264, 111120
210. de Haas, F., Boisset, N., Taveau, J.-C.,
Lambert, O., Vinogradov, S. N., and Lamy, J.
N. (1996) Biophys. J. 70, 19731984
211. Zuckerkandl, E. (1965) Sci. Am. 212(May),
110118
212. Perutz, M. F. (1986) Nature (London) 322, 405
213. Wood, W. B., and Edgar, R. S. (1967) Sci. Am.
217(Jul), 6074
214. Kikuchi, Y., and King, J. (1975) J. Mol. Biol. 99,
645672
215. Casjens, S., and King, J. (1975) Ann. Rev.
Biochem. 44, 555611
216. Pollard, T. D., and Craig, S. W. (1982) Trends
Biochem. Sci. 7, 5558
217. Ito, K., Date, T., and Wickner, W. (1980) J. Biol.
Chem. 255, 21232130
218. McDonnell, P. A., Shon, K., Kim, Y., and
Opella, S. J. (1993) J. Mol. Biol. 233, 447463
219. Sanders, J. C., Haris, P. I., Chapman, D., Otto,
C., and Hemminga, M. A. (1993) Biochemistry
32, 1244612454
220. Guy-Caffey, J. K., and Webster, R. E. (1993) J.
Biol. Chem. 268, 54965503
221. Duda, R. L., Martincic, K., and Hendrix, R. W.
(1995) J. Mol. Biol. 247, 636647
222. Xie, Z., and Hendrix, R. W. (1995) J. Mol. Biol.
253, 7485
223. Marvik, C. J., Dokland, T., Nokling, R. H.,
Jacobsen, E., Larsen, T., and Lindqvist, B. H.
(1995) J. Mol. Biol. 251, 5975
224. LeCuyer, K. A., Behlen, L. S., and Uhlenbeck,
O. C. (1995) Biochemistry 34, 1060010606
225. McKenna, R., Ilag, L. L., and Rossmann, M. G.
(1994) J. Mol. Biol. 237, 517543
226. Dokland, T., McKenna, R., Ilag, L. L., Bowman,
B. R., Incardona, N. L., Fane, B. A., and Rossmann,
M. G. (1997) Nature (London) 389, 308313
227. Thuman-Commike, P. A., Greene, B., Jakana,
J., Prasad, B. V. V., King, J., Prevelige, P. E., Jr.,
and Chiu, W. (1996) J. Mol. Biol. 260, 8598
228. Parker, M. H., Stafford, W. F., III, and Prevelige,
P. E., Jr. (1997) J. Mol. Biol. 268, 655665
376 Chapter 7. How Macromolecules Associate
289. Herrmann, H., Hner, M., Brettel, M., Mller,
S. A., Goldie, K. N., Fedtke, B., Lustig, A.,
Franke, W. W., and Aebi, U. (1996) J. Mol. Biol.
264, 933953
289a. Herrmann, H., Strelkov, S. V., Feja, B., Rogers,
K. R., Brettel, M., Lustig, A., Hner, M., Parry,
D. A. D., Steinert, P. M., Burkhard, P., and
Aebi, U. (2000) J. Mol. Biol. 298, 817832
289b. Heimburg, T., Schuenemann, J., Weber, K.,
and Geisler, N. (1996) Biochemistry 35, 1375
1382
290. Geisler, N., Schnemann, J., and Weber, K.
(1992) Eur. J. Biochem. 206, 841852
290a. Bellin, R. M., Sernett, S. W., Becker, B., Ip, W.,
Huiatt, T. W., and Robson, R. M. (1999) J. Biol.
Chem. 274, 2949329499
291. Leterrier, J. F., Ks, J., Hartwig, J., Vegners, R.,
and Janmey, P. A. (1996) J. Biol. Chem. 271,
1568715694
292. Betts, J. C., Blackstock, W. P., Ward, M. A., and
Anderton, B. H. (1997) J. Biol. Chem. 272,
1292212927
293. Carter, J., Gragerov, A., Konvicka, K., Elder,
G., Weinstein, H., and Lazzarini, R. A. (1998)
J. Biol. Chem. 273, 51015108
294. Osborn, M., and Weber, K. (1986) Trends
Biochem. Sci. 11, 469472
295. Gibson, M. A., Kumaratilake, J. S., and Cleary,
E. G. (1989) J. Biol. Chem. 264, 45904598
296. Steinert, P. M. (1991) J. Structural Biol. 107,
157174
297. Cohlberg, J. A. (1993) Trends Biochem. Sci. 18,
360362
298. Nakajima-Iijima, S., Hamada, H., Reddy, P.,
and Kakunaga, T. (1985) Proc. Natl. Acad. Sci.
U.S.A. 82, 61336137
299. Orlova, A., Chen, X., Rubenstein, P. A., and
Egelman, E. H. (1997) J. Mol. Biol. 271, 235243
300. Fulton, A. B. (1984) The Cytoskeleton: Cellular
Architecture and Choreography, Chapman and
Hall, New York
301. Bershadsky, A. D., and Vasiliev, J. M. (1988)
Cytoskeleton, Plenum, New York
302. DeRosier, D. J., Tilney, L. G., and Egelman, E.
(1980) Nature (London) 287, 291296
303. Collier, N. C., and Wang, K. (1982) J. Biol.
Chem. 257, 6937943
304. Moon, R. T., and Lazarides, E. (1983) Proc.
Natl. Acad. Sci. U.S.A. 80, 54945499
305. Meek, R. L., Lonsdale-Eccles, J. D., and Dale,
B. A. (1983) Biochemistry 22, 48674877
306. Steinert, P. M., and Idler, W. W. (1979)
Biochemistry 18, 56645669
307. Dustin, P. (1980) Sci. Am. 243(Aug.), 6776
308. Mandelkow, E., and Mandelkow, E.-M. (1994)
Current Opinion in Structural Biology 4, 171179
309. Hyams, J. S., and Lloyd, C. W., eds. (1994)
Microtubules, Wiley-Liss, New York
310. Sosa, H., and Milligan, R. A. (1996) J. Mol.
Biol. 260, 743755
311. Margulis, L., To, L., and Chase, D. (1978)
Science 200, 11181124
312. Thormhlen, M., Marx, A., Mller, S. A., Song,
Y.-H., Mandelkow, E.-M., Aebi, U., and
Mandelkow, E. (1998) J. Mol. Biol. 275, 795809
313. Weber, K., Pollack, R., and Bibring, T. (1975)
Proc. Natl. Acad. Sci. U.S.A. 72, 459463
314. Wolf, S. G., Nogales, E., Kikkawa, M.,
Gratzinger, D., Hirokawa, N., and Downing,
K. H. (1996) J. Mol. Biol. 262, 485501
315. Nogales, E., Wolf, S. G., and Downing, K. H.
(1998) Nature (London) 391, 199203
316. Pennisi, E. (1998) Science 279, 176177
317. Zheng, Y., Wong, M. L., Alberts, B., and
Mitchison, T. (1995) Nature (London) 378,
578583
318. Hackney, D. D. (1995) Nature (London) 376,
215216
229. Butcher, S. J., Dokland, T., Ojala, P. M.,
Bamford, D. H., and Fuller, S. D. (1997) EMBO
J. 16, 44774487
230. Olson, N. H., Baker, T. S., Willingmann, P.,
and Incardona, N. L. (1992) J. Structural Biol.
108, 168175
231. Hayashi, M., Aoyama, A., Richardson, D. L.,
Jr., and Hayashi, M. N. (1988) in The Bacterio-
phages (Calendar, R., ed), pp. 171, Plenum,
New York
232. Butcher, S. J., Bamford, D. H., and Fuller, S. D.
(1995) EMBO J. 14, 60786086
233. Berger, B., Shor, P. W., Tucker-Kellogg, L., and
King, J. (1994) Proc. Natl. Acad. Sci. U.S.A. 91,
77327736
234. Steven, A. C., Greenstone, H. L., Booy, F. P.,
Black, L. W., and Ross, P. D. (1992) J. Biol.
Chem. 228, 870884
235. van Driel, R. (1980) J. Mol. Biol. 138, 2742
236. King, J., and Mykolajewycz, N. (1973) J. Mol.
Biol. 75, 339358
237. Feller, S. M., Ren, R., Hanafusa, H., and
Baltimore, D. (1994) Trends Biochem. Sci. 19,
453458
238. Pawson, T. (1995) Nature (London) 373, 573
580
239. Koch, C. A., Anderson, D., Moran, M. F., Ellis,
C., and Pawson, T. (1991) Science 252, 668674
240. Maignan, S., Guilloteau, J.-P., Fromage, N.,
Arnoux, B., Becquart, J., and Ducruix, A.
(1995) Science 268, 291293
241. Ullner, M., Selander, M., Persson, E., Stenflo,
J., Drakenberg, T., and Teleman, O. (1992)
Biochemistry 31, 59745983
242. Meininger, D. P., Hunter, M. J., and Komives,
E. A. (1995) Protein Sci. 4, 16831695
243. Bersch, B., Hernandez, J.-F., Marion, D., and
Arlaud, G. J. (1998) Biochemistry 37, 12041214
244. Rand, M. D., Lindblom, A., Carlson, J.,
Villoutreix, B. O., and Stenflo, J. (1997) Protein
Sci. 6, 20592071
245. Mller, K., Gombert, F. O., Manning, U.,
Grossmller, F., Graff, P., Zaegel, H., Zuber, J.
F., Freuler, F., Tschopp, C., and Baumann, G.
(1996) J. Biol. Chem. 271, 1650016505
246. Fry, M. J., Panayotou, G., Booker, G. W., and
Waterfield, M. D. (1993) Protein Sci. 2, 1785
1797
247. Malek, S. N., Yang, C. H., Earnshaw, W. C.,
Kozak, C. A., and Desiderio, S. (1996) J. Biol.
Chem. 271, 69526962
248. Tong, L., Warren, T. C., King, J., Betageri, R.,
Rose, J., and Jakes, S. (1996) J. Mol. Biol. 256,
601610
249. Xu, R. X., Word, J. M., Davis, D. G., Rink, M.
J., Willard, D. H., Jr., and Gampe, R. T., Jr.
(1995) Biochemistry 34, 21072121
250. Mikol, V., Baumann, G., Zurini, M. G. M., and
Hommel, U. (1995) J. Mol. Biol. 254, 8695
251. Pascal, S. M., Yamazaki, T., Singer, A. U., Kay,
L. E., and Forman-Kay, J. D. (1995) Biochemis-
try 34, 1135311362
252. Zhou, M.-M., Meadows, R. P., Logan, T. M.,
Yoon, H. S., Wade, W. S., Ravichandran, K. S.,
Burakoff, S. J., and Fesik, S. W. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 77847788
253. Metzler, W. J., Leiting, B., Pryor, K., Mueller,
L., and Farmer, B. T., II. (1996) Biochemistry 35,
62016211
254. Viguera, A. R., Arrondo, J. L. R., Musacchio,
A., Saraste, M., and Serrano, L. (1994)
Biochemistry 33, 1092510933
255. Liang, J., Chen, J. K., Schreiber, S. L., and
Clardy, J. (1996) J. Mol. Biol. 257, 632643
256. Lim, W. A., Richards, F. M., and Fox, R. O.
(1994) Nature (London) 372, 375379
257. Guruprasad, L., Dhanaraj, V., Timm, D.,
Blundell, T. L., Gout, I., and Waterfield, M. D.
(1995) J. Mol. Biol. 248, 856866
258. van der Geer, P., and Pawson, T. (1995) Trends
Biochem. Sci. 20, 277280
259. Zhou, M.-M., Ravichandran, K. S., Olejniczak,
E. T., Petros, A. M., Meadows, R. P., Sattler,
M., Harlan, J. E., Wade, W. S., Burakoff, S. J.,
and Fesik, S. W. (1995) Nature (London) 378,
584592
260. Rameh, L. E., Arvidsson, A.-k, Carraway, K.
L., III, Couvillon, A. D., Rathbun, G.,
Crompton, A., VanRenterghem, B., Czech, M.
P., Ravichandran, K. S., Burakoff, S. J., Wang,
D.-S., Chen, C.-S., and Cantley, L. C. (1997) J.
Biol. Chem. 272, 2205922066
261. Hemmings, B. A. (1997) Science 275, 1899
262. Blomberg, N., Baraldi, E., Nilges, M., and
Saraste, M. (1999) Trends Biochem. Sci. 24, 441
445
263. Koshiba, S., Kigawa, T., Kim, J.-H., Shirouzu,
M., Bowtell, D., and Yokoyama, S. (1997) J.
Mol. Biol. 269, 579591
264. Saras, J., and Heldin, C.-H. (1996) Trends
Biochem. Sci. 21, 455458
265. Cabral, J. H. M., Petosa, C., Sutcliffe, M. J.,
Raza, S., Byron, O., Poy, F., Marfatia, S. M.,
Chishti, A. H., and Liddington, R. C. (1996)
Nature (London) 382, 649652
266. Ponting, C. P. (1997) Protein Sci. 6, 464468
267. Resnick, D., Pearson, A., and Krieger, M.
(1994) Trends Biochem. Sci. 19, 58
268. Chang, Y., Mochalkin, I., McCance, S. G.,
Cheng, B., Tulinsky, A., and Castellino, F. J.
(1998) Biochemistry 37, 32583271
269. de Vos, A. M., Ultsch, M. H., Kelley, R. F.,
Padmanabhan, K., Tulinsky, A., Westbrook,
M. L., and Kossiakoff, A. A. (1992) Biochemis-
try 31, 270279
270. De Serrano, V. S., and Castellino, F. J. (1992)
Biochemistry 31, 1169811706
271. Sun, M.-F., Zhao, M., and Gailani, D. (1999) J.
Biol. Chem. 274, 3637336377
272. Macias, M. J., Hyvnen, M., Baraldi, E., Schultz,
J., Sudol, M., Saraste, M., and Oschkinat, H.
(1996) Nature (London) 382, 646649
273. Bogusky, M. J., Dobson, C. M., and Smith, R.
A. G. (1989) Biochemistry 28, 67286735
274. Hoffmann, W., and Hauser, F. (1993) Trends
Biochem. Sci. 18, 239243
275. Lamb, J. R., Tugendreich, S., and Hieter, P.
(1995) Trends Biochem. Sci. 20, 257259
276. Das, A. K., Cohen, P. T. W., and Barford, D.
(1998) EMBO J. 17, 11921199
277. Barlow, P. N., Luisi, B., Milner, A., Elliott, M.,
and Everett, R. (1994) J. Mol. Biol. 237, 201211
278. Lichtarge, O., Yamamoto, K. R., and Cohen, F.
E. (1997) J. Mol. Biol. 274, 325337
279. Saurin, A. J., Borden, K. L. B., Boddy, M. N.,
and Freemont, P. S. (1996) Trends Biochem. Sci.
21, 208214
280. Prez-Alvarado, G. C., Kosa, J. L., Louis, H.
A., Beckerle, M. C., Winge, D. R., and Summers,
M. F. (1996) J. Mol. Biol. 257, 153174
281. Porter, K. R., and Tucker, J. B. (1981) Sci. Am.
244(Mar), 5767
282. Weber, K., and Osborn, M. (1985) Sci. Am.
253(Oct), 110120
283. Amos, L. A., and Amos, B. W. (1991) Molecules of
the Cytoskeleton, The Guilford Press, New York
284. Lazarides, E. (1980) Nature (London) 283, 249
256
285. Fuchs, E., and Weber, K. (1994) Ann. Rev.
Biochem. 63, 345382
286. Fuchs, E., and Cleveland, D. W. (1998) Science
279, 514519
287. Kaufmann, S. H. (1989) J. Biol. Chem. 264,
1394613955
288. Ip, W., Hartzer, M. K., Pang, Y.-Y. S., and
Robson, R. M. (1985) J. Mol. Biol. 183, 365375
References
377
319. Raff, E. C., Fackenthal, J. D., Hutchens, J. A.,
Hoyle, H. D., and Turner, F. R. (1997) Science
275, 7073
320. Angelastro, J. M., and Purich, D. L. (1992) J.
Biol. Chem. 267, 2568525689
321. Terry, B. J., and Purich, D. L. (1980) J. Biol.
Chem. 255, 1053210536
322. Rakic, P., Knyihar-Csillik, E., and Csillik, B.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 9218
9222
323. Rodionov, V. I., and Borisy, G. G. (1997)
Science 275, 215218
References
Study Questions
1. Rewrite Equations 6-75 through 6-77 in terms of
dissociation constants. These may be labeled K
1
,
K
2
, K
i
, etc., as is conventional, but you may prefer
to use K
1d
, K
2d
, K
id
, etc., to avoid confusion.
2. A molecule has two identical binding sites for a
ligand X. The Gibbs energy of interaction between
ligands bound to the same molecule, , is defined
as the change in Gibbs energy of binding of the
ligand to the molecule that results from the prior
binding of a ligand at the adjacent site. If the
saturation fraction is Y, show from the equation for
the binding isotherm that the following equation
holds when Y = 1/ 2:
3. The hydrogen ion binding curve for succinate is
shown is Fig. 7-4. From the curve estimate and
the microscopic association constants.
4. A linear chain molecule has a very large number
of identical binding sites for a ligand X. The Gibbs
energy of interaction between ligands bound to
adjacent sites is . Interactions between non-
nearest neighbors are considered negligible. If the
binding constant for a site adjacent to unoccupied
sites is K
r
, the binding isotherm is given by
[Applequist, J. (1977) J. Chem. Ed. 54, 417]. Show
from the equation for the binding isotherm that
the following equation holds at Y = 1/ 2;
dY/d ln [ X ] = 1/4 e
/ 2RT
5. The binding of adenosine to polyribouridylic acid
[poly(U)] has been studied by the method of
equilibrium dialysis [Huang and Tso (1966) J. Mol.
Biol. 16, 523]. The table below gives the fraction of
poly(U) sites occupied, Y at various molar concen-
trations of free adenosine [A] at 5C. Assuming
that the nearest-neighbor interaction model is
correct, determine the intrinsic association con-
stant for the binding of adenosine to poly U and
the free energy of interaction of adjacent bound
adenosines. Do the bound molecules attract or
repel each other?
324. Hoenger, A., and Milligan, R. A. (1996) J. Mol.
Biol. 263, 114119
325. Fan, J., Griffiths, A. D., Lockhart, A., Cross, R.
A., and Amos, L. A. (1996) J. Mol. Biol. 259,
325330
326. Hirose, K., Fan, J., and Amos, L. A. (1995) J.
Mol. Biol. 251, 329333
327. Knop, M., and Schiebel, E. (1997) EMBO J. 16,
69856995
328. Moritz, M., Braunfeld, M. B., Sedat, J. W.,
Alberts, B., and Agard, D. A. (1995) Nature
(London) 378, 638640
329. Berridge, M. J. (1990) J. Biol. Chem. 265, 9583
9586
330. Arnold, C. S., Johnson, G. V. W., Cole, R. N.,
Dong, D. L.-Y., Lee, M., and Hart, G. W. (1996)
J. Biol. Chem. 271, 2874128744
331. Bosc, C., Cronk, J. D., Pirollet, F., Watterson,
D. M., Haiech, J., Job, D., and Margolis, R. L.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 2125
2130
dY/d ln[X] =
1
2(1 + e
/RT
)
0.51 0 3.07 0.72
2.10 0 4.00 0.92
2.70 0.15 6.50 0.93
2.96 0.36 8.50 0.93
3.01 0.52 10.00 1.00
[A] x 10
3
Y [A] x 10
3
Y
Y =
K[X]e
/RT
1
2{K[X]e
/RT
1)
2
+ 4K[X]}
1/2
1
2
+
378 Chapter 8. Lipids, Membranes, and Cell Coats
A. Lipid Structures
1. Fatty Acids, Fatty Alcohols, and Hydrocarbons
2. Acylglycerols, Ether Lipids, and Waxes
3. Phospholipids
4. Glycolipids
5. Sphingolipids
6. Sterols and Other Isoprenoid Lipids
B. Membranes
1. The Structure of Membranes
Lipids of membranes
Liquid crystals, liposomes, and artificial membranes
Physical properties of membrane lipids
Functions of phospholipid head groups
Non-bilayer structures of phospholipids
Membrane fluidity and life
Electrical properties of membranes
The two sides of a membrane
2. Membrane Proteins
Integral membrane proteins
Anchors for proteins
Analyzing erythocyte membranes
Glycoproteins
Connections to the cytoskeleton
Integrins and focal adhesions
Other cell adhesion molecules
Peripheral proteins of the outer membrane surface
3. Enzymes and Membrane Metabolism
C. The Transport of Molecules through Membranes
1. Facilitated Diffusion and Active Transport
2. Pores, Channels, and Carriers
Porins
Aquaporins
Ion channels
Channel-forming toxins and antibiotics
Ionophores and other mobile carriers
3. The 12-Helix Major Facilitator Superfamily
Entrance of sugars into cells
Cotransport of sugars and other nutrients
with H
+
or Na
+
4. Active Transport Systems
Periplasmic permeases
The bacterial phosphotransferase system
380
380
382
382
387
389
389
390
390
392
392
394
396
397
397
400
401
401
401
402
403
404
405
405
406
407
409
410
410
411
411
411
412
414
414
415
415
416
417
417
419
420
420
421
422
422
425
427
427
427
428
431
431
431
431
432
436
436
440
440
440
443
444
452
384
386
398
418
438
439
442
380
381
393
435
Boxes
Box 8-A The Platelet-Activating Factor
Box 8-B Dipalmitoylphosphatidylcholine and the
Surfactant System of the Lungs
Box 8-C Electron Paramagnetic Resonance (EPR)
Spectra and Spin Lables
Box 8-D Colicins: Antibiotic Proteins
Box 8-E Genetic Defects of Collagen Structure
Box 8-F Skin
Box 8-G The Biochemistry of Teeth
Tables
Table 8-1 Some Important Fatty Acids
Table 8-2 Fatty Acid Composition (in %) of Some
Typical Fats and Oils
Table 8-3 Estimated Chemical Compositions of
Some Membranes
Table 8-4 Types of Vertebrate Collagen
5. Transport of Ions
Anions
Cation channels
Active transport of cations
ATP-driven ion pumps
6. Exocytosis, Endocytosis, and the Flow of
Membrane Constituents
D. Communication
E. The Extracellular Matrix and Cell Walls
1. The Structure of Bacterial Cell Walls
The outer membrane of gram-negative bacteria
Teichoic and teichuronic acids
Mycobacteria
Other bacterial coats
2. The Surroundings of Animal Cells
The collagens
Elastic fibers
Cartilage and basement membranes
Fibrillin and Marfans syndrome
The cuticles of invertebrates
Bones, teeth, and shells
3. Cell Walls of Fungi and Green Plants
References
Study Questions
Contents
Portion of an endocytic vacuole forming in the
plasma membrane of a cultured fibroblast. The
view is from the inside of the cell and shows a
large clathrin cage assembling to form a coated
vesicle. The overall diameter of the vacuole is
~ 0.2 m. Clathrin cages vary in size and in the
number of faces but are typically ~ 0.1 m in
diameter (see Fig. 8-27 and associated references).
Courtesy of Barbara Pearse.
379
The boundary between a living cell and its sur-
roundings is the incredibly thin (710 nm) plasma
membrane. This vital partition, which controls the
flow of materials into and out of a cell and which
senses and controls the response of cells to hor-
mones and other external signals, consists largely of
phospholipids together with embedded proteins. The
nonpolar chains of the phospholipids stick together to
form a double molecular layer or bilayer which provides
the basic structure of almost all biological membranes.
Phospholipids, together with other natural materials
that have a high solubility in apolar solvents or are
structurally related to compounds with such solubility
properties, are classified as lipids. The most abundant
lipids are the fats, compounds that are stored by animals
and by many plants as an energy reserve (Fig. 8-1).
Other lipids form the outer cuticle of plants and yet
others serve as protective coatings on feathers and
hair. Vitamins A, D, K, and E and ubiquinone are all
lipids as are a variety of hormones and such light-
absorbing plant pigments as the chlorophylls and
carotenoids. Many of these compounds are dissolved
in or partially embedded in the plasma membrane of
bacteria or in the mitochondrial and chloroplast mem-
branes of higher organisms. Membranes serve many
purposes. The most obvious is to divide space into
compartments. Thus, the plasma membrane forms cell
boundaries and mitochondrial membranes separate
the enzymes and metabolites of mitochondria from
those of the cytosol. Membranes are semipermeable
and regulate the penetration into cells and organelles
of both ionic and nonionic substances. Many of these
materials are brought into the cell against a concentra-
tion gradient. Hence, osmotic work must be done in a
process known as active transport. Many enzymes,
Figure 8-1 Electron micrograph of a thin section of a fat
storage cell or adipocyte. L, the single large fat droplet; N,
nucleus; M, mitochondria; En, endothelium of a capillary
containing an erythrocyte (E); CT, connective tissue ground
substance which contains collagen fibers (Co) and fibroblasts
(F). The basement membranes (BM) surrounding the endo-
thelium and the fat cell are also marked. From Porter and
Bonneville.
6
Courtesy of Mary Bonneville.
Lipids, Membranes, and Cell Coats
8
BM
CT
BM
En
E
G
L
Co
F
N
G
M
380 Chapter 8. Lipids, Membranes, and Cell Coats
including those responsible for most of the oxidative
metabolism of cells, are found in membranes of bacteria
and of mitochondria. Within the chloroplasts of green
leaves, highly folded membranes containing chlorophyll
absorb energy from the sunlight. Thin membranes
contain the photoreceptor proteins that function in
vision. Electrical impulses are transmitted along the
membranes of nerve cells.
The outer surfaces of membranes are designed to
interact with the cells external world. Special receptors
sense the presence of hormones. Binding proteins
await the arrival of needed nutrients and help to bring
them into cells. Highly individual arrangements of
protein and of the carbohydrate fuzz of glycoproteins
and sphingolipids screen the outer surface, helping to
prevent attack by foreign bacteria, viruses, and toxins.
A. Lipid Structures
Unlike proteins, polysaccharides, and nucleic acids,
most lipids are not polymers. However, they are made
by linking together smaller molecules.
15
Among the
building blocks of lipids are fatty acids, glycerol,
phosphoric acid, and sugars. Many lipids have
both polar and nonpolar regions. This gives them an
amphipathic character, i.e., a tendency toward both
hydrophobic and hydrophilic behavior, and accounts
for their tendency to aggregate into membranous
structures. Notice that in the following structure the
carbon atoms of glycerol have been numbered 1 to 3.
Although glycerol is not chiral, the positioning of the
two hydroxymethyl groups is not equivalent. If the
priority, used in the RS system (page 42) for the C-1
group is raised (e.g., by ester formation) to be higher
than that for the C-3 group, the molecule would have
the S configuration. The C-1 group is said to be pro-S
and the C-3 group pro-R. According to the stereo-
chemical numbering (sn) system, which is discussed
further in Chapter 9, the carbon in the pro-S position
is numbered 1.
1. Fatty Acids, Fatty Alcohols, and Hydrocarbons
The names and structures of some fatty acids are
summarized in Table 8-1. Notice that these acids have
straight carbon chains and may contain one or more
double bonds. Except for the smallest members of
the series, which are soluble in water, fatty acids are
strongly hydrophobic. However, they are all acids
with pK
a
values in water of ~4.8. To the extent that
free fatty acids occur in nature, they are likely to be
found in interfaces between lipid and water with the
carboxyl groups dissociated and protruding into the
water. However, most naturally occurring fatty acids
a
The number of carbon atoms is given first, then the number of
double bonds. The positions of the lowest numbered carbon of
each double bond and whether the configuration is cis (c) or trans
(t) are indicated in parentheses.
b
Official IUPAC names of these and other acyl groups have been
designated by the Commission of the Nomenclature of Organic
Chemistry in Pure and Applied Chemistry 10, 111125 (1965). In
a number of cases IUPAC inserted an o in the traditional name,
e.g., palmityl became palmitoyl and crotonyl became crotonoyl.
However, acetyl was not changed. In many cases the systematic
names, e.g., hexadecanoyl (from hexadecanoic acid), are preferable
and IUPACIUB recommends that alkyl radicals always be designated
by systematic names, e.g., hexadecyl, not palmityl alcohol. The
older use of palmityl for both acyl and alkyl radicals was one
reason for IUPACs adoption of new names for acyl radicals.
c
Systematic names are not often used because of their complexity,
e.g., linolenic acid is cis,cis,cis-9,12,15-octadecatrienoic acid.
Saturated fatty acids
1 Methanoic Formic Formyl
2 Ethanoic Acetic Acetyl
3 Propanoic Propionic Propionyl
4 Butanoic Butyric 4:0 Butyryl
12 Dodecanoic Lauric 12:0 Lauroyl
b
14 Tetradecanoic Myristic 14:0 Myristoyl
16 Hexadecanoic Palmitic 16:0 Palmitoyl
18 Octadecanoic Stearic 18:0 Stearoyl
20 Eicosanoic Arachidic 20:0
22 Docosanoic Behenic 22:0
24 Tetracosanoic Lignoceric 24:0
Unsaturated fatty acids
c
4 Crotonic 4:1(2t) Crotonoyl
16 Palmitoleic 16:1(9c)
18 Oleic 18:1(9c) Oleoyl
18 Vaccenic 18:1(11c)
18 Linoleic 18:2(9c,12c)
18 Conjugated 18:2(9c,11t)
linoleic
18 Linolenic 18:3(9c,12c,15c)
20 Arachidonic 20:4(5c,8c,11c,14c)
TABLE 8-1
Some Important Fatty Acids
No. carbon Systematic Common Common name
atoms name name Abbreviation
a
of acyl group
Stearic acid, 18 carbon atoms, major
component of animal triacylglycerols
C
CH
3
O
HO
C
CH
2
OH
CH
2
OH
HO
H
pro-S group
1
2
3
Labeled by the stereochemical
numbering (sn) system
A fatty acid
Glycerol
381 A. Lipid Structures
Like plants, animals contain palmitic and oleic
acids. In addition, large amounts of stearic acid and
small amounts of the C
20
, C
22
, and C
24
acids are also
present. Phospholipids of photoreceptor membranes
of the retina contain fatty acid chains as long as C
36
.
7
The variety of fatty acids found in animals is greater
than in a given plant species. A large fraction of the
fatty acids present in most higher organisms are unsat-
urated and contain strictly cis double bonds. Table 8-2
shows the fatty acid composition of some typical
triacylglycerol mixtures.
are esterified or combined via amide linkages in com-
plex lipids. For example, ordinary fats are largely the
fatty acid esters of glycerol called triacylglycerols
(triglycerides).
There is a seemingly endless variety of fatty acids,
but only a few of them predominate in any single
organism. Most fatty acid chains contain an even
number of carbon atoms. In higher plants the C
16
palmitic acid and the C
18
unsaturated oleic and
linoleic acids predominate. The C
18
saturated stearic
acid is almost absent from plants and C
20
to C
24
acids
are rarely present except in the outer cuticle of leaves.
Certain plants contain unusual fatty acids which may
be characteristic of a taxonomic group. For example,
the Compositae (daisy family) contain acetylenic fatty
acids and the castor bean contains the hydroxy fatty
acid ricinoleic acid.
Bacteria usually lack polyunsaturated fatty acids
but often contain branched fatty acids, cyclopropane-
containing acids, hydroxy fatty acids, and unesterified
fatty acids. Mycobacteria, including the human patho-
gen Mycobacterium tuberculosis, contain mycolic acids.
In these compounds the complex grouping R contains
a variety of functional groups including OH, OCH
3
,
HO
C
C
O
HO
H
H
CH
2
C C
HO
O
C
OH
H
R
O
H
Branched fatty acids of the anteiso series have a branch here
and a 5-carbon starter piece derived from isoleucine
Branched fatty acids of the iso series
contain a 5-carbon starter piece derived
from leucine or a 4-carbon piece derived from valine
A major fatty acid of lactobacilli
C
22
or C
24
alkyl group
A complex chain of about 60 carbon atoms
with a variety of functional groups (see text)
A mycolic acid
Lactobacillic acid
Branched fatty acids
C=O, COOH, cyclopropane rings, methyl branches,
and C=C bonds. Each species of Mycobacterium contains
about two dozen different mycolic acids
9,10
as well as
other complex C
30
C
56
fatty acids (see Eq. 21-5).
11
Certain polyunsaturated fatty acids are essential
in the human diet (see Box 21-B). One of these,
arachidonic acid (which may be formed from dietary
linoleic acid), serves as a precursor for the formation
of the hormones known as prostaglandins and a series
of related prostanoids. Lipids of animal origin also
TABLE 8-2
Fatty Acid Composition (in %) of Some Typical
Fats and Oils
a
No. of carbon atoms and (following colon)
the number of double bonds
Fats and oils 14 16 18 16:1 18:1 18:2
Human depot fat 3 23 4 8 45 10
Beef tallow 4 30 25 5 36 1
Corn oil 13 2 31 54
Lard 1 28 15 3 42 9
a
From Gunstone.
8
One of a family of hormones, this one is
synthesized in tissues from arachidonic acid
COOH
O
H
H
H
HO
H OH
Prostaglandin PGE
2
Accounts for 60% of the fatty acids in seeds of
Crepis foetida, a member of the compositae family
C
C C
O
HO
Crepenynic acid 18:2 (9c, 12a)
Ricinoleic acid (12-hydroxyoleic acid)
C
O
HO
HO H
Accounts for up to 90% of the fatty acids of Ricinus communis (castor bean)
COOH
A nutritionally essential fatty acid (Box 21-A)
is shown here in a folded conformation
Arachidonic acid
382 Chapter 8. Lipids, Membranes, and Cell Coats
contain unusual unsaturated fatty acids. Among
them, conjugated linoleic acids are receiving atten-
tion for their possible cancer-preventive action. The
predominant form in meats, dairy products, and the
human body is the C
18
9-cis, 11-trans isomer whose
two double bonds are conjugated.
11a
Other lipid components include the fatty alcohols
which are formed by reduction of the acids. These are
esterified with fatty acids to form waxes. Both fatty
alcohols and free fatty acids occur in waxes together
with the esterified forms. These mixtures are found
on exterior surfaces of plants and animals. Plants
and, to a limited extent, animals are able to decarboxy-
late fatty acids in a multistep process to alkanes and
these too are important constituents of some waxes.
Small amounts of fatty acid amides such as cis-9,10-
octadecenoamide are present in low concentrations in
the cerebrospinal fluid of cats and rats as well as humans.
This compound accumulates in cats that are deprived
of sleep. When the synthetic compound was injected
into rats they fell into apparently normal sleep.
12
Insects make unsaturated as well as saturated hydro-
carbons. The former as well as long-chain alcohols and
their esters often form the volatile pheromones with
which insects communicate. Thus, the female pink
bollworm attracts a male with a sex pheromone con-
sisting of a mixture of the cis,cis and cis,trans isomers
of 7,11-hexadecadienyl acetate,
13
and European corn
borer males are attracted across the cornfields of Iowa
by cis-11-tetradecenyl acetate.
14
Addition of a little of
the trans isomer makes the latter sex attractant much
more powerful. Since more than one species uses the
same attractant, it is possible that the males can distin-
guish between different ratios of isomers or of mixtures
of closely related substances.
2. Acylglycerols, Ether Lipids, and Waxes
The components of complex lipids are linked in a
variety of ways. Often, glycerol acts as the central unit,
e.g., combining in ester linkage with three fatty acids to
formtriacylglycerols (triglycerides), the common fats
of adipose tissues and plant oils. Diacyl- and monoacyl-
glycerols (diglycerides, and monoglycerides) are
present to a lesser extent. In addition, small amounts
of alkyl ethers or alkenyl ethers are often present in
isolated lipids. They are especially abundant in fish
liver oils.
These ether lipids are all chiral molecules with an R
configuration but are derivatives of the nonchiral
glycerol. The carbon atoms of glycerol are numbered
using the stereochemical system which is described
on p. 470. The ether linkage is to the sn-1 carbon atom.
Most phospholipids are derivatives of the sn-3 phosphate
ester of glycerol.
Triacylglycerols and the ether lipids described in
the previous section are classified as neutral lipids.
Other neutral lipids are alcohols, waxes, aldehydes,
and hydrocarbons derived from fatty acids. These
sometimes have specific biological functions. For exam-
ple, fatty aldehydes are important in the biolumines-
cence of bacteria (Eq. 23-47).
3. Phospholipids
As major constituents of biological membranes,
phospholipids play a key role in all living cells.
The two principal groups of phospholipids are the
1-Alkyl-2,3-diacyl-sn-glycerol, an alkyl ether lipid
C
CH
2
CH
2
H
O
O C R
3
O
O CH
2
R
1
C R
2
O
C
CH
2
CH
2
H
O
O C R
3
O
O CH
C R
2
O
1
1
CHR
1
1-Alkenyl-2,3-diacyl-sn-glycerol, an alkenyl ether
H
2
N
C
O
9
cis-9,10-Octadecenoamide (Oleic acid amide)
A wax
C
O
O
An ester of a fatty acid and a fatty alcohol
C
O
O
C
O
O
C
O
O
CH
2
C H
CH
2
*
Notice the chiral center, designated by the asterisk
A triacylglycerol (fat)
383
glycerophospholipids (glycero-
phosphatides; Fig. 8-2) which con-
tain the alcohol glycerol and the
sphingophospholipids which
contain the alcohol sphingosine
(Section 5). The glycerophospho-
lipids can be thought of as arising
from the building blocks glycerol,
fatty acids, the dihydrogen phos-
phate ion H
2
PO
4

, and the appro-


priate alcohol by removal of four
molecules of water (Eq. 8-1). They
are derivatives of sn-glycerol-3-
phosphate. Esterification of this
alcohol with two fatty acids gives
a phosphatidic acid (Fig. 8-2).
Formation of a phosphate diester
linkage to one of the alcohols
choline, serine, or ethanolamine
yields a glycerophospholipid.
The resulting three groups
of phospholipids are called
phosphatidylcholine (lecithin),
phosphatidylserine, and
phosphatidylethanolamine,
respectively (Fig. 8-2).
The phosphate and choline,
ethanolamine, or serine portions
of the phosphatide are electrically
charged and provide a polar head
for the molecule. In all three cases
the positively charged group is
able to fold back and form an ion
pair with the negatively charged
phosphate group. However, the
methyl groups surrounding the
nitrogen in phosphatidylcholine
prevent a very close approach and
with phosphatidylserine the adja-
cent carboxylate group weakens
this electrostatic interaction. Un-
like the triacylglycerols, most of
which are liquid at body tempera-
ture, phospholipids are solid at
this temperature. This property,
like the ionic properties of the
phosphatides, is doubtless related
to their suitability for functioning
in biological membranes.
(8-1) A phosphatidylcholine (lecithin).
A. Lipid Structures
Figure 8-2 Structures of some phosphatides (glycerophospholipids).
O
C
O
C
H
2
C
H
2
C
H
O
P
O
Y

O
O
OH
O CH
2
CH
2
N
+
(CH
3
)
3
O CH
2
CH
2
NH
3
+
COO

O CH
2
CH
2
NH
3
+
O CH
2
CH CH
2
OH
OH
OH
O
HO
OH HO
OH
O CH
2
CH
Y =
CH
2
1
OH
4
O
5
Phosphatidic acid
Phosphatidylcholine (lecithin)
Phosphatidylserine (serine cephalin)
Phosphatidylethanolamine
(ethanolamine cephalin)
Phosphatidylglycerol
Diphosphatidylglycerol (cardiolipin),
a special component of bacterial and
mitochondrial membranes
PC
PA
PS
PE
PI Phosphatidylinositol
Abbrev.
C
O
The ester linked fatty acid in this position is
replaced in plasmalogens by a vinyl ether group:
and in ether phosphatides by an alkoxy group:
O
R
O
R

C
O
HO
C
O
HO
C
H
2
C
H
2
C
H
O
O
H
O
H
P
OH
OH

O
O
N
CH
3
H
2
C
H
3
C
H
3
C
CH
2
O
C
O
O
C
O
C
H
2
C
H
2
C
H
O
P
O
O
O
O
N
CH
3
H
2
C
H
3
C
H
3
C
CH
2
+
+
H
H

2H
2
O
sn-Glycerol 3-phosphate
(L--glycerol phosphate)
C
CH
2
OH
CH
2
O
HO
H
PO
3
2
1
2
3
384 Chapter 8. Lipids, Membranes, and Cell Coats
of all eukaryotes and have a specific role in regulating
responses of cells to hormones and other external
agents. See Chapter 11 for details. Phosphatidylinositol
also forms part of anchors used to hold certain proteins
onto membrane surfaces (see Fig. 8-13).
Bacteria and plants often make the anionic
phosphatidyglycerol in which the second glycerol is
esterified at its sn-1 position with the phosphate. Bacteria,
as well as mitochondria, contain diphosphatidylglycerol
(cardiolipin) in which phosphatidyl groups are attached
at both the 1 and 3 positions of glycerol (Fig. 8-2).
Ether phospholipids, analogous to the ether lipids
described in Section 2, are also widely distributed. The
alkenyl ether analogs of phosphatidylcholine (Fig. 8-2)
are called plasmalogens.
17
In neutrophils the 1-O-alkyl
ethers contain the major share of the cells arachidonic
acid, which is esterified in the 2 position.
18,19
In halophilic (salt loving), thermophilic, and meth-
anogenic bacteria, most of the lipids present are either
Part of the immune response consists of the
release from stimulated neutrophils, macrophages,
and other cells of a platelet activating factor
(PAF), a material that activates blood platelets.
Activated platelets aggregate, a response that ini-
tiates clot formation. They may also be lysed and
release stored substances that include platelet-
derived growth factors (see Chapter 11) and fibrin
stabilizing factor, a proenzyme that is converted to
the protein-crosslinking enzyme transglutaminase
(Eq. 2-17). See Fig. 12-17. However, the principal
interest in the platelet-activating factor has arisen
from its powerful effect in inducing inflammation
BOX 8-A THE PLATELET-ACTIVATING FACTOR
in surrounding tissues.
PAF has been identified as the following simple
ether phospholipid:
ad
A remarkably potent compound, its effects on plate-
lets are observed at concentrations of 10
11
to 10
10
M. Both lyso-PAF and the glycero-1- phosphocho-
line enantiomer are inactive. Specific receptors for
this factor are evidently present on platelet surfaces.
eg
One effect of PAF on platelets is to induce a
rapid (510s) cleavage of phosphatidylinositol
4,5-bisphosphate by phospholipase C to give diacyl-
glycerol and inositol 1,4,5-trisphosphate. The
subsequent effects of these two substances in causing
a rapid influx of Ca
2+
and in inducing a series of
secondary responses are outlined in Fig. 11-9. Among
these responses are the release of the materials
stored in the platelets granules. PAF also appears
to inhibit adenylate cyclase
b
and causes vasodila-
tion, a property not expected for a compound that
stimulates clotting. Receptors for PAF are also
present in the brain, where this phosholipid may
function in regulation of development.
h
Lecithins and related phospholipids usually con-
tain a saturated fatty acid in the C-1 position but an
unsaturated acid, which may contain from one to
four double bonds, at C-2. Arachidonic acid is often
present here. Hydrolysis of the ester linkage at C-2
yields a 1-acyl-3-phosphoglycerol, better known as a
lysophosphatidylcholine. The name comes from the
powerful detergent action of these substances which
leads to lysis of cells. Some snake venoms contain
phospholipases that form lysophosphatidylcholine.
Lysophosphatidic acid (1-acyl-glycerol-3-phosphate)
is both an intermediate in phospholipid biosynthesis
(Chapter 21) and also a signaling molecule released
into the bloodstream by activated platelets.
15
Another group of phosphatides contain the
hexahydroxycyclohexane known as inositol (Fig. 8-2,
see also Chapter 21).
16
Phosphatidylinositol, as well
as smaller amounts of phosphatides derived from
phosphate esters of inositol are present in membranes
Scanning EM of
activated blood
platelets.
Quest, Photo
Researchers
Transmission EM
of thin section through
activated blood platelets.
Photo Researchers
Platelet-activating factor (PAF)
1-O-alkyl-2-acetyl-sn-glycerophosphocholine
O
O-alkyl
C
H
CH
3
O
O
P O
O
O
N
H
3
C
H
3
C
CH
3
+

385
Many other phospholipids are present in small
amounts or in a limited number of species. These
include phosphonolipids, which contain a CP bond
and are abundant in ciliate protozoa such as Tetrahymena
and in some other invertebrates.
26
Phosphonoethyl-
amine replaces phosphoethanolamine in these lipids.
A consequence of this structural alteration is a high
degree of resistance to the action of the enzyme phos-
pholipase C. The phosphonolipids of the external
membrane of Tetrahymena are also ether lipids with an
alkoxy group in the sn-1 position. This makes them
phospholipids and glycolipids containing the C
20
isoprenoid phytanyl group or the C
40
diphytanyl
group
20 25
(see also Section 4), related isoprenoid
alcohols, or long-chain 1,2-diols.
25
An example of a
diphytanylglycerophospholipid is the following:
A. Lipid Structures
BOX 8-A (continued)
Stimulated platelets release arachidonic acid
rapidly from their phospholipids, apparently as
a result of activation of phospholipase A
2
. The
released arachidonate can in turn be metabolized
to endoperoxides and thromboxane A
2
(Chapter 21).
These compounds are also potent activators of
platelets and cause a self-activating or autocrine
effect.
i,j
While PAF has a beneficial function, it can
under some conditions contribute in a dangerous way
to inflammation and to allergic responses including
anaphylaxis,
j
asthma
g
and cold-induced urticaria.
k
Although the effect of PAF is separate from those of
histamine and of leukotrienes, these agents may act
cooperatively to induce inflammation.
l
The biosynthesis of PAF is discussed in Chapter
21, Section C. One pathway is deacylation at the
glycerol C-2 of a longer chain alkyl phospholipid
followed by acetylation at the same position.
m,n
a
Cusack, N. J. (1980) Nature (London) 285, 193
b
Hanahan, D. J. (1986) Ann. Rev. Biochem. 55, 483 509
c
Prescott, S. M., Zimmerman, G. A., and McIntyre, T. M. (1990) J.
Biol. Chem. 265, 17381 17384
d
Winslow, C. M., and Lee, M. L., eds. (1987) New Horizons in
Platelet Activating Factor Research, Wiley, New York
e
Hwang, S.-B., Lam, M.-H., and Pong, S.-S. (1986) J. Biol. Chem.
261, 532 537
f
Ishii, I., Izumi, T., Tsukamoto, H., Umeyama, H., Ui, M., and
Shimizu, T. (1997) J. Biol. Chem. 272, 7846 7854
g
Bazan, N. G. (1995) Nature (London) 374, 501 502
h
Hattori, M., Adachi, H., Aoki, J., Tsujimoto, M., Arai, H., and
Inoue, K. (1995) J. Biol. Chem. 270, 31345 31352
i
Bussolino, F., Sironi, M., Bocchietto, E., and Mantovani, A.
(1992) J. Biol. Chem. 267, 14598 14603
j
Darius, H., Lefer, D. J., Smith, J. B., and Leefer, A. M. (1986)
Science 232, 58 60
Kadsurenone
CH
3
O
CH
3
O
O
OCH
3
O
HO P CH
2
CH
2
NH
3
O
O
+

Phosphonoethylamine
k
Grandel, K. E., Farr, R. S., Wanderer, A. A., Eisenstadt, T. C.,
and Wasserman, S. I. (1985) N. Engl. J. Med. 313, 405 409
l
Tomeo, A. C., Egan, R. W., and Durn, W. N. (1991) FASEB J. 5,
2850 2855
m
Billah, M. M., Eckel, S., Myers, R. F., and Siegel, M. I. (1986)
J. Biol. Chem. 261, 5824 5831
n
Lee, T.-c, Ou, M.-c, Shinozaki, K., Malone, B., and Snyder, F.
(1996) J. Biol. Chem. 271, 209 217
o
Stafforini, D. M., McIntyre, T. M., Zimmerman, G. A., and
Prescott, S. M. (1997) J. Biol. Chem. 272, 17895 17898
p
Miwa, M., Hill, C., Kumar, R., Sugatan, J., Olson, M. S., and
Hanahan, D. J. (1987) J. Biol. Chem. 262, 527 530
q
Hwang, S.-B., Lam, M.-H., Biftu, T., Beattie, T. R., and Shen, T.-Y.
(1985) J. Biol. Chem. 260, 15639 15645
Platelets can inactivate PAF by the inverse sequence:
deacetylation followed by acylation with arachidonic
acid. PAF can also be hydrolyzed by a PAF acetylhydro-
lase, a phospholipase.
o
Absence of this enzyme in the
brain may be related to a human brain malformation.
h
Some tissues may contain a lipid that inhibits binding
of PAF.
p
The following compound from a Chinese herb
binds to PAF receptors and may be the forerunner of
useful drugs.
q
O C
H
2
C
H CH
2
O
O
P
O
O
O

CH
2
C
H
OH
HOH
2
C
1
A phospholipid derived from 2,3-di-O-phytanyl-sn-glycerol
Phytanyl groups
386 Chapter 8. Lipids, Membranes, and Cell Coats
BOX 8-B DIPALMITOYLPHOSPHATIDYLCHOLINE AND THE SURFACTANT SYSTEM OF THE LUNGS
When air is exhaled the small alveoli of the
lungs could collapse if it were not for the surface
active material (surfactant) present in the fluid that
coats the lungs.
a e
In fact, the lack of adequate
surfactant is the cause of respiratory distress
syndrome, a major cause of death among prema-
ture infants and a disease that may develop in
acute form in adults. The surfactant material
forms a thin film of high fluidity at the airliquid
interface and lowers the surface tension from the
72 mN/m of pure water to <10 mN/m.
f,g
(Pay
attention to the definition of surface tension.
h
)
About 65% by weight of the surfactant is lecithin,
mostly dipalmitoylphosphatidylcholine (see Fig.
8-4), a phospholipid resistant to attack by oxygen.
Phosphatidylglycerol, in an unusually high concen-
tration, accounts for ~ 12% of the human surfactant.
Other phospholipids, plasmalogen,
i
cholesterol,
proteins, and calcium ions are also present.
The surfactant contains four unique proteins,
designated SP-A, SP-B, SP-C, and SP-D.
c,e
The major
protein (SP-A) is a sialic acid-rich glycoprotein
derived from a 26-kDa peptide, which contains a
short collagen-like domain.
e,j,k
Like collagen, this
domain contains glycosylated hydroxyproline.
The C-terminal domain is a Ca
2+
-dependent C-type
lectin (Chapter 4), while the N-terminal domain is
involved in oligomer formation through disulfide
bridges. The overall structure is similar to that of
the complement protein C1q (Chapter 31).
e,j
Protein
D is also collagen-like
l
but evidently plays a very
different functional role than SP-A. The latter asso-
ciates with the major surfactant lipids but SP-D does
not. It does bind phosphatidylinositol
m
and gluco-
sylceramide, lipids that are present in small amounts.
Perhaps SP-D helps to remove these polar lipids
which might interfere with surfactant action.
e
Both
proteins A and D may also have functions in the
immune system.
l
Proteins SP-B and SP-C are small extremely
hydrophobic polypeptides consisting of 79 and
35 amino acid residues, respectively.
n,o
Aliphatic
branched amino acids constitute 23 of the 35 residues
of the C-terminal part of protein C, which is also
palmitoylated on two cysteine residues. SP-B is
formed from a large 381-residue precursor. The
mature protein contains seven cysteines and disulfide
bridges. Both proteins have major effects on the
properties of the surfactant mixture. They promote
rapid reorganization of lipid layers, an important
consideration for the functioning of the surfactant.
Infants lacking SP-B suffer severe respiratory failure
with high mortality.
e
The properties of the surfactant allow for rapid
formation of a large area of lipid monolayerair
interface. The low surface tension and the ability
to rapidly spread the mixture of lipids and proteins
are essential.
o
At the end of the expiration stage of
breathing the surfactant is present in the interface as
a strong, tightly packed monolayer whose properties
reflect the rigidity of the dipalmitoylphosphatidyl-
choline. The excess surfactant in the alveolar fluid
forms liposome-like bilayer structures and also
associates with proteins and calcium ions to form a
lattice-like material called tubular myelin. Lipid
in this form must be transferred rapidly into the air
liquid interface during inspiration.
p
The ability of
phospholipids to pass through a hexagonal phase
(Fig. 8-12) may also be important for this transition.
q,r
One enzyme present in the surfactant fluid is an
acid phosphatase able to hydrolyze phosphatidyl-
glycerol phosphate, perhaps functioning in the final
step of biosynthesis of the phosphatidylglycerol
present in the surfactant.
c
Study of the action of the
natural lung surfactant has led to development of
artificial surfactant mixtures that are being used
effectively to save many lives.
d
a
Rooney, S. A. (1979) Trends Biochem. Sci. 4, 189191
b
Persson, A., Chang, D., Rust, K., Moxley, M., Longmore, W.,
and Crouch, E. (1989) Biochemistry 28, 63616367
c
Rooney, S. A., Young, S. L., and Mendelson, C. R. (1994) FASEB J.
8, 957 967
d
Jobe, A. H. (1993) N. Engl. J. Med. 328, 861 868
e
Kuroki, Y., and Voelkers, D. R. (1994) J. Biol. Chem. 269, 25943
25946
f
Shiffer, K., Hawgood, S., Haagsman, H. P., Benson, B., Clements,
J. A., and Goerke, J. (1993) Biochemistry 32, 590597
g
Pastrana-Rios, B., Flach, C. R., Brauner, J. W., Mautone, A. J.,
and Mendelsohn, R. (1994) Biochemistry 33, 51215127
h
Bangham, A. D. (1992) Nature (London) 359, 110
i
Rana, F. R., Harwood, J. S., Mautone, A. J., and Dluhy, R. A.
(1993) Biochemistry 32, 2731
j
McCormack, F. X., Calvert, H. M., Watson, P. A., Smith, D. L.,
Mason, R. J., and Voelker, D. R. (1994) J. Biol. Chem. 269, 5833
5841
k
Taneva, S., McEachren, T., Stewart, J., and Keough, K. M. W.
(1995) Biochemistry 34, 1027910289
l
Crouch, E., Persson, A., Chang, D., and Heuser, J. (1994) J. Biol.
Chem. 269, 1731117319
m
Ogasawara, Y., Kuroki, Y., and Akino, T. (1992) J. Biol. Chem.
267, 2124421249
n
Korimilli, A., Gonzales, L. W., and Guttentag, S. H. (2000) J.
Biol. Chem. 275, 8672 8679
o
Pastrana, B., Mautone, A. J., and Mendelsohn, R. (1991)
Biochemistry 30, 1005810064
p
Lipp, M. M., Lee, K. Y. C., Zasadzinski, J. A., and Waring, A. J.
(1996) Science 273, 11961199
q
Perkins, W. R., Dause, R. B., Parente, R. A., Minchey, S. R.,
Neuman, K. C., Gruner, S. M., Taraschi, T. F., and Janoff, A. S.
(1996) Science 273, 330332
r
Discher, B. M., Maloney, K. M., Grainger, D. W., Sousa, C. A.,
and Hall, S. B. (1999) Biochemistry 38, 374383
387
The plasma membrane of mammalian male germ cells
contains the following sulfogalactosylglycerolipid.
It is found only in spermatozoa and testes, in which it
accounts for 5 8% of total lipid, and in the brain, in
which it accounts for only 0.2% of total lipid.
31
A variety of acylated glucolipids and
phosphoglucolipids, including monoglucosyl
and diglucosyl diacylglycerols, have been
identified in membranes of the cell-wall-less
bacteriumAcholeplasma laidlawii.
32,32a
The
following glycolipid from the methanogen
Methanosarcina
33
is identical to the core
structure of eukaryotic glycosylphospha-
tidylinositol membrane protein anchors
(Fig. 8-13).
In addition to the previously mentioned phytanyl ether
phospholipids, methanogens contain diphytanyl
tetraether lipids that are both glycerophospholipids
and glycolipids.
O
OH
H
2
C
HO
O
O
3
SO
OH
H
2
C
C
CH
2
H
O
O
R'
C
R
O

Sulfogalactosylglycerolipid, a sulfate ester


resistant to phospholipase A
1
as well. These two pro-
perties appear to protect the naked cell membranes of
the protozoa from their own phospholipases which
may be secreted into the environment.
26
Marine algae
form an arsenic-containing phospholipid O-phospha-
tidyltrimethylarsonium lactic acid.
27
They apparently do this as part of a scheme for detoxi-
fying arsenate taken up with phosphate from the
phosphate-poor ocean water.
4. Glycolipids
The polar heads of the glycoglycerolipids lack
phospho groups but contain sugars in glycosidic link-
age.
28
Large amounts of the galactolipids shown in the
following structure are found in chloroplasts.
29
The
monogalactosyl diacylglycerol is said to be the most
abundant polar lipid in nature.
Marine algae as well as aquatic higher plants accumu-
late arsenophospholipids.
30
O
CH
2
As O
CH
2
CH
3
HO OH
O
CH
2
C
CH
2
H
HO
O
P
O
CH
2
C
H
2
C
O
O
O
H
OH
C O
R
1
+

O
H
2
N
O
CH
2
OH
HO
HO
HO
OH
HO
O HO
P O

O
O
diphytanylglycerol
myo-Inositol
1
6
4
d-Glucosamine
A. Lipid Structures
O
C
O
CH
2
C
O
C
O
H
2
C
H
O
OH
CH
2
HO
O HO
OH
A second galactose
may be added here
A galactolipid of chloroplaststypically 96%
of the fatty acyl groups are from linolenic acid
As
CH
2
C
OH
H
H
3
C
H
3
C
H
3
C COOH
+
Trimethylarsonium lactic acid
Arsenophospholipid of aquatic plants
O
C
O
CH
2
C
O
C
O
CH
2
H
O
OH
CH
2
O
HO
O
3
S
HO
R
1
R
2
Sulfolipid of
chloroplasts
6-Sulfo-6-deoxy--d glucopyranosyl diglyceride

Chloroplasts also contain the following sulfolipid, an


anionic sulfonate.
388 Chapter 8. Lipids, Membranes, and Cell Coats
5. Sphingolipids
The backbone of the sphingolipids is the basic alco-
hol sphingosine (sphingenine) or a related long chain
base. At least 60 such bases have been identified.
34,35
They vary in chain length from C
14
to C
22
and include
members of the branched iso and anteiso series. Up
to two double bonds may be present. Sphingosine
contains 18 carbon atoms and is formed from palmitic
acid and serine (Fig. 21-6). An intermediate in the
formation of sphingosine is the saturated sphinganine
(dihydrosphingosine), which is also a common com-
ponent of animal sphingolipids. Hydroxylation of
sphinganine to phytosphingosine occurs in both plants
and animals, especially within glycolipids. The name
comes from the fact that phytosphingosine was first
discovered in plants.
Sphingosine-containing lipids are classified as
sphingophospholipids (sphingomyelins) and
sphingoglycolipids. In both cases the sphingosine is
combined in amide linkage with a fatty acid to form a
ceramide (Fig. 8-3) which still contains a free hydroxyl
group able to combine with another component. In the
sphingomyelins, which were first isolated from human
brain by Thudicum in 1884, the additional component
is usually phosphocholine (Fig. 8-3). Ceramide amino-
ethylphosphonates and related glycolipids occur in
some invertebrates.
36,37
The cerebrosides are glycosides of ceramide
containing galactose or glucose. They are found in
relatively large amounts in the brain where monoga-
lactosylceramide predominates. Cerebrosides also
occur in other animal tissues and to a lesser extent in
plants. Many glycosphingolipids contain oligosaccha-
rides of various sizes. When the oligosaccharide
contains one or more residues of sialic acid the com-
pound is known as a ganglioside.
38
Sialic acids are
never found in plants. However, plants and fungi
contain phytoglycolipids, which resemble ganglio-
sides. Some contain inositol phosphates as well as
sugars.
39 41
The structures of gangliosides may be very
complex. Like glycoproteins, these substances are often
located at the outer surface of cells where they may act
as receptors for toxins, viruses, and hormones (see
Section D).
42
Some of them carry attached blood
group antigens (Box 4-C).
43,44
The sulfate esters of
cerebrosides, known as sulfatides (Fig. 8-3), are also
important components of membranes.
45
Gliding bacteria such as Capnocytophaga contain
sulfonolipids:
46
O
O
O
O
C
CH
2
CH
2
O
H
PO
3

-glycerol
C
H
H
2
C
H
2
C
Glcp12Galf-O
(sn-glycerol3-P)
From a methanogenic bacterium.
21
O
3
S
CH
2
C
NH
C
R O
H OH
H

A 1-deoxy-1-sulfonate analog of ceramides from


gliding bacteria. R is a long-chain alkyl group.
HOH
2
C
NH
2
H
HO
H
CH
2
NH
2
H
C
14
H
29
HO
CH
2
NH
2
HO
H
C
14
H
29
HO
H
H
OH
HO
H
Sphingosine (sphingenine)
Sphinganine (dihydrosphingosine)
Phytosphingosine
N-acylcapnine
A dipthytanyl tetraether lipid
389
These are analogs of the ceramides. The sulfono-
lipids seem to be necessary for the gliding movement
of these bacteria across solid surfaces.
47
6. Sterols and Other Isoprenoid Lipids
A large group of isoprenoid lipids, including
sterols, terpenes, and carotenoid compounds, are
often present in membranes or in extracted lipids.
Among these are the fat-soluble vitamins A, D, E, and
K and the high polymers rubber and gutta-percha.
Figure 8-3 Structures of some sphingolipids.
An OH group occurs here in some brain cerebrosides
Y = H
The compounds in which Y = H are ceramides.
Sphingolipids are often named as ceramide
derivatives.
Cerebrosides or ceramide mono- and oligosaccharides
The galactose bears a 3-sulfate group in cerebroside
sulfatides, e.g., in lactosyl ceramide sulfate
Present in sphingolipid of red blood cell membranes
Phosphocholine
O P
O
O
O CH
2
CH
2
N (CH
3
)
3
P
O
O
CH
2
CH
2
NH
3
Y =

+
+
Y =
Sphingomyelins
Ceramide aminoethyl phosphonates
Y = d-Gal or d-Gal14Glc
Y = GalNAc13Gal14Gal14Glc
O P
O
O
O Inositol Mannose

Y = Present in phytoglycolipid of yeast


C
N H
O
H
2
C
O
Y
H
H
OH
A. Lipid Structures
The phytyl group of chlorophyll (Fig. 23-30), the phy-
tanyl and diphytanyl groups of the lipids of methano-
gens, and the side chain of the pigment heme a (Fig.
16-5) are all related and are all derived from the pre-
cursor prenyl pyrophosphate (isopentenyl diphos-
phate). The whole group of compounds are often
referred to as polyprenyl or by the older designation
isoprenoid. The major discussion of polyprenyl
compounds is found in Chapter 22 but the role of
cholesterol and related compounds in membranes and
the function of polyprenyl groups as membrane anchors
for some proteins will be considered in this chapter.
390 Chapter 8. Lipids, Membranes, and Cell Coats
B. Membranes
In Chapter 7 we examined ways in which protein
subunits can be stacked to form helices and closed
oligomers. Another important arrangement of cell
constituents is that of flat sheets or membranes.
48 54
Chemists, physicists, and biologists have mounted a
sustained effort to understand these thin but remark-
ably tough outer surfaces of cells. However, consider
the fact that a 7 - to10-nm-thick plasma membrane of a
cell of 10-m diameter has less than 1/ 1000 the thick-
ness of the cell and occupies only 0.5% of the total
volume. The technical difficulties in studying such a
membrane are great and are compounded by the fact
that a cell contains more than one kind of membrane.
Membranes from many sources have been studied.
One of these is myelin, the multilayered insulation
that surrounds the axons of many nerve cells.
55 57
Myelin is derived from the plasma membrane of
Schwann cells which lie adjacent to many neurons.
Schwann cells literally wrap themselves around neu-
ronal axons. Their cytoplasm is squeezed out leaving
little but tightly packed membrane layers. Myelin
membranes are the most stable known and also have
the highest lipid content (80%). Another readily avail-
able experimental material is the plasma membrane
of the human red bood cell, which can be prepared
by osmotic rupture of the cells. The remaining
erythrocyte ghosts contain ~ 1% of the dry matter
of the cell and may have been studied more than any
other membrane. A much investigated specialized
membrane is the outer portion of the visual receptor
cells known as rods (Chapter 23), which contains a
closely packed and regular array of flat discs, each one
consisting of a pair of membranes. Both membranes
and cell walls of many kinds of bacteria have also been
investigated.
Prenyl diphosphate (pyro-
phosphate) (isopentenyl
pyrophosphate), precursor
of steroid,carotenoid, and
other isoprenoid substances Cholesterol, a sterol
-Carotene, a yellow pigment
of carrots and other plants;
converted in the human body
to vitamin A.
Notice the center of symmetry
in this 40-carbon molecule
derived from 8 prenyl units
CH
3
H
3
C
CH
3
CH
3
CH
3
CH
3
H
3
C CH
3
H
3
C
CH
3
H
3
C
H
2
C
CH
2
O
P
O

O
O
P
O O

OH
1. The Structure of Membranes
Membranes consist largely of protein and lipid.
The ratio (by weight) of protein to lipid varies from 0.25
in myelin to ~ 3.0 in bacterial membranes. In mem-
branes of erythrocytes it is about 1.2 and a ratio of
about 1.0 may be regarded as typical for animal cells.
Small amounts of carbohydrates (< 5%) are present, as
are traces of RNA (< 0.1%).
In 1926, Gorter and Grendel calculated that the
erythrocyte ghost contained just enough lipid to form
a 3.0- to 4.0-nm-thick layer around the cell. Apparently
they reached this correct conclusion only because their
measurements of pressures of surface films contained
compensating errors.
58
Nevertheless, this information,
together with the known propensity of lipids to aggre-
gate in micelles in which the hydrocarbon tails
clustered together and the polar heads protruded
into the surrounding water,
59
led Danielli and Davsen
in 1935 to propose the lipid bilayer structure for
membranes.
60
Its essential features are indicated in
Fig. 8-4. Hydrophobic bonding holds the extended
hydrocarbon chains together, while the polar groups
of the phospholipid molecules may interact with
proteins on the sides of the bilayer. The original pro-
posal assumed an extended structure for the proteins,
which would allow them to coat the bilayer uniformly
on both sides. However, this is not correct. Proteins
are sometimes embedded in the bilayer, sometimes
protrude through it, and sometimes are attached on
one surface, most often the cytoplasmic surface of
the plasma membrane. These concepts were brought
together by Singer and Nicolson in 1972 in the fluid
mosaic model of membrane structure
61
(Fig. 8-5).
The lipid bilayer still provides the basic structure upon
which the complex membranes of living organisms are
assembled.
52,54,62 65
The term fluid refers to the fact
CH
3
H
3
C
CH
3
H
H H
HO
H
3
C H
CH
3
391
~7 nm
~3 nm ~2 nm ~2 nm
Typical appearance
of osmium-stained
membranes in the
electron microscope
Cross-sectional
area of hydrocarbon
chain is 0.2 nm
2
Distearoyl phosphatidylcholine
O
P
O O
O
C
O

O
N
CH
3
H
3
C
H
3
C
O
O
C
2.0 nm
CH
3
CH
3
~4 nm
Schematic diagram of
phospholipid bilayer
Polar head
O
P O O
O
C
O

O
N
CH3
H3C
H3C
O
O
C
O
P O O
O
C
O

O
N
CH3
H3C
H3C
O
O
C
O
P O O
O
C
O

O
N
CH3
H3C
H3C
O
O
C
O
P O O
O
C
O

O
N
CH3
H3C
H3C
O
O
C
O
P O O
O
C
O

O
N
CH3
H3C
O
O
C
CH3
O
P O O
O
C
O

O
N
CH3
H3C
O
O
C
CH3
O
P O O
O
C
O

O
N
CH3
H3C
O
O
C
CH3
O
P O O
O
C
O

O
N
CH3
H3C
O
O
C
CH3
Figure 8-4 Bimolecular lipid layers and membranes. (Top) A molecule
of phosphatidylcholine. (Center) Lipid bilayer structure. (Bottom)
Bilayer structure as seen by the electron microscope with osmium
tetroxide staining.
Figure 8-5 The fluid mosaic model of Singer and Nicolson.
61
Some
integral membrane proteins, which are shown as irregular solids, are
dissolved in the bilayer. Transmembrane proteins protrude from both
sides. One of these is pictured as a seven-helix protein, a common
type of receptor for hormones and for light absorption by visual pig-
ments. Other proteins adhere to either the outer or the inner surface.
Many membrane proteins carry complex oligosaccharide groups which
protrude from the outer surface (Chapter 4). A few of these are indicated
here as chains of sugar rings.
Figure 8-6 (A) Molecular packing of 2,3-
dimyristoyl-D-glycero-1-phosphocholine dihy-
drate. The two molecules in the asymmetric
unit are labeled 1 and 2. The position of the
water molecules is indicated either by W1W4
or by small open circles. Hydrogen bonds are
represented by dotted lines. From Pascher et
al.
66
(B) Two-dimensional orthorhombic
arrangement of hydrocarbon chains in a crystal-
line alkane. The a b plane corresponds to the
plane of the bilayer surface; the long axes of the
acyl chains project from the page. From Cameron
et al.
67
7
1
2
3
4
5
6
7
1
2
3
4
5
6
S
S

H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
H
a
b
B. Membranes
A
B
392 Chapter 8. Lipids, Membranes, and Cell Coats
Figure 8-7 Atomic force microscope image of a dimyristoyl-
phosphatidylethanolamine bilayer deposited by the Langmuir
Blodgett technique (see Fig. 8-8) at a specific molecular area
of 0.41 nm
2
and a surface pressure of 40 mN/m on a freshly
cleaved mica substrate. The images were taken under water.
The long, uniformly spaced rows are roughly 0.7 0.9 nm in
spacing. The modulation along the rows, with rounded
bright spots roughly every 0.5 nm, corresponds to the indi-
vidual headgroups of the phosphatidylethanolamine mole-
cules. The lattice spacing is identical to that measured by
X-ray diffraction at the airwater interface. The area per
molecule in the AFM image is ~ 0.4 nm
2
. From Zasadzinski
et al.
68
that at temperatures suitable for growth and metabo-
lism the hydrocarbon chains are not rigidly packed in
the center of the bilayer but are molten (see Section 2).
However, at a low enough temperature they become
rigid and pack together in a manner similar to that of
the chains in crystals of the phosphatidylcholine shown
in Fig. 8-6A. These crystals consist of stacked bilayers
of thickness 5.5 nm.
66,69
The scanning tunneling and
atomic force microscopes have provided direct views
of a similar arrangement of side chains in a monomo-
lecular fatty acid layer
68,70,71
(Fig. 8-7). Measurements
on multilamellar vesicles of dipalmitoylphosphatidyl-
choline give bilayer thicknessess from 5.4 nm for dehy-
drated vesicles to 6.7 nm for the biologically relevant
fully hydrated bilayers.
72
Lipids of membranes. Approximately 1500
different lipids have been identified in the myelin of
the central nervous system of humans. About 30 of
these are present in substantial amounts.
73
The distri-
bution of the different lipids varies markedly between
membranes from different sources (Table 8-3) making
generalization difficult. However, phospholipids are
apparently always present and, except in chloroplasts,
make up from 40% to over 90% of the total lipid (Table
8-3).
Five kinds of phospholipid predominate: phospha-
tidylcholine, phosphatidylethanolamine, phosphatidyl-
serine, phosphatidylglycerols, and sphingomyelin.
Usually there are also small amounts of phosphatidyli-
nositol. The major phospholipid in animal cells is
phosphatidylcholine, but in bacteria it is phosphati-
dylethanolamine. The phospholipids of E. coli consist
of 80% phosphatidylethanolamine, 15% phosphati-
dylglycerol, and 5% diphosphatidylglycerol (cardio-
lipin). Significant amounts of cardiolipin are found
only in bacteria and in the inner membrane of mito-
chondria. Sphingomyelin is almost absent from mito-
chondria, endoplasmic reticulum, or nuclear membranes.
Glycolipids are important constituents of the
plasma membranes, of the endoplasmic reticulum,
and of chloroplasts. The cerebrosides and their sulfate
esters, the sulfatides, are especially abundant in myelin.
In plant membranes, the predominant lipids are the
galactosyl diglycerides.
29,74
The previously described
ether phospholipids (archaebacteria), ceramide amino-
ethylphosphonate (invertebrates), and sulfolipid
(chloroplasts) are also important membrane components.
Cholesterol makes up 17% of myelin and is present
in plasma membranes. However, it usually does not
occur in bacteria and is present only in trace amounts
in mitochondria. Related sterols are present in plant
membranes. Esters of sterols occur as transport forms
but are not found in membranes. Membrane bilayers,
likewise, contain little or no triacylglycerols, the latter
being found largely as droplets in the cytoplasm.
Quantitatively minor membrane components with
important biological functions include ubiquinone,
which is present in the inner mitochondrial membrane,
and the tocopherols. Plant chloroplast membranes
contain chlorophyll, carotenes, and other lipid-soluble
pigments.
Liquid crystals, liposomes, and artificial mem-
branes. Phospholipids dissolve in water to form true
solutions only at very low concentrations (~ 10
10
M for
distearoyl phosphatidylcholine). At higher concentra-
tions they exist in liquid crystalline phases in which
the molecules are partially oriented. Phosphatidylcho-
lines (lecithins) exist almost exclusively in a lamellar
(smectic) phase in which the molecules form bilayers.
In a warm phosphatidylcholinewater mixture con-
taining at least 30% water by weight the phospholipid
forms multilamellar vesicles, one lipid bilayer sur-
rounding another in an onion skin structure. When
such vesicles are subjected to ultrasonic vibration they
break up, forming some very small vesicles of diameter
down to 25 nm which are surrounded by a single
bilayer. These unilamellar vesicles are often used for
study of the properties of bilayers. Vesicles of both
types are often called liposomes.
75 77
When liposomes are stained with osmium tetroxide
or potassium permanganate, embedded, and sectioned
0.5 nm
393
for electron microscopy, their membranes show a
characteristic three layered structure similar to that
observed for biological membranes. Two darkly
stained lines ~ 2 2.5 nm thick are separated by a clear
space ~ 2.5 3.5 nm wide in the center. Both myelin
and the retinal rod outer segments show closely spaced
pairs of such membranes with a combined width of
18 nm. These results seemed to support the original
DavsenDanielli model. However, many questions
must be raised about the interpretation of these results.
Why does OsO
4
stain only the outer protein layer when
it is known to react also with double bonds of hydro-
carbon side chains of lipids to form osmate esters
which are readily reduced to a diol and osmium?
77a-c,78
Membranes from which most of the lipid has been
extracted still stain with OsO
4
to give three-layered
electron micrographs. Perhaps little can be concluded
from the three-layered appearance. We have learned
that it is difficult to determine even the thickness, let
alone the complete structure of an object that is only
6 10 nm thick.
Strong support for the lipid bilayer model comes
from the preparation of another type of artificial mem-
brane which can be made from a solution of phospha-
tidylcholine or of a mixture of phospholipids plus
cholesterol in a hydrocarbon solvent. A droplet of
solution is placed on a small orifice in a plastic sheet,
separating two compartments filled with an aqueous
medium (Fig. 8-8). The solution in the orifice quickly
drains, just as does a soap bubble, and the resulting
film eventually becomes so thin that the bright colors
disappear and a black membrane is formed. Similar
membranes, but without a residual content of hydro-
carbon solvent, have been created by apposition of
two lipid monolayers formed at an airwater inter-
face.
79,80
The thickness of such artificial membranes is
thought to be only 6 9 nm. Resilient and self-sealing,
the membranes can be stained with OsO
4
to give a
typical three-layered pattern.
B. Membranes
O
O
Os
O
O
O
O
O
Os
O
An osmate ester formed
from two unsaturated groups
Protein 22 59 60 76 75 48
Total lipid 78 41 40 24 25 52
f
Phosphatidylcholine 7.5 13 6.9 8.8
Phosphatidylethanolamine 11.7 6.5 6.5 8.4 18
Phosphatidylserine 7.1 2.5 3.1
Phosphatidylinositol 0.6 0.4 0.3 0.75
Phosphatidylglycerol 4
Cardiolipin
g
0.4 4.3 3
Sphingomyelin 6.4 0.5 6.5
Glycolipid 22.0 9.5 Trace Trace 23
Cholesterol 17.0 2.0 9.2 0.24
Total phospholipid 33 27 24 22.5 25 4.7
Phospholipid as a percentage 42 66 60 94 >90% 9
of total lipid
TABLE 8-3
Estimated Chemical Compositions of Some Membranes
Percentage of total dry weight of membrane
a
a
Dewey, M. M. and Barr, L. (1970) Curr. Top. Membr. Transp. 1, 6.
b
Kaback, H. R. (1970) Curr. Top. Membr. Transp. 1, 35 99.
c
Mizushima, S. and Yamada, H. (1975) Biochim. Biophys. Acta. 375, 44 53 .
d
Yamato, I. Anraku, Y. and Hirosawa, H. (1975) J. Biochem. (Tokyo) 77, 705 718. These investigators found 67% protein, 21% lipids, 10%
carbohydrate, and 2% RNA.
e
Lichtenthaler, H. K. and Park, R. B. (1963) Nature (London) 198, 1070 1072.
f
About 14% is accounted for by chlorophyll, carotenoids, and quinones.
e
g
Diphosphatidylglycerol (Fig. 8 -2).
Compound
Myelin
(bovine)
Retinal
rod
Plasma
membrane
(human
erythrocyte)
Mitochondrial
membranes
E. coli
b,c,d
(inner and outer
membranes) Chloroplasts
e
394 Chapter 8. Lipids, Membranes, and Cell Coats
The study of monolayers formed on a water sur-
face has also provided important information. A thin
film of an amphiphilic (containing both polar and non-
polar groups) compound such as a fatty acid is prepared.
This is done by depositing a small quantity of the
compound dissolved in a volatile solvent on a clean
aqueous surface between the barriers of a Langmuir
trough (Fig. 8-8).
81,82
The difference in surface tension
() across the barriers is measured with a suitable
device
81
for different areas of the monolayer, i.e., for
different positions of the moveable barrier. The value
of is low for expanded monolayers and falls to nearly
zero when the surface is no longer completely covered.
The pressure reaches a plateau when a compact mono-
layer is formed, after which it rises again (Fig. 8-8B).
At very high values of the monolayer collapses
(buckles). Both the cross-sectional area per molecule
in the monolayer and the collapse pressure can be
determined. For typical fatty acids, regardless of chain
length, the area covered is only ~ 0.2 nm
2
per molecule
indicating that the fatty acid chains are stacked verti-
cally to the surface in the monolayer. The collapse
Figure 8-8 (A) The LangmuirAdam film balance. Ten-
sion on the moveable barrier is recorded for different areas
of the surface between the barriers. This gives the surface
pressure , which is the difference between the surface
tension (
o
) of a clean aqueous surface and that of a spread
monolayer (): =
o
. Courtesy of Jones and Chapman.
81
(B) Surface pressure () area per molecule isotherm for a
typical fatty acid (e.g., pentadecanoic acid C
14
H
29
CO
2
H) at
the aqueous air interface. From Knobler.
81a
Moveable
barrier
Fixed
barrier
Aqueous phase
Trough

0

A
B
Solid
Condensed
Expanded
0.2 0.3 0.4 0.5 0.6 0.7 0.8
nm
2
/molecule


(
m
N
m
-
1
)
pressure is higher for longer molecules as a result of the
greater number of van der Waals interaction between
the chains. LangmuirBlodgett layers are prepared
by transferring one or more monolayers onto a smooth
solid surface (Fig. 8-7).
82,83
Physical properties of membrane lipids. A
completely extended C
18
fatty acid chain as shown in
Fig. 8-4 has a length of ~ 2.0 nm and occupies, either in
crystals or in monolayers, when viewed end-on, an
area of ~ 0.2 nm
2
. The hydrocarbon layer in a lipid
bilayer containing such chains would have a thickness
of about 4.0 nm; that determined by X-ray diffraction
for myelin is ~ 3.5 nm. However, for artificial black
membranes the thickness of the hydrocarbon layer can
be as little as 3.1 nm when all solvent is removed.
84
These and many other results
85
indicate that the hydro-
carbon chains are to some extent folded and that the
membrane is expanded over that expected according
to the simplest model.
Structure determinations on crystalline alkanes
confirm that the chains exist in a completely extended
conformation and that adjacent chains often pack
together in the orthorhombic arrangement shown in
Fig. 8-6B. As the temperature of such crystals is raised
a series of solidsolid phase transitions is observed
below the melting point of the crystals.
86
These can be
detected by changes in the infrared absorption spectrum
or by small amounts of heat absorption revealed by
differential scanning calorimetry (Fig. 8-9). Each
new phase permits a greater degree of mobility for the
hydrocarbon chains. Thus, at a high enough tempera-
ture but below the melting point, the chains are able
to rotate freely about their own axes in a so-called
hexagonal phase. Now the chains are packed in a
hexagonal array instead of the orthorhombic array of
Fig. 8-6B. At intermediate temperatures, some of the
chains may assume nonplanar conformations and
changes in the tilt of the hydrocarbon chains (Fig. 8-6)
may occur.
Similar phase transitions are observed for bilay-
ers.
88 90
For dipalmitoyl phosphatidylcholine the first
detectable subtransition
91
is centered at a temperature
T
s
of 18C. The second, known as the pretransition,
occurs at 35C (T
p
). The structure below T
s
may be
described as rigid or crystalline and that above T
s
as
a gel in which the hydrocarbon side chains twist and
turn much more freely but in which the orthorhombic
packing is maintained.
86
Above T
p
the head groups
become disordered. Although the orthorhombic pack-
ing of the tails may be maintained, there are several
distinct phases,
92,93
including one or more in which the
gel is thought to assume a structure analogous to that
in the hexagonal phase of hydrocarbons. At 41C the
main transition occurs.
395
Figure 8-9 Differential scanning calorimetric curves for 1-stearoyl-2-
linoleoyl-sn-glycerol. (A) Crystals of the compound grown from a hexane
solution were heated from 10 to 35C at a rate of 5C per minute and the
heat absorbed by the sample was recorded. (B) The molten lipid was
cooled from 35 to 10C at a rate of 5 per minute and the heat evolved
was recorded as the lipid crystallized in the phase and was then trans-
formed through two sub- phases. (C) The solid was reheated. From Di
and Small.
87
Courtesy of Donald M. Small.
10 0 10 20 30
Temperature (C)
sub
2
sub
1
sub
2 sub
1
Crystallization
Melt
' Melt
(a)
(b)
(c)
Cooling
Heating
Heat
absorbed
Heat
absorbed
Heat
evolved
Reheating
This is a sharper transition with a well-defined melting
temperature designated T
m
. Above T
m
the lipid is in
the lamellar liquid crystalline or L

state. The bilayer


continues to hold together, but the fatty acid chains
have melted and are now free to rotate and undergo
twisting movements more freely than at lower temper-
atures (Fig. 8-11). The main transition is highly, but
not completely, cooperative. Thus, the melting of the
membrane occurs over a range of several degrees.
The presence in biological membranes of a variety of
different components containing a variety of fatty acid
chains leads to a broadening of the melting range.
The behavior of bilayers is strongly influenced
by the lipid composition. Phospholipids containing
saturated, long-chain fatty acids have high transition
temperatures. The presence of unsaturated fatty acyl
groups with cis double bonds in membrane lipids
encourages folding of the hydrocarbon chains and
lowers T
m
. Even a single double bond lowers T
m
, the
decrease being greatest when the double bond is near
the center of the chain.
94 96
While T
m
for dipalmitoyl
phosphatidylcholine is 41C, that of 1,2-dipalmitoyl
phosphatidyl-sn-glycerol, which lacks the phospho-
choline head group, is 70C. This falls to 11.6C for
the polyunsaturated 1-stereoyl-2-linoleoyl-sn-glycerol,
whose melting curve is shown in Fig. 8-9.
87
This lipid
also shows a complex phase behavior and a melting
point for the stable, crystalline phase higher than
that of the phase.
Inclusion of other molecules of irregular shape
within membranes also lowers T
m
. However, a mole-
cule of cholesterol can pack into a bilayer with a cross-
sectional area of 0.39 nm
2
, just equal to that of two
hydrocarbon chains.
49
It tends to harden membranes
above T
m
but increases mobility of hydrocarbon chains
belowT
m
.
97 100
A complex of cholesterol and phospha-
tidylcholine may form a separate phase within the
membrane.
101,102
The ether-linked plasmalogens may
account for over 30% of the phosphoglycerides of the
white matter of the brain and of heart and ether linked
phospholipids are the major lipids of many anaerobic
bacteria.
103
Their T
m
values are a few
degrees higher than those of the corre-
sponding acyl phospholipids.
104
Between the pretransition temper-
ature and T
m
solid and liquid regions
may coexist within a bilayer.
101
The
termlateral phase separation has been
applied to this phenomenon.
105,106
Since changes in the equilibrium be-
tween solid and liquid can be induced
readily, e.g., by changes in the ionic
environment surrounding the bilayer,
lateral phase separation may be of
significance in such phenomena as
nerve conduction.
107
The phase transitions in bilayers
can be recognized in many ways.
Differential scanning colorimetry has
already been mentioned. Another
approach is to measure the spacing
between molecules by X-ray diffrac-
tion. The cross-sectional area occupied
by a phospholipid in a bilayer is
always greater than the 0.40 nm
2
expected for closest packing of a pair
of extended hydrocarbon chains.
39,85
High temperature
Low temperature
Liquid crystalline, partially ordered
disordered chains, biologically relevant
Gel phase, orthorhombic,
polar head groups disordered
Sub-gel, interdigitated
Crystalline phase, dehydrated highly
ordered all-trans extended chains
P

(Gr) Rippled;
G
h
, Hexagonal
G
o
, Orthorhombic, like alkanes
L

L
'
(G
d
)
T
m
41C
L

L
C
(G
S
)
T
P
35C
T
S
18C
B. Membranes
396 Chapter 8. Lipids, Membranes, and Cell Coats
bilayer into two or more phases can be observed using
2
H- or
31
P- NMR.
118 120
The orientation and dynamic
behavior of various head groups has been explored,
110,121
as have effects of mixing into the bilayer other lipids
such as glycosphingolipids
122
and cholesterol.
123,124
Crystalline phospholipids are being investigated by
solid-state NMR.
125
Fourier transform infrared spectroscopy
126,127
also
provides information about conformation of both
hydrocarbon chains and head groups. EPR spectros-
copy (Box 8-C) with doxyl probes on carbon atoms at
different depths within the bilayer has also been em-
ployed.
128
In recent years molecular dynamics simulations
have been used to predict behavior of membranes. As
is indicated in Fig. 8-11, the molten interior of the liquid
crystalline L

state is portrayed clearly.


129 131
In the gel
state the hydrocarbon chains maintain a closer packing
and undergo coordinated movement.
88
It is difficult to
know how realistic the simulations are. To calibrate
the method efforts are made to correctly predict a series
of known properties such as density and area per lipid
(0.61 nm).
130
Functions of phospholipid head groups. The
dipolar ionic head groups of phosphatidylcholine and
phosphatidylethanolamine occupy about the same
cross-sectional area as the two hydrocarbon tails.
Thus, they are in rather close contact with each other.
In crystals chains of hydrogen-bonded atoms may be
formed. In phosphatidylethanolamine the phosphate
and NH
3
+
ions may alternate in these chains.
132
In phosphatidylcholine, in which the nitrogen is sur-
rounded by methyl groups and cannot form this kind
of chain, water molecules bridge between the phos-
phates but the positive charges still interact with the
adjacent negative charges.
The chains of hydrogen bonds between the head
groups of phosphatidylethanolamine help to stabilize
the bilayer and are apparently responsible for the
elevation of T
m
by 10 30 above that observed for
phosphatidylcholine.
132
In contrast, the negatively
charged carboxyl groups of phosphatidylserine make the
membrane less stable. The melting point is increased
if the pH is lowered, protonating these groups. Their
presence also makes the membrane sensitive to the
concentration of cations.
133
The same is true of phos-
phatidylglycerol, whose head group contains a
negatively charged phosphate without an attached
BelowT
m
the spacing between chains is about 0.42 nm
corresponding to close packing of the fatty acid chains
in a hexagonal array with an area per phospholipid of
0.41 nm
2
. As the temperature is raised above T
m
the
spacing increases
85
to give an average area per phos-
pholipid of 0.640.73 nm
2
. Another technique (Box 8-C)
is to study a spin label by EPR while yet another is to
observe the fluorescence of a polarity-dependent
fluorescence probe such as N-phenylnaphthylamine
or other fluorescent probes
108
(see Chapter 23). The
compound is incorporated into the membrane and
undergoes changes in the intensity of its fluorescence
when the state of the membrane is altered.
A variety of NMR techniques are being applied
109 113
both to liposomes and to natural membranes.
111,114
Incorporation of
13
C or
2
H into various positions in the
hydrocarbon chains has allowed measurements of the
relative degree of mobility of the chains at different
depths in the bilayer (Fig. 8-10).
109,115 117
The results
are in agreement with statistical mechanical predictions
that configurational freedom increases with depth
toward the midplane of the bilayer. Separation of a
H N
H
H O
P
O
O
O
Glycerol
H N
H
H O
P
O
O
O
Glycerol
H N
H
H
+ + +
Figure 8-10
2
H NMR spectra of dimyristoyl phosphatidyl-
choline-d
27
/water in lamellar phases at 40C. One chain of
the phosphatidylcholine is fully deuterated, containing 27
atoms of
2
H. The mole ratios of water to lipid were 5.0 in (A)
and 25.0 in (B). The average interfacial areas per alkyl chain
as measured by X-ray diffraction were 0.252 nm
2
for (A) and
0.313 nm
2
for (B).
2
H NMR spectra are presented as powder
patterns because the lipid molecules are randomly oriented
in the magnetic field of the spectrometer as if in a powder.
This gives rise to pairs of peaks symmetrically located on
both sides of the origin. The separation distances are a
measure of the quadrupole splitting of the NMR absorption
line caused by the
2
H nucleus. The various splittings of the
resonances of the 13 CH
2
and one CH
3
groups reflect
differences in mobility.
109
The peaks have been assigned
tentatively as indicated. From Boden, Jones, and Sixl.
115
Courtesy of N. Boden.
3-9
10
11
12
13 2
2
14
14
2
2
13
12
11
10
3-9
3-8
9
10
11
12
2
13
14 14
12
10
9
3-8
2
13
2
2
11
10 kHz
A
B
397
counterion. Addition of calcium ions increases T
m
greatly and causes either phosphatidyglycerol or
phosphatidylserine to form a separate phase with a
more crystalline-like packing of the hydrocarbon side
chains.
134
Hydrogen bonding between head groups
also occurs with glycolipids.
135
Non-bilayer structures of phospholipids.
Under appropriate conditions some aqueous phospho-
lipids can exist in non-bilayer phases, a fact that may
be of considerable biological importance.
119,136,137
In
the presence of Ca
2+
some pure phospholipids can be
converted to the inverted hexagonal or H
II
phase
(Fig. 8-12).
136,138 140
In this phase the phospholipid
heads are clustered together in cylindrical inverted
micelles which pack in a hexagonal array. The ease
with which this transition can occur is increased by the
presence of small amounts of diacylglycerols or lyso-
lecithins.
141
Some lipids, such as the galactosyldia-
cylglycerol of chloroplasts, do not form bilayers but
prefer the hexagonal phase structure.
29,32a
This is
Figure 8-11 Results of simulated motion in a lipid bilayer
consisting of 64 molecules of dipalmitoylphosphatidylcho-
line and 23 water molecules per lipid at a pressure of 2 atm
and 50C. The view is that observed after 500 ps of simula-
tion. Bold lines represent the head group and glycerol parts
of the structures and the thin lines the hydrocarbon chains.
The gray spheres represent water molecules. From Berger,
Edholm, and Jhnig.
130
Courtesy of Dr. Olle Edholm.
thought to be a result of very high curvature of a
bilayer that arises from the sizes and packing of their
head groups. Another phase, even though it is liquid,
has a three-dimensional cubic symmetry.
142 144a
It
apparently consists of a complex arrangement of
polyhedral bilayer surfaces with interpenetrating
water channels between them.
143
Membrane fluidity and life. In agreement with
the known behavior of bilayers, the lipids of most
membranes in all organisms are partially liquid at
those temperatures suitable for life. Organisms have
developed at least three distinct means of ensuring
that membrane lipids remain liquid.
145
(1) In our
bodies (as well as in E. coli) the unsaturated fatty acids
that are present lower the melting point. Mutants of E.
coli that are unable to synthesize unsaturated fatty acids
cannot live unless these materials are supplied in the
medium.
146
(2) In Bacillus subtilis, which contains no un-
saturated fatty acids when grown at 37C, and in other
gram-positive bacteria, more than 70% of membrane
31
P NMR spectrum
Mol % egg PC
0
30
50
25 ppm
H
Phospholipid phase
?
Figure 8-12 (A)
31
P NMR spectra of different phospholipid
phases. Hydrated soya phosphatidylethanolamine adopts
the hexagonal H
II
phase at 30C. In the presence of 50 mol%
of egg phosphatidylcholine only the bilayer phase is observed.
At intermediate (30%) phosphatidylcholine concentrations
an isotropic component appears in the spectrum. (B) Inverted
micelles proposed to explain lipidic particles seen in freeze
fracture micrographs of bilayer mixture of phospholipids,
e.g., of phosphatidylethanolanine + phosphatidylcholine +
cholesterol. From de Kruijft et al.
119
Courtesy of B. de Kruijft.
B. Membranes
A
B
398 Chapter 8. Lipids, Membranes, and Cell Coats
BOX 8-C ELECTRON PARAMAGNETIC RESONANCE (EPR) SPECTRA AND SPIN LABELS
Unpaired electrons have magnetic moments and
are therefore suitable objects for magnetic resonance
spectroscopy. The technique is similar to NMR spec-
troscopy, but microwave frequencies of ~10
10
Hz are
employed, the energies being ~100 times greater than
those used in NMR.
a,b
Unpaired electrons are found
in organic free radicals and in certain transition metal
ions, both of which are important to many enzymatic
processes. Furthermore, spin labels in the form of
stable organic free radicals, can be attached to macro-
molecules at many different points.
a,c e
Coupling of
such artificially introduced unpaired electrons with
the magnetic moments of other unpaired electrons
or of magnetic nuclei can often be observed by EPR
techniques.
The conditions for absorption of energy in the
EPR spectrometer are given by the equation
h = gH
o
which is identical in form to that for NMR spectros-
copy. Here H
o
is the external magnetic field strength
and is a constant called the Bohr magneton. The
value of g, the spectroscopic splitting factor, is
one of the major characteristics needed to describe
an EPR spectrum. The value of g is exactly 2.000 for
a free electron but may be somewhat different in
radicals and substantially different in transition
metals. One factor that causes g values to vary with
environment is spin-orbit coupling which arises
because the p and d orbitals of atoms have directional
character. For the same reason g sometimes has three
discrete values for the three different directions (g
value anisotropy). The g value parallel to the direc-
tion of H
o
(g
||
) often differs from that in the perpen-
dicular direction (g

). Both values can be ascertained


experimentally.
A second feature of an EPR spectrum is hyper-
fine structure which results from coupling of the
magnetic moment of the unpaired electron with
nuclear spins. The coupling is analogous to the spin
spin coupling of NMR (Chapter 3). The hyperfine
splitting constant A, like the coupling constant J of
NMR spectroscopy, is given in Hertz. Splitting may
be caused by a magnetic atomic nucleus about which
the electron is moving or by some adjacent nucleus
or other unpaired electron. Sometimes important
chemical conclusions can be drawn from the presence
or absence of splitting. Thus, the EPR spectrum of a
metal ion in a complex will be split by nuclei in the
ligand only if covalent bonding takes place.
It is customary in EPR spectroscopy to plot
the first derivative of the absorption rather than the
absorption itself. Thus, for the paramagnetic nitroxide
2,2,6,6-tetramethylpiperidine-1-oxyl the EPR spec-
trum consists of three equally spaced bands whose
peaks are marked at the points where the steep
lines in the middle of the first derivative plots cross
the horizontal axis.
Coupling with the
14
N nuclear spin causes split-
ting into three lines as shown in the accompanying
figure.
This nitroxide is more soluble in liquid regions
of bilayers than it is in solid regions. As bilayers
are warmed in the EPR spectrometer, the solubility
of this spin-labeled compound in the lipid can be
followed (see figure). The lower of the three spectra
approximates that of the spin label in water alone,
while the others are composite spectra for which
part of the spin label has dissolved in the phospho-
lipid bilayers.
EPR spectrum of tetramethylpiperidine-1-oxyl dissolved
in an aqueous dispersion of phospholipids. (Top) above
the main bilayer transition temperature T
t
; (center) be-
tween T
t
and pretransition temperature; (bottom) below
pretransition temperature. From Shimshick and
McConnell.
f
N
O
2,2,6,6-Tetramethylpiperidine-1-oxyl
This "spin" label is often
attached by covalent linkage
at this point to larger molecules.
399
BOX 8-C (continued)
Since frequencies for EPR spectroscopy are ~100
times higher than those for NMR spectroscopy,
correlation times (Chapter 3) must be less than
~10
9
s if sharp spectra are to be obtained. Sharp
bands may sometimes be obtained for solutions, but
samples are often frozen to eliminate molecular
motion; spectra are taken at very low temperatures.
For spin labels in lipid bilayers, both the bandwidth
and shape are sensitively dependent upon molecular
motion, which may be either random or restricted.
Computer simulations are often used to match
observed band shapes under varying conditions with
those predicted by theories of motional broadening
of lines. Among the many spin-labeled compounds
that have been incorporated into lipid bilayers are
the following:
Much of the interpretation of the observed
changes in EPR spectra of spin labels is empirical.
For example, the spectra in the accompanying figure
can be interpreted to indicate that the spin label
dissolves in the lipid to a greater extent at higher
temperatures. The ratio f (defined in the figure) is
an empirical quantity whose change can be moni-
tored as a function of temperature. Plots of f vs T
have been used to identify transition and pretransi-
tion temperatures in bilayers.
f
EPR spectroscopy is used widely in the study of
proteins and of lipidprotein interactions.
c
It has
often been used to estimate distances between spin
labels and bound paramagnetic metal ions.
g
A high-
resolution EPR technique that detects NMR transi-
tions by a simultaneously irradiated EPR transition
is known as electron-nuclear double resonance
(ENDOR).
h
a
Berliner, L. J., and Reuben, J., eds. (1989) Spinlabeling. Theory and
Applications, Vol. 8, Plenum, New York ((Biological Magnetic
Resonance Series)
b
Cantor, C. R., and Schimmel, P. R. (1980) Biophysical Chemistry,
Freeman, San Francisco, California (pp. 525 536, 1352 1362)
c
Marsh, D. (1983) Trends Biochem. Sci. 8, 330 333
d
Esmann, M., Hideg, K., and Marsh, D. (1988) Biochemistry 27,
3913 3917
e
Millhauser, G. L. (1992) Trends Biochem. Sci. 17, 448 452
f
Shimshick, E. J., and McConnell, H. M. (1973) Biochemistry 12,
2351 2360
g
Voss, J., Salwinski, L., Kaback, H. R., and Hubbell, W. L. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 12295 12299
h
Lubitz, W., and Babcock, G. T. (1987) Trends Biochem. Sci. 12, 96
100
fatty acids contain methyl branches (Chapter 21)
147,148
which can decrease the melting point and increase the
monolayer surface area by a factor of as much as 1.5.
(3) Yet another mechanism for lowering the melting
point of fats is the incorporation of cyclopropane-
containing fatty acids (Chapter 21).
On the other hand, as we have already seen, cho-
lesterol tends to reduce the mobility of molecules in
membranes and causes phospholipid molecules to
occupy a smaller area than they would otherwise.
Myelin is especially rich in long-chain sphingolipids
and cholesterol, both of which tend to stabilize artificial
bilayers. Within our bodies, the bilayers of myelin tend
to be almost solid. Bilayers of some gram-positive
bacteria growing at elevated temperatures are stiffened
by biosynthesis of bifunctional fatty acids with co-
valently joined tails that link the opposite sides of
a bilayer.
149
Why must membrane lipids be mobile? One
reason is probably to be found in the participation of
membranes in many vital transport processes. Biologi-
cal membranes have a relatively high permeability to
neutral molecules (including H
2
O),
64,150
and it has been
suggested that above T
m
fatty acid chains are free to
rotate by 120 around single bonds from trans to gauche
conformations. When such rotation occurs about
adjacent, or nearly adjacent single bonds, kinks are
formed. If a kink originates near the bilayer surface,
as will usually be the case, a small molecule may jump
into the void created. Since the kink can easily migrate
through the bilayer, a small molecule may be carried
through with it.
151,152
The same factors may assist
larger protein molecules which function in membrane
transport. They probably also account for the substan-
tial degree of hydration of bilayers which involves
both the polar head groups and water diffusing through
the nonpolar interior.
153
Not only can molecules diffuse through membranes
but also membrane lipids and proteins can move with
respect to neighboring molecules. The rates of lateral
B. Membranes
OH
CH
3
CH
3
H
N
O
O
C
N
O
(CH
2
)
n
HOOC O
(CH
2
)
m
C H3
400 Chapter 8. Lipids, Membranes, and Cell Coats
diffusion of lipids in bilayers and of antigenic proteins
on cell surfaces are rapid.
80,154
If diffusion of phospho-
lipids is assumed to occur by a pairwise exchange of
neighboring molecules, the frequency of such exchanges
can be estimated
155
as ~ 10
7
s
1
. However proteins may
meet many obstacles to free diffusion.
156
Lateral diffu-
sion is often measured by the technique of fluorescence
recovery after photobleaching. One small spot in
a bilayer that contains a dye attached to a lipid or a
protein is bleached by a laser beam. Lateral diffusion
of nearby unbleached molecules into the bleached
spot can then be observed.
80
Lateral diffusion can also
be observed by NMR spectroscopy
157
and by single-
particle tracking.
158,159
In addition to diffusion there
may often be a flow of membrane constituents in
directions dictated by metabolism.
160
Although lateral
diffusion is fast a flip-flop transfer of a phospholipid
from one side of the bilayer to the other may require
many seconds.
161
However, a sudden increase in the
calcium ion concentration, an important intramolecular
signal (Chapter 11), activates a scramblase protein
which promotes a rapid transbilayer movement of
phospholipids.
162
Electrical properties of membranes. Biological
membranes serve as barriers to the passage of ions and
polar molecules, a fact that is reflected in their high
electrical resistance and capacitance. The electrical
resistance is usually 10
3
ohms cm
2
, while the capaci-
tance is 0.5 1.5 microfarad (F) cm
2
. The correspond-
ing values for artificial membranes are ~ 10
7
ohms cm
2
and 0.6 0.9 F cm
2
. The lower resistance of biological
membranes must result from the presence of proteins
and other ion-carrying substances or of pores in the
membranes. The capacitance values for the two types
of membrane are very close to those expected for a
bilayer with a thickness of ~ 2.5 nm and a dielectric
constant of 2.
54,84,163
The electrical potential gradient
is steep.
Outer cell surfaces usually carry a net negative
charge, the result of phosphate groups of phospholipids,
of carboxylate groups on proteins, and of sialic acids
attached to glycoproteins. This negatively charged
surface layer attracts ions of the opposite charge (coun-
terions), including protons, and repels those of the
same charge. The result is development of a diffuse
electrical double layer consisting of the fixed
negative charges on the surface and a positive ionic
atmosphere extending into the solution for a distance
that depends upon the ionic strength.
164 166
This ionic
atmosphere is analogous to that postulated by the
DebyeHckel theory (Chapter 6). At the physiological
ionic strength of 0.145 M the thickness of the double
layer, taken as the distance at which the electrical
potential falls to a certain fraction of that at the cell
surface,
164,165
is about 0.8 nm. However, the double-
layer thickness increases to about three times this
value at an ionic strength of 10
3
M and to still greater
distances at lower ionic strengths.
The net surface charge of a cell and the associated
electrical double layer are important in interactions
between cells and may influence the development of
extracellular structure such as basement membranes.
The net negative charge on cells also gives rise to an
experimentally measurable electrophoretic mobility.
A characteristic of living cells is the maintenance
of ionic gradients across the plasma membrane. Thus
almost all cells accumulate K
+
, even pumping it from
very dilute external solutions. Cells also exclude sodium,
pumping it out from the cytoplasm by mechanisms
considered in Section C,2. If a microelectrode is inserted
through a cell membrane and the potential difference
is measured between the inside and outside of the cell,
a resting potential which, in nerve cells, may be as
high as 90 mV is observed. The origin of the potential
appears to lie in the concentration differences of ions.
From the value of G for dilution of an ion (Eq. 6-25)
and the relationship between G and electrode poten-
tial (Eq. 6-63), the Nernst equation (Eq. 8-2) can be
derived. According to this equation, which applies to
a single ion for which the membrane is permeable,
a 10-fold concentration difference across the membrane
for a monovalent ion (n = 1) would lead to a 59-mV
membrane potential, E
m
. Since membranes are relatively
impermeable to sodium ions, it is generally conceded
that for many membranes the origin of the membrane
potential lies mainly with the potassium ion concen-
tration difference which is maintained by the Na
+
, K
+
-
ATPase (Section C). A more complete equation takes
account of K
+
, Na
+
, and Cl
-
together with their respec-
tive permeabilities.
167 169
Note also that Eq. 6-64 is also
often called the Nernst equation.
170
Protons are also pumped across cytoplasmic and
inner mitochondrial membranes, a topic of Chapter 18.
The flow of protons from inside to outside also contri-
butes to the membrane potential. The positive charges
of H
+
, K
+
, and other cations associated with the exter-
nal membrane surface are balanced by the negative
charges of protein molecules as well as Cl

and phos-
phate anions that are in or near to the inner surface of
the membranes.
Another possibility for proton flow has intrigued
biophysicists for years. Membranes often display a
substantial electrical conductivity in a lateral direction
along the membrane surface.
171 173
Electrical conduction
may involve movement of protons along hydrogen-
bonded lines, e.g., involving ethanolamine head
groups or phosphate groups and bridging water as
E
RT
nF
c
c n
c
c
m
at C =

ln
.
log
1
2
1
2
0 059
25
(8-2)
401
previously discussed (see also Eq. 9-96). Alternatively,
conduction may depend upon membrane-associated
proteins.
174
This lateral proton conduction may be
important to many proton-driven membrane processess,
such as rotation of bacterial flagella, ATP synthesis, and
pumping of ions (Chapter 18).
The two sides of a membrane. Many observa-
tions indicate great differences between the inside and
outside of the membranes that surround cells.
51,175,176
Bretscher and Raff
62
observed that, among the phos-
pholipids of the erythrocyte membrane, phosphatidyl-
choline predominates in many mammals but is replaced
by sphingomyelin in ruminants. Sheep erythrocytes
are resistant to cobra venom phospholipase A, which
is known to remove the fatty acid from the central
position on the glycerol of phosphatidylcholine, causing
lysis of the cells. The resistance of sheep erythrocytes
suggested that the sphingomyelin is on the outside of
the membrane while the phosphatidylethanolamine
and other phospholipids are inside. By inference,
phosphatidylcholine is also largely on the outside of
plasma membranes. Supporting this conclusion is the
observation that most of the reactive amino groups of
phosphatidylethanolamine and phosphatidylserine
are found on the inside (cytoplasmic) surfaces.
177
Since the total content of phosphatidylcholine and
sphingomyelin often exceeds that of phosphatidyletha-
nolamine and phosphatidylserine, the bilayer would
be incomplete on the inside of the membrane were it
not for the presence of proteins, which contribute more
to the inside than to the outside surface.
Glycolipids are usually on the outside of plasma
membranes with the attached sugar chains projecting
into the surrounding water. An important generalization
is that sugar groups attached either to lipids or to proteins
tend to be on outer cell surfaces or on materials that are
being exported from cells. An exception is found in the
abundant galactolipids of chloroplasts.
2. Membrane Proteins
The many proteins present within or attached to
membranes have a variety of functions. Some are
obviously structural, tying other proteins to a mem-
brane or providing a base for projecting fimbriae,
flagella, and other appendages. Some proteins of the
outer surface act as anchoring points for macromolecules
that lie between cells. The inner surfaces of membranes
are attached to the cytoskeleton (Chapter 7). Many
of the proteins embedded in membranes control the
passage of materials across membranes. Others serve
as receptors that sense the presence of specific com-
pounds or of light. Membranes may also contain
foreign proteins such as subunits of virus coats. Pro-
teins that are deeply embedded in membranes are
referred to as intrinsic or integral membrane
proteins. Proteins that are more loosely associated
with the membrane, principally at the inner surface,
are called peripheral.
61
Integral membrane proteins. Membrane pro-
teins are hard to crystallize
178
and precise structures
are known for only a few of them.
179 181
A large frac-
tion of all of the integral membrane proteins contain
one or more membrane-spanning helices with loops
of peptide chain between them. Folded domains in
the cytoplasm or on the external membrane surface
may also be present. The best-known structure of a
transmembrane protein is that of the 248-residue
bacteriorhodopsin. It consists of seven helical segments
that span the plasma membrane (Fig. 23-45) and serves
as a light-activated proton pump. Other proteins with
similar structures act as hormone receptors in eukary-
otic membranes. A seven-helix protein embedded in a
membrane is depicted in Fig. 8-5 and also, in more
detail, in Fig. 11-6.
The most hydrophobic integral membrane proteins
can be extracted into organic solvents such as mixtures
of chloroform and methanol. One such proteolipid
protein, the 23.5-kDa lipophilin, accounts for over half
the protein of myelin.
57,182
The purified protein from
rat brain contains 66% of nonpolar amino acids and six
molecules of covalently bound palmitic acid and other
fatty acids per peptide chain in thioester linkage to
cysteine side chains. This protein evidently has four
transmembrane helical segments with the six fatty acid
chains incorporated into the membrane bilayer. It also
has cytoplasmic and extracellular loops, one of which
binds inositol hexakisphosphate (Ins P-6). (Fig. 11-9).
183
The myelin proteolipid is an essential component of
the myelin sheath and defects in this protein are asso-
ciated with some demyelinating diseases
57
which are
discussed in Chapter 30.
There are many known topologies for helix-bundle
membrane proteins with the number of membrane-
spanning helices ranging from 1 to 14 or more e.g.,
see Fig. 8-23.
184 186
A topology can often be predicted
using suitable computer programs.
187 191
A first step
is to identify all sequences of 20 or more residues that
could form a helix sufficiently hydrophobic to allow
good nonpolar interactions with the bilayer core.
Sequences that can form amphipathic (amphiphilic)
helices must also be considered because two or more
of these can pack together in a membrane with their
hydrophilic sides together, sometimes forming pores
(see Section C,1).
192
Predictions of membrane protein structure are in
part based on the positive-inside rule which states
that positively charged lysine and arginine residues
will not pass through a membrane but will remain on
the negatively charged cytoplasmic surface. Often,
N-terminal parts of a protein will pass through a
B. Membranes
402 Chapter 8. Lipids, Membranes, and Cell Coats
membrane and will contain glutamate or aspartate
residues whose side chains carry negative charges.
These tend to remain outside of the membrane where
they are attracted to the positive charges on the mem-
brane outer surface. Positively charged residues may
interact with phosphate groups of phospholipids and
tyrosine and tryptophan side chains may interact with
the carbonyl groups of the ester linkages.
192a,b
After probable transmembrane helical regions have
been identified a residue-by-residue attempt can be
made to identify cytosolic and extracellular loops.
Chemical reactivities of the naturally occurring side
chains can also be examined. Residues within the
helix bundle will be protected. Mutations can be pre-
pared systematically by scanning mutagenesis. For
example, presumed extracellular loops in the erythro-
cyte band 3 protein (next section) have been mutated
by introduction of N-glycosylation acceptor sites (Asn-
X-Ser / Thr). If the sequence is at least 1214 residues
away from transmembrane sequences it will probably
be glycosylated in a suitable laboratory test system.
193
Within a transmembrane domain amino acid residues
can be systematically replaced with alanine (alanine
scanning mutagenesis) or with cysteine and effects on
the protein can be observed. Substitution with cys-
teine allows another possibility: Two cysteines on
adjacent transmembrane helices may be linked as
disulfides if they are close enough.
194,195
Discovery of
such neighboring pairs can be very valuable in at-
tempting to establish relationships of one helix to an-
other. A cloned gene can also be split into two pieces
prior to crosslinking of cysteine side chains. This may
facilitate mapping of tertiary interactions within trans-
membrane proteins.
196
Molecular dynamics methods
for modeling helix bundle proteins are also being
developed.
197
Not all integral membrane proteins have a helix
bundle structure. Some of the simplest transmembrane
proteins are the subunits of bacterial viruses and pili
(Figs. 7-7 and 7-9) The 7-nm -helical rods of phage
M13 have a 20-residue hydrophobic section (residues
25-46) which is preceded by a negatively charged se-
quence that appears to form a short amphipathic helix
that lies on the external surface of the membrane and
helps to anchor the subunit. A positively charged cluster
at the opposite end of the hydrophobic helical region
remains in the cytoplasm.
180,186,198
The porins, which
form pores in outer membranes of bacteria and ribosomes,
are large 16-strand cylinders (Section C,1).
179,180
It
has been suggested that this different basic architecture
may be related to the fact that the porin polypeptides
must be exported through the inner membrane before
being refolded in the periplasm.
199
Keep in mind that
a strand of nine residues can span the 33-nm bilayer
core just as well as can a 22-residue helix. Beta struc-
tures as well as shorter helices may well be present in
proteins that also contain membrane-spanning helices.
A third important structural pattern involves
extensive use of amphipathic helices that lie partially
embedded in a membrane surface. For example, the
blood lipoproteins are lipid particles partially coated
by amphipathic helices (Chapter 21).
200,201
Anchors for proteins. Proteins with membrane-
spanning sequences are usually anchored into the
membranes with the help of many polar side chains,
often including glycosylated residues, in the ends and
loops that protrude on the two sides of the membranes.
Other proteins are anchored by insertion into mem-
branes of nonpolar groups. These include fatty acyl
groups at N termini, fatty acid ester groups on serine,
threonine, or cysteine side chains, and polyprenyl groups
attached to cysteine side chains as thioethers.
202
Many
membrane-anchored proteins carry the saturated 14-
carbon myristoyl group in amide linkage with an
N-terminal glycine of the protein.
203 206
The fatty acid
chain is added at the time of the protein synthesis, i.e.,
cotranslationally. It can intercalate into the membrane
bilayer but provides a relatively weak anchor.
207
The
16-carbon palmitoyl group is typically added to a
cysteine thiol in recognition sequences at various
positions in a protein.
182,202,208,209
The modification
occurs after protein synthesis, i.e., posttranslationally.
Because thioesters are relatively unstable, palmitoy-
lation is regarded as a reversible modification.
210
A third lipid anchor is provided by the polyprenyl
farnesyl (15-carbon) and geranylgeranyl (20-carbon)
groups in thioether linkage to cysteine residues. These
must be present in specific recognition sequences at
the C termini of proteins, most often with the sequence
CAAX.
211 215
The prenylation (also called isoprenyla-
tion) reaction is followed by proteolytic removal of the
last three residues (AAX) and methylation of the new
C-terminal carboxyl group as is discussed in Chapter
11, Section D,3. See also Chapter 22, Section A,4.
A lipoprotein present in the periplasmic space of
E. coli is anchored to the outer bacterial membrane by
a triacylated modified N-terminal cysteine containing
a glyceryl group in thioether linkage as shown in the
following structure (see also Section E,1).
A related anchor that uses diphytanylglycerylation
is found in certain proteins of archaeobacteria.
216
N
N
O
O
S
O
O
O
O
COOH
H
H
Protein
403
A series of glycosylphosphatidylinositol (GPI)
anchors that are covalently linked to a variety of pro-
teins utilize diacylglycerol or alkylacylglycerol for
attachment to a bilayer. The proteins are joined through
their C-terminal carboxyl groups to the diacylglycerol
by a chain of covalently linked ethano-
lamine, phosphate, mannose, gluco-
samine, and myo-inositol as shown in
Fig. 8-13.
217 223
The proteins are linked
to the diacylglycerol through the
conserved structure: H
2
N-protein
ethanolamineP6Man12Man1
6Man14GlcNAcInsdiacylglycerol.
The structure in Fig. 8-13, which
anchors the small Thy-1 antigen to
surfaces of rat thymocytes,
224
contains
additional mannose, N-acetylgalac-
tosamine, and ethanolamine phos-
phate. These groups may be missing
or substituted by other groups in other
anchors.
225
For example, that of human
erythrocyte acetylcholinesterase lacks
the extra mannose and GalNAc of the
Thy-1 anchor but contains a palmitoyl
group attached to an oxygen atom of
the inositol. This provides an addi-
tional hydrocarbon tail that can enter
the bilayer.
218
Other PI-anchored
proteins include enzymes, such as
alkaline phosphatase and lipoprotein
lipase, and surface proteins of the
parasites Trypanosoma,
225
Leishmania,
and Toxoplasma.
220
Some adhesion
molecules and a variety of other outer
surface proteins are similarly attached
to membranes. Analysis of the genome
of Caenorhabditis elegans suggests that
the nematode contains over 40 GPI-
tailed proteins and perhaps more than
120.
225a
Analyzing erythrocyte mem-
branes. The proteins of red blood cell
membranes were among the first to be
studied. Because membrane proteins
are present in small amounts and tend
to be hard to dissolve without denatur-
ation, they have been difficult to study.
H
3
C N
N
O
O
S
O
O
COOH
H
H
Protein
B. Membranes
They usually do not dissolve readily in water, but red
cell membranes can be almost completely solubilized in
water using a 5 x 10
3
M solution of the chelating agent
EDTA (Table 6-9) or by 0.1 M tetramethyl ammonium
bromide.
226
These observations suggested that ionic
linkages between proteins, or between proteins and
phospholipids, are important to membrane stability.
Nonionic detergents such as those of the Triton X series
or -octylglucopyranoside also solubilize most mem-
brane proteins,
178,227 229
whereas ionic detergents such
as sodium dodecyl sulfate (SDS) often cause unfolding
and denaturation of peptide chains.
Gel electrophoresis of plasma membrane proteins
in SDS solution yields ~ 10 prominent bands and at
Figure 8-13 Structure of glycosylphosphatidylinositol (also called phospha-
tidylinositol-glycan) membrane anchors. The core structure is shown in black.
The green parts are found in the Thy-1 protein and / or in other anchors.
Man1
or
Acyl
Membrane
(1/2 thickness)
P
O
O

O NH
3
+
Man
Man
GlcN
Inositol
C O
GalNAc
O
O
CH
3
NH
HO
HO
HO
Man
Ethanolamine
1
2
(C-terminus)
HO
O
HO
O
P
O
O
O
N
O
O
H
HO
O
HO
O
O
O
O
HO
O
O
O
O
HO
H
2
N
OH
O
O
HO
HO
O HO
P
H
H
O O

1
C
2
O
1

P
O
O
NH
3
6

1
4
4
O
O 6
+

Protein
404 Chapter 8. Lipids, Membranes, and Cell Coats
Spectrin
Ankyrin
Pallidin
Dematin
Hemoglobin
72kDa
A B C D E
least 30 less intense bands ranging in molecular mass
from 10 to 360 kDa.
230,231
Proteins of erythrocyte ghosts,
and the usual system for numbering them, are shown
in Fig. 8-14. Some very important proteins known to
be present in this membrane, such as (Na
+
+ K
+
)-activated
ATPase (Section C,2), are found in such low quantities
(e.g., a few hundred molecules in a single red blood
cell)
62
that they do not show up in electropherograms.
Mitochondrial membranes appear to be more complex
than plasma membranes, but myelin has a somewhat
simpler composition.
Glycoproteins. Many of the integral proteins of
membranes are glycoproteins.
234 236
These may some-
times be recognized in electropherograms because they
are stained by the periodic acid-Schiff (PAS) procedure.
At least 20 glycoproteins are present in erythrocyte ghosts,
and glycoproteins appear to be prominent protein
components of the plasma membranes of all eukaryotes
and of primitive archaebacteria such as Halobacterium.
The most abundant glycoprotein of red blood cell
membranes is the 95-kDa PAS-reactive band 3 protein
(Fig. 8-14) which makes up ~ 25% of the total mem-
brane protein.
236 240
A variety of asparagine-linked
oligosaccharides based on the core hexasaccharide
structure shown in Chapter 4, Section D,2 are present
and apparently project into the surrounding medium.
Electron micrographs of freeze-fractured surfaces
through the membrane bilayer (Fig. 8-15) show ~ 4200
particles of 8 nm diameter per square micrometer,
randomly distributed and apparently embedded in
the membrane. These probably represent dimers of
the glycoprotein. The amino acid sequence of the
911-residue protein suggests 13 membrane-spanning
helices in the 550-residue C-terminal domain and that
the 41-kDa N-terminal domain projects inward into
the cytoplasm.
236,240
Among the first 31 residues are
16 of aspartate or glutamate. These provide a highly
negatively charged tail that is able to interact electro-
statically with other proteins.
236
Among these are com-
ponents of the cytoskeleton, which appears to be anchored
to the membrane via the band 3 protein. The band 3
protein is also a substrate for transglutaminase (Eq. 2-23)
which creates covalent crosslinks to other proteins.
237
Another major function of the band 3 glycoprotein is
to form channels for the transport of anions (Section C,2).
Another integral glycoprotein of erythrocytes, the
31-kDa glycophorin A (PAS-1),
235,241 244
is ~ 60% by
weight carbohydrate. Its 131-residue chain has a
single ~ 23-residue membrane-spanning helix. The
first 50 residues at the N terminus, which project from
the outside surface of the membrane, include many
serines and threonines. Their side chains carry 15
O-linked tetrasaccharides and one complex N-linked
oligosaccharide. There are a total of ~ 160 sugar residues
per peptide chain, largely N-acetylgalactosamine,
galactose, and sialic acid. Some of the oligosaccharides
contain MNO blood group determinants
234,241
(Box 4-C).
Because of a high content of sialic acid, glycophorin
also carries a large negative charge. The 35-residue
C-terminal domain is hydrophilic and rich in proline,
glutamate, and aspartate. It probably extends into the
cytoplasm and may bind calcium ions or interact with
NH
3
+
groups on phospholipid heads.
If all the sugar residues of the glycophorin mole-
cules in an erythrocyte were spread over the surface of
the cell they could cover approximately one-fifth of its
surface in a loose network. However, it is more likely
that they form bushy projections of a more localized
sort. These oligosaccharides not only act as immuno-
logical determinants but also serve as receptors for
influenza viruses. Other glycoproteins related to
glycophorin A occur in smaller amounts.
244
Figure 8-14 SDS-polyacrylamide gel electrophoresis of
human erythrocyte ghosts. (A) From untreated cells. (B) From
cells digested externally with chymotrypsin. (C) Inside-out
vesicles prepared from cells pretreated with chymotrypsin.
(D) The same inside-out vesicles after further treatment with
chymotrypsin. (E) Polypeptides released by the chymotryptic
treatment of the inside-out vesicles. The peptides are numbered
according to the system of Steck
232
; Hb, hemoglobin. From
Luna et al.
233
C O
CH
2
CH
2
O
H
C
CH
3
H
H
H
3
C
C H
3
C
H
3
C
H
3
C
n
Polyoxyethylene p-t-octyl phenol: the Triton X series of detergents.
n = 9 10 for Triton X-100 and 7 8 for Triton X-114.
405
Connections to the cytoskeleton. About one-
third of the protein of the red blood cell membrane is
accounted for by a pair of larger hydrophobic peptides
called spectrin with molecular masses of 280 kDa (
chain) and 246 kDa ( chain).
245 250a
These are found
in bands 1 and 2 of Fig. 8-14. The spectrin monomers
consist largely of 106- to 119- residue repeat sequences
each of which folds into a short triple-helical bundle
(Fig. 8-16B). The beaded-chain monomers associate
readily to dimers, long ~ 100 nm thin flexible rods
which associate further to ()
2
tetramers. The latter,
in turn, bind to monomers or to small oligomers of
actin. In red blood cells the actin crosslinks the spec-
trin tetramers into a two-dimensional fishnet (Fig. 8-
16A), the ~ 85,000 spectrin tetramers uniformly
covering the entire inner surface (130 m
2
) of the
erythrocyte.
245,251
The inner location of spectrin was
established by the fact that chemical treatments
252
that
covalently label groups on proteins exposed on the
outer surface of erythrocytes (e.g., iodination with
lactoperoxidase; Chapter 16) did not label spectrin.
257
Spectrin also binds to one domain of another large
215-kDa peripheral protein called ankyrin (band 2.1,
in Fig. 8-14) which anchors the spectrin network to the
membrane.
258,259
Ankyrin is actually a multigene
family of related proteins that are present in many
metazoan tissues.
258,260
These are modular proteins
Figure 8-15 Freeze-fractured membranes of two erythrocyte
ghosts. The upper fracture face (PF) shows the interior of
the membrane half closest to the cytoplasm. The smooth
region is lipid and contains numerous particles. The lower
face, the extracellular half (EF), possesses fewer particles.
The space between the two is nonetched ice. See Figs. 1-4
and 1-15 for electron micrographs of sections through bio-
logical membranes. Courtesy of Knute A. Fisher.
B. Membranes
with separate binding domains for spectrin and band
3 protein. The latter domain contains 24 ankyrin
repeats, 33-residue modules, also found in a variety
of other proteins.
258,259
Ankyrin binds firmly to the band 3 glycoprotein
which is embedded in the membrane. The 78 kDa
band 4.1 protein is another major component of the
erythrocyte membrane skeleton.
261,262
Protein 4.1
binds both to ankyrin and also to glycophorin C,
providing another anchor to an integral membrane
protein (Fig. 8-16C). Spectrin:actin:protein 4.1 in a
1:2:1 ratio are the major components of the membrane
skeleton.
262
Other less abundant proteins include
adducin, protein 4.2 (pallidin, which interacts with
band 3 protein),
263
protein 4.9 (dematin, an actin
bundling protein),
264
and the muscle proteins tro-
pomyosin and tropomodulin (Chapter 19). While
spectrin and ankyrin of erythrocyte membranes have
been studied most intensively, related proteins occur
in other cells.
265
Spectrin of brain and other tissues is
also known as fodrin.
266
Dystrophin and -actinin,
actin-crosslinking proteins of muscle, are also mem-
bers of the spectrin superfamily.
248
Protein 4.1 also
occurs in various organisms,
267
in various tissues, and
in various locations in cells.
268
What is the function of the membrane skeleton?
There is a group of hereditary diseases including
spherocytosis in which erythrocytes do not maintain
their biconcave disc shape but become spherical or
have other abnormal shapes and are extremely frag-
ile.
269 272
Causes of spherocytosis include defective
formation of spectrin tetramers and defective association
of spectrin with ankyrin or the band 4.1 protein.
265,273
Thus, the principal functions of these proteins in
erythrocytes may be to strengthen the membrane and
to preserve the characteristic shape of erythrocytes
during their 120-day lifetime in the bloodstream. In
other cells the spectrins are able to interact with micro-
tubules, which are absent from erythrocytes, and to
microtubule-associated proteins of the cytoskeleton
(Chapter 7, Section F).
270
In nerve terminals a protein
similar to erythrocyte protein 4.1 may be involved in
transmitter release.
274
The cytoskeleton is also actively
involved in transmembrane signaling.
Integrins and focal adhesions. Mature erythro-
cytes have no nucleus and lack the microtubules and
actin filaments that span other cells. In nonerythroid
cells the major connections of the cytoskeleton to
the membrane are through large heterodimeric
membrane-spanning proteins called integrins (Fig.
8-17). These proteins, as well as the ends of the actin
filaments, tend to be concentrated in regions long
observed and described by microscopists as focal
adhesions.
275 279
These are also sites of interaction
with the external proteins that form the extracellular
matrix (ECM). There are at least 16 different integrin
406 Chapter 8. Lipids, Membranes, and Cell Coats
subunits, whose external domains consist of up to
1114 residues, at least 8 subunits with external do-
mains of up to 678 residues, and at least 22 distinctly
different heterodimers.
280 282
The N-terminal part
of each subunit contains seven repeats of ~ 60 resi-
dues each, probably arranged as a -propeller (see Fig.
11-7D or 15-23).
281
The integrins are structurally com-
plex. Some contain a nucleotide-binding domain (see
Figs. 2-13 and 2-27C).
283
Integrins have differing and
quite exacting specificities toward the proteins of the
external matrix to which they bind. They span the
cell membrane and appear to be actively involved in
communication between the cytoskeleton and external
proteins.
275,277,284
They are often described as
receptors for the proteins that bind to them.
Other cell adhesion molecules. Long before the
discovery of integrins another class of transmembrane
adhesion molecules were recognized as members of
the immunoglobulin family. These were called cell
Glycophorin C
Ankyrin
Anion exchanger
(Band 3)
Membrane
4.1
4.1
Actin
4.9
Spectrin
chain
chain
Tropomodulin
Tropomyosin
Adducin
Actin
4.2
Figure 8-16 The erythrocyte membrane skeleton. (A)
Electron micrograph showing a region of the membrane
skeleton (negatively stained, X 200,000) and artificially
spread to a surface area nine to ten times as great as in the
native membrane. Spreading makes it possible to obtain
clear images of the skeleton whose protein components are
so densely packed and so subject to thermal flexing on the
native, unspread membrane that it is difficult to visualize the
individual molecules and the remarkably regular way that
they are connected. The predominantly hexagonal and
pentagonal network is composed of spectrin tetramers cross-
linked by junctions containing actin oligomers and band 4.1
protein. Band 4.9 protein and tropomyosin are probably also
bound to the oligomers, whose length (13 actin monomers
long) corresponds to the length of a tropomyosin molecule.
From Byers and Branton.
253,254
Courtesy of Daniel Branton.
(B) Proposed triple -helical structure of a single spectrin
repeat unit.
255
Courtesy of Ruby I. MacDonald and Alfonso
Mondragn. Each spectrin chain consists of 20 and each
spectrin chain of 17 such repeats, which have only partially
conserved sequences. The and chains are thought to
associate in a side-by-side fashion and the heterodimers
in an end-to-end fashion to give tetramers. These are the
rod-like structures seen in (A) and (C). (C) Cross section of
the unspread membrane and cytoskeleton as pictured by
Luna and Hitt.
256
Major interactions among components of
the cytoskeleton are shown. Apparent sizes of the protein
subunits, based on migration positions in SDS-polyacryla-
mide gel electrophoresis are: spectrin (260 and 225 kDa),
adducin (105 and 100 kDa), band 3 (90 to 100 kDa), protein
4.1 (78 kDa), protein 4.2 (pallidin, 72 kDa), dematin (protein
4.9, 48 kDa), glycophorin C (~25 kDa), actin (43 kDa), and
tropomyosin (29 and 27 kDa). Courtesy of Elizabeth J. Luna.
A
B
C
407
adhesion molecules (CAMs) by Edelman.
286,287
These
molecules go by names such as intercellular adhesion
molecule-1 (ICAM-1), neural cell adhesion molecule
(NCAM),
288,289
liver cell adhesion molecule (LCAM),
vascular cell adhesion molecule (VCAM), and platelet
endothelial cell adhesion molecule (PECAM).
290,291
Many of these proteins, including the T-cell antigen
CD2,
291a
were first recognized on leukocyte surfaces
as differentiation antigens and are often designated by
the Cluster of Differentiation names (Chapter 31).
For example, ICAM-1 is also called CD54 and PECAM
CD31.
290,292
The genes for these proteins are often
expressed differentially in various tissues and the
mRNA molecules formed undergo alternative splic-
ing.
287,289
The extracellular domains of these adhesion
molecules consist largely of Ig domains and most have
a single transmembrane helix and a small cytoplasmic
C-terminal domain. Some of the alternatively spliced
forms are attached to the membrane by PGI tails.
287,289
ICAM-1 (Fig. 8-18) has five Ig domains and VCAM-1
has seven,
293
but some CAMs have only two.
294
The
CAMs are glycoproteins, often with large N-linked
oligosaccharides attached. The widely distributed N-
CAM contains long 28 linked polysialic acid chains
on two of the three N-glycosylation sites in the fifth Ig
domain.
289
The CAMs are often referred to as recep-
tors. Their ligands include surface proteins such as
fibronectin (next section) but also the integrins, which
are also called receptors. Integrins are coreceptors for
receptors of the Ig superfamily. Each of the corecep-
tors in a pair has binding sites for other ligands as well
(Fig. 8-18). The CAMs and many other adhesion
molecules are most abundantly expressed in embryon-
ic tissues in which cells often migrate to new locations
and for which the communication with neighboring
cells is especially active. Many adhesion molecules
bind only weakly and reversibly to their ligands,
allowing cells to move.
The cadherins are calcium-dependent adhesion
proteins that mediate direct cell cell interactions.
295,296
The external parts of the cadherins also have repeated
structural domains with the Ig fold.
297-298b
They have
high affinity for each other, allowing cadherins from
two different cells to interact and tie the cells together
with a zipper-like interaction that is stabilized by the
bound Ca
2+
ions,
297,300
and may be relatively long-
lived. The gene for cadherin E is often mutated in
breast cancers and may be an important tumor
suppressor gene (Box 11-D).
301
Peripheral proteins of the outer membrane
surface. Many integral membrane glycoproteins have
their sugar-bearing portions exposed on the outer
surface of the plasma membrane. Among these are
receptors, ion pumps, and biochemical markers of
individuality. In addition to these proteins, which are
actually embedded in the bilayer, there are external
peripheral proteins. One of the best known of these is
B. Membranes
Figure 8-17 Working model of the proteinprotein interactions in focal adhesions determined by in vitro binding experi-
ments and immunolocalization. In addition, several interactions are of relatively low affinity in solution but may be enhanced
at the membrane surface. Abbreviations are: ECM, extracellular matrix; PM, plasma membrane; p-Tyr-?, unknown phosphoty-
rosine-containing protein; R/E/M, member of the radixin/ezrin/moesin family; VASP, vasodilator-stimulated phosphopro-
tein. Diagram is modified from Simon et al.
285

Integrin
ECM
p-Tyr?
Vinculin
Paxillin
Talin
Zyxin
Tenuin
F-actin
VASP
PM
-Actinin
pp125
FAK
pp60
src
Others
R/E/M
Tensin
408 Chapter 8. Lipids, Membranes, and Cell Coats
Figure 8-18 (A) Diagram of the ICAM-1 molecule. The structures labeled D1D5 are the Ig domains. The glycosylation sites
are labeled with small lollipops and approximate sites for binding of the chain (CD11a) of the integrin called LFA-1, for the
macrophage antigen Mac-1, for fibrinogen, for a ligand from erythrocytes infected by the malaria organism Plasmodium falci-
parum (PFIE), and for human rhinoviruses (HRV) are labeled. The binding sites are indicated schematically but each one is a
complex interacting surface complementary to its ligand. From Bella et al.
299
Courtesy of Michael Rossman. (B) Structure of
two N-terminal domains of E-cadherin. The molecules of the dimer are related by a noncrystallographic twofold symmetry
axis running vertically in the plane of the page. Clusters of three calcium ions are bound in the linker regions, connecting the
N- and C-terminal domains of each molecule and, in this view, are separated by the twofold axis. The N- and C-terminal
domains are composed of seven-stranded -barrels showing the same topology and similar three-dimensional structures.
From Nagar et al.
296
A
B
fibronectin (from fibra, fiber, and nectare, to
bind).
302 309
This very large 470-kDa glycoprotein is a
disulfide-linked dimer. Appearing under the electron
microscope as having two 60-nm arms,
305
fibronectin
molecules join together and surround animal cells,
anchoring other proteins and carbohydrates of the
ECM to the cells. Fibronectin binds tightly to several
different cell surface integrins and also to collagen and
to glycosaminoglycans of the matrix. It binds to the
blood-clotting protein fibrinogen, to actin, and also to
staphylococci and other bacteria. Each peptide chain
of the fibronectin dimer is organized as several do-
mains. Fibrin and staphylococci bind to the N-termi-
nal domain. A second domain binds collagen and a
domain near the C-terminus binds heparin. Several
different integrins of cell surfaces bind to the region of
the 8th, 9th, and 10th type III repeats.
308
The specificity
of the binding depends to a large extent on the presence
of the specific tripeptide sequence Arg-Gly-Asp
(RGD) in a type III repeat (Fig. 8-19B). Peptide
sequences such as the PHSRN (Fig. 8-19B) and oth-
ers
310
also participate in binding to specific integrins.
Initial interactions are noncovalent but fibrin and
collagen gradually become covalently attached to
fibronectin through isopeptide linkages formed by
transglutaminase (Eq. 2-23). This enzyme is abundant
409
in many developing tissues but often disappears as the
tissues mature.
311,312
Fibronectin was formerly known
as the large external transformation-sensitive pro-
tein, a name derived from the fact that its quantity is
greatly reduced in many virus-transformed cells. This
might explain the loss of adhesiveness and of contact
inhibition observed for cancer cells (Box 11-D).
There are 20 isoforms of human fibronectin.
These arise by alternative splicing of the primary
gene transcript. Their formation is developmentally
regulated.
308,315
One of the isoforms is present as a
soluble protein (cold-insoluble globulin) in blood
plasma.
302
The fibronectins belong to a larger family
of cytoadhesins, among which are the blood-clotting
proteins fibrinogen and von Willebrand factor
(Chapter 12), and also thrombospondin, vitronectin,
316
tenascin,
317 320
laminin,
321,322
osteopontin,
323
and
collagens. These proteins all have a modular construc-
tion with repeated domains, often of several different
types.
324
For example, tenascin contains 14
1
/2 EGF
domains (Table 7-3), 16 fibronectin type III repeats,
and a C-terminal segment homologous to fibrino-
gens.
325
Laminin (see Fig. 8-33) also has EGF-like
modules. Most of these proteins also have in common
the presence of the RGD sequence, which binds the
cytoadhesins to those integrins and other molecules
that carry RGD receptor sites.
304,326 328
The extracellu-
lar matrix is discussed further in Section E,2.
B. Membranes
Figure 8-19 (A) Schematic diagram of a human fibronectin molecule showing one complete
~ 250-kDa chain consisting largely of 29 type I, II, or III fibronectin domains with a 12-kDa
type III connecting segment or V-region. The second chain, for which only the C-terminal
portion is shown, is identical to the first except for the absence of the V-region as a result of
alternative splicing of the mRNA. The two chains are joined in antiparallel fashion by a pair
of S S bridges near the C terminus.
313
The kDa sizes refer to fragments generated by the
action of thermolysin. After Ingham et al.
306
(B) MolScript ribbon drawing of the structure of
the ninth and tenth type III repeats in human fibronectin. The biologically active peptide
segments PHSRN and RGD are represented in a stick and ball mode. The arrow marks valine
1416, next to which a polyglycine linker of various lengths has been inserted experimentally
in order to study interactions between modules.
308
From Spitzfaden et al.
314
12 11 10 15
S
S S
S
11 10 9 8 7 6 5 4 3 2 1 14 13 12
14 13 12
12 11 10 15
C
C
40 kDa
V
30 kDa 110 kDa
2 9 8 7 1 6 5 4 3 2 1
70 kDa 9 kDa
N
Type III domain Type II domain Type I domain
A
B
3. Enzymes and Membrane Metabolism
Many of the proteins of membranes are enzymes.
For example, the entire electron transport system of
mitochondria (Chapter 18) is embedded in membranes
and a number of highly lipid-soluble enzymes have
been isolated. Examples are phosphatidylserine
decarboxylase, which converts phosphatidylserine to
phosphatidylethanolamine in biosynthesis of the
latter, and isoprenoid alcohol phosphokinase,
which participates in bacterial cell wall synthesis
(Chapter 20). A number of ectoenzymes are present
predominantly on the outsides of cell membranes.
329
Enzymes such as phospholipases (Chapter 12), which
are present on membrane surfaces, often are relatively
inactive when removed from the lipid environment
but are active in the presence of phospholipid bilay-
ers.
330,331
The distribution of lipid chain lengths as
well as the cholesterol content of the membrane can
affect enzymatic activities.
332
Why are membranes so important to cells? Be-
sides their obvious importance in enclosing and defin-
ing the limits of cells, membranes are the result of a
natural aggregation of amphipathic molecules. They
also represent a natural arrangement for boundaries
between different aqueous phases within a cell. In
addition, membranes are the habitat for many rela-
tively nonpolar molecules formed by metabolism.
These include proteins with hydrophobic surfaces and
410 Chapter 8. Lipids, Membranes, and Cell Coats
those with membrane anchors. The semiliquid interi-
or of the membrane permits distortion of the bilayer
and the addition or subtraction of proteins (and low-
molecular-mass materials) in response to metabolic
processes in the adjacent cytoplasm.
The principal factor providing stability to macro-
molecules and membranes is the hydrophobic nature
of reduced organic compounds. This characteristic
leads to the separation of lipids, proteins, and other
molecules from the aqueous cytoplasm into oligomeric
aggregates and membranes. However, the best cata-
lysts, including most enzymes, are soluble in water.
Thus, membranes represent thin regions of relative
stability adjacent to aqueous regions in which chemi-
cal reactions occur readily and which tend to contain
the more polar, the smaller, and the more water-solu-
ble materials. The stability of membrane surfaces
provides a means of bringing together reactants and of
promoting complex sequences of biochemical reac-
tions. For example, membranes contain both oxidative
enzymes and reactive, dissolved quinones. The mem-
branecytoplasmic interfaces may often be the meta-
bolically most active regions of cells.
Despite their stability, membrane components
have a metabolism of their own which is related to the
high concentrations of oxidizing enzymes located in or
on membranes. Oxidative reactions provide a mecha-
nism for modification of hydrophobic membrane
constituents. For example, sterols, prostaglandins, and
other regulatory molecules are initially synthesized as
hydrophobic chains attached to water-soluble head
groups (Chapter 21). The hydrophobic products of
these synthetic reactions tend to be deposited in mem-
branes. However, attack by oxygen leads to introduc-
tion of hydroxyl groups and to a gradual increase in
water solubility. As the hydrophilic nature of the
compound is increased through successive enzymatic
hydroxylation reactions, the hydrophobic membrane
constituents eventually redissolve in the water and are
completely metabolized. Another process that actively
degrades membrane lipids is attack by hydrolytic
enzymes such as the phospholipases.
C. The Transport of Molecules through
Membranes
Small neutral molecules, such as water or ethanol,
can penetrate membranes by simple diffusion.
64,150
The rate is determined by the solubility of a substance
in the membrane, by its diffusion coefficient (see Eq.
9-24) in the membrane, and by the difference in its
concentration between the outside and the inside of
the cell. This concentration difference is commonly
referred to as the concentration gradient across the
membrane. The ease of diffusion through a mem-
brane is described quantitatively by a permeability
coefficient P which is related to the diffusion coeffi-
cient D (Eq. 8-3).
J = D
m
Kc / x = Pc
Here J is the flux of molecules across the membrane,
i.e., the number of molecules crossing one cm
2
per
second. D
m
is the diffusion coefficient in the bilayer,
while K is a partition coefficient, the ratio of the
concentration of the diffusing solute in the bilayer to
that in water. The concentration gradient of the solute
across the membrane is c, while x is the membrane
thickness in centimeters. The permeability coefficient
P for H
2
O through biological membranes
333
is about
1 10 m s
1
. For H
+
and OH

P is 0.1 m s
1
. For
halide ions diffusing across liposome bilayers P ranges
from 10
5
to 10
3
m s
1
,
334
fast enough to be of some
biological significance. However, for most other ions
P is less than 10
6
m s
1
. Because of their high lipid
content membranes are quite permeable to nonpolar
materials. For example, anesthetics usually have a
high solubility in lipids, enabling them to penetrate
nerve membranes.
1. Facilitated Diffusion and Active Transport
While simple diffusion may account for the en-
trance of water, carbon dioxide, oxygen, and anesthetic
molecules into cells, movement of most substances is
facilitated by protein channels and transporters.
335
Genes of 76 families of such proteins have been locat-
ed in the genomes of 18 prokaryotes.
335a
Some of these
provide for facilitated diffusion.
169,335
Like simple
diffusion, it depends upon a concentration gradient
and molecules always flow from a higher to a lower
concentration. A distinguishing feature of facilitated
diffusion is a saturation effect, i.e., a tendency to
reach a maximum rate of flow through the membrane
as the concentration of the diffusing substance, on the
high concentration side, is increased. In this character-
istic it is similar to enzymatic action (Chapter 9).
In active transport a material is carried across a
membrane against a concentration gradient, i.e., from
a lower concentration to a higher concentration. This
process necessarily has a positive Gibbs energy change
(as given by Eq. 6-25) of approximately 5.71 log c
2
/c
1
kJ mol
1
, where c
2
and c
1
are the higher and lower
concentrations, respectively. The transport process
must be coupled with a spontaneous exergonic reac-
tion. In primary active transport there is a direct
coupling to a reaction such as the hydrolysis of ATP to
pump the solute across the membrane. Secondary
active transport utilizes the energy of an electro-
chemical gradient established for a second solute;
that is, a second solute is pumped against a concentra-
tion gradient and the first solute is then allowed to
(8-3)
411
cross the membrane through an exchange process with
the second solute (antiport or exchange diffusion).
Alternatively, both the first and the second solutes
may pass through the membrane bound to the same
carrier (cotransport or symport). Another form of
active transport is group translocation, a process in
which the substance to be transported undergoes
covalent modification, e.g., by phosphorylation. The
modified product enters the cell and within the cell
may be converted back to the unmodified substance.
Transport processes, whether facilitated or active,
often require the participation of more than one mem-
brane protein. Sometimes the name permease is used
to describe the protein complexes utilized.
Like facilitated diffusion, active transport depends
upon conformational changes in carrier or pore pro-
teins, the equilibrium between the two conformations
depending upon the coupled energy-yielding process.
Thus, if ATP provides the energy a phosphorylated
carrier will probably have a different conformation
than the unphosphorylated protein. A carrier with
Na
+
bound at one site may have a different affinity for
glucose than the same carrier lacking Na
+
. A hypo-
thetical example of the kind of cycle that can function
in active transport is provided by the picture of the
sodium pump given in Fig. 8-25.
2. Pores, Channels, and Carriers
To accommodate the rapid diffusion that is often
needed to supply food, water, and inorganic ions to
cells, membranes contain a variety of small pores and
channels. The pores may be nonspecific or they may
be selective for anions or cations or for some other
chemical characteristics. They may be permanently
open or sometimes closed and referred to as gated.
The gating may be controlled by the membrane electri-
cal potential, by a hormone, by the specific ligand, or
by other means. Some pores may be small enough to
allow only small molecules such as H
2
O to pass
through. Others may be large enough to allow for
nonspecific simple diffusion of molecules of low mo-
lecular mass. Structures are known for only a few.
Large pores tend to be nonspecific, but when the
solute approaches the pore diameter in size the speci-
ficity increases. Furthermore, diffusion of ions
through pores is influenced strongly by any charged
groups in or near the pore. Thus, a cation will not
enter a pore containing a net positive charge in its
surface. Any electrical potential difference across the
membrane, resulting from accumulation of excess
negative ions within the cell, will also affect the diffu-
sion of ions.
336,337
Porins. The outer membranes of gram-negative
bacteria contain several 34- to 38-kDa proteins known
as porins. They form a large number of trimeric pores
which allow molecules and ions with molecular mass-
es < 600 Da to enter. However, even small proteins are
excluded. This appears to be a means for protecting
the bacteria against enzymes such as lysozyme (Chap-
ter 12). Four distinct porins are among the most abun-
dant proteins of the outer membrane of E. coli.
338
They
are designated according to the names of their genes.
OmpF (a nonspecific, open channel) and OmpC
(osmoporin) are encoded by OmpF (outer membrane
protein F) and OmpC genes, respectively. Maltoporin
(LamB) is selective for maltodextrins but also allows
other small molecules and ions to pass.
338 341a
Its
name comes from its original discovery as a receptor
for bacteriophage lambda. PhoE (phosphoporin) is a
porin with a preference for anions such as sugar phos-
phates while OmpF prefers cations.
342
Porin FepA is
ligand gated, opening to take up the chelated iron
from ferric enterochelin (Chapter 16). OmpF, OmpC,
and PhoE all have 16-stranded -barrel structures (Fig.
8-20).
199,343
Maltoporin forms a quite similar 18-
stranded barrel. Similar porins are present in many
bacteria.
199,344
The porin monomers associate to form trimeric
channels as is shown in Fig. 8-20B. They all have a
central water-filled, elliptical channel that is constricted
in the center to an eye ~ 0.8 x 1.1 nm in size. In this
restriction zone the channel is lined with polar residues
that provide the substrate discrimination and gating.
For example, in OmpF and PhoE there are many posi-
tively and negatively charged side chains that form the
edge of the eye (Fig. 8-20C). The electrostatic potential
difference across the outer membrane is small, but
apparently determines whether the porins are in an
open or a closed state.
344a
The voltage difference has
opposite effects on OmpF and PhoE, apparently as a
result of the differing distribution of charged
groups.
342,345,346
A key role in determining the voltage
dependence may be played by Lys 18 (Fig. 8-20C).
342
Polyamines (Chapter 24), which are present in the
outer membrane, induce closing of porin channels
by binding to specific aspartate and tyrosine side
chains.
347
The most abundant protein in the E. coli outer
membrane is OmpA. It appears to form a transmem-
brane helical bundle. Although it is regarded primarily
as a structural protein it too acts, in monomeric form,
as an inefficient diffusion pore.
350
Mitochondrial
outer membranes contain nonspecific pores (mito-
chondrial porins) that allow passage of sucrose and
other saccharides of molecular mass up to 2 to 8
kDa.
351,352
Similar pore-forming proteins have been
found in plant peroxisomes.
353
Aquaporins. Many biological membranes are not
sufficiently permeable to water to allow for rapid
osmotic flow. For example, the kidney membranes in
C. The Transport of Molecules through Membranes
412 Chapter 8. Lipids, Membranes, and Cell Coats
portions of Henles loop have permeabilities as high
as 2500 m s
1
(compared to 1020 m s
1
in a 1:1
cholesterol / phospholipid bilayer).
354-355a
This high
permeability is provided by aquaporin-1 (AQP-1),
formerly called CHIP (channel-forming integral mem-
brane protein) or AQP-CHIP. Aquaporin-1 was first
identified in erythrocyte membranes and is present in
many tissues. The 28-kDa subunits of the protein form
six-helix bundles, each with a pore in the center. These
are associated as tetramers in the membrane.
356 358
Other aquaporins with related sequences occur
broadly. There are at least ten in mammals.
359,359a
Plants, which must accommodate to heavy loss of
water in hot dry weather, have aquaporins in both
plasma membranes and tonoplasts.
360
Bacteria also
have aquaporins.
356,361
A defect in aquaporin-2 of the
kidney collecting duct leads to nephrogenic diabetes
insipidus, in which the kidneys fail to concentrate
urine in response to secretion of the hormone
vasopressin.
355a,362,363
Ion channels. Most organisms contain a large
number of ion channels. One of these, which plays a
key role in nerve conduction, is the voltage-gated K
+
channel. It is closed most of the time but opens when
a nerve impulse arrives, dropping the membrane
potential from its resting 50 - to 70-mV (negative inside)
value to below zero. This voltage change opens the
channel, allowing a very rapid outflow of K
+
ions.
364 367
The channels then close spontaneously. There are
many different K
+
channels but most have a similar
architecture.
368 370
The three-dimensional structure
has been determined for the membrane-spanning part
of the K
+
channel of Streptomyces lividans (Fig. 8-21).
The funnel-shaped tetrameric molecule has a narrow
conduction channel which contains the selectivity
filter.
The conduction channel is lined largely with
hydrophobic groups. The selectivity filter, which
discriminates between K
+
and Na
+
, is a short (~ 1.2-
nm-long) narrow (~ 1.0-nm-diameter) portion of the
Figure 8-20 MolScript ribbon drawings of the OmpF porin
of E. coli. (A) View of the 340-residue monomer. (B) View
of the trimer looking down the threefold axis. From Wa-
tanabe et al.
348
From atomic coordinates of Cowan et al.
349
(C) Molecular model of the constriction zone of the PhoE
porin. Locations of key residues are shown, with positions
of homologous residues in OmpF given in parentheses.
Extracellular loops have been omitted. Constructed from
coordinates of Cowan et al.
349
by Samartzidou and Del-
cour.
342
Courtesy of Anne Delcour.
A B
C
413
pore that is aligned roughly with
the center of the bilayer. It is
formed by four extended peptide
chains, one from each subunit, each
having the signature sequence
for K
+
channels, TVGYG. In this
sequence the peptide carbonyl
groups all point into the channel.
The consecutive groups of four
carbonyls along the channel form
binding sites for K
+
, whose ionic
diameter (Table 6-10) is 0.27 nm.
The K
+
must lose its hydration
sphere to fit into the 1.0-nm chan-
nel. The site of strongest binding,
occupied by K
+
in Fig. 8-21C, lies
just at the C-terminal ends of four
helices, and the partial negative
charges of the helix dipoles proba-
bly contribute to the binding. The
pore may constrict to strengthen
the bonds. When a second K
+
ion
enters the channel it appears to
bind ~ 0.75 nm from the central K
+
,
repelling it and weakening its
interaction with the filter, allowing
it to pass through the pore. Rb
+
(0.30 nm diameter) and Cs (0.34 nm
diameter) also pass through.
How are the smaller Na
+
(0.19
nm diameter) and Li
+
(0.12 nm
diameter) excluded from the pore?
The pore is too small for the hydrated
ions and perhaps too large to bind
the dehydrated ion well enough
to let it escape from its hydration
sphere. Four negatively charged
side chains in the cytoplasmic
mouth of the pore presumably
discourage anions from entering.
At the other end, the extracellular
entryway is a site that can be blocked
specifically by 35- to 40-residue
scorpion toxins.
368
The S. lividans K
+
channel is
not voltage gated. Voltage-gating
mechanisms must be learned from
study of other channels! The voltage-
dependent K
+
channels from the rat
have an
4

4
composition. The
and subunits coassemble in the
endoplasmic reticulum and remain
as a permanent complex,
370ac
After
insertion into the plasma mem-
brane the subunits form a chan-
nel as in Fig. 8-21. However, an
additional intracellular domain of
C. The Transport of Molecules through Membranes
Figure 8-21 Views of the tetrameric K
+
channel from Streptococcus lividans.
(A) Ribbon representation as an integral membrane protein. Aromatic amino
acids on the membrane-facing surface are also shown. (B) Stereoscopic view.
(C) Stereoscopic view perpendicular to that in (B) with a K
+
ion in the center.
From Doyle et al.
366
A
C
B
414 Chapter 8. Lipids, Membranes, and Cell Coats
the subunits, together with the
4
complex, provide
an elaborate and as yet poorly understood internal
structure. The subunits are oxidoreductases contain-
ing bound NADH, whose function is also uncertain.
There are many more pores and carriers of various
kinds in biological membranes. Some, like the S. lividans
K
+
channel, facilitate diffusion of a single ion or
compound. They are uniporters. Others promote
cotransport in which an ion such as H
+
or Na
+
also
passes through the carrier. In some cases a pore is
formed by a single molecule. Other pores follow a
twofold, threefold, or fourfold axis (Fig. 8-21) of an
oligomeric protein.
371
Two different conformations
for the protein, one in which a gate opens to one
side of the membrane and one in which it opens to the
other, are usually involved. Interconversion between
the two conformations is spontaneous but the equilib-
rium between them may be influenced by the binding
of the solute, by the membrane potential, or by bind-
ing of inhibitors or activators. The latter may bind
differently to the parts of the carrier (pore) protein
exposed on the two sides of the membrane.
Channel-forming toxins and antibiotics. Some
of the bacterial toxins known as colicins (Box 8-D) kill
susceptible bacteria by creating pores that allow K
+
to
leak out of the cells. One part of the complement
system of blood (Chapter 31) uses specific proteins to
literally punch holes in foreign cell membranes. Mel-
litin, a 26-residue peptide of bee venom,
372,373
as well as
other hemolytic toxins and antibiotic peptides of insects,
amphibians, and mammals (Chapter 31) form amphip-
athic helices which associate to form voltage-depen-
dent anion-selective channels in membranes.
374 377
The polypeptide antibiotic gramicidin A consists
of 15 nonpolar residues of alternating d- and l-config-
uration; two molecules can form a channel with a
right-handed helix structure.
378,379
The central 0.48-nm
hole in the helix is large enough to allow unhydrated
cations such as Na
+
or K
+
to pass through. The same
peptide, under other conditions, forms left-handed
helical channels whose structures are known.
379
Aggregates of
10
helices may form channels between
helices of suzukacillin.
380
For this antibiotic as well
as for alamethicin (Chapter 30) the conductance
depends upon the membrane potential, a characteristic
shared with the pores of nerve membranes. A variety
of synthetic channel-forming peptides have been
prepared. Some of these form -helical bundles with
a central pore. A five helix bundle of this type
381
has
a structure reminiscent of the acetylcholine receptor
channel of neurons.
382
Ionophores and other mobile carriers. Facili-
tated diffusion of a molecule or ion is sometimes
accomplished by binding to a mobile carrier. An
example is the diffusion of a complex of K
+
with the
low-molecular-mass lipid-soluble carrier, or ionophore
valinomycin (Fig. 8-22). The K
+
valinomycin com-
plex diffuses the short distance to the other side of the
membrane and discharges the bound ion. If the rates
of binding to a carrier and of release from the carrier
are greater than those of the diffusion process,
MichaelisMenten kinetics are observed. The maxi-
mum velocity V
max
and Michaelis constant K
m
can
be defined as in Eq. 9-15 for enzymatic catalysis.
Valinomycin is a depsipeptide which contains
ester linkages as well as amide linkages. The anti-
biotic is made up of d- and l-valine, l-lactic acid, and
d-hydroxyisovaleric acid. When incorporated into an
artificial membrane bathed in a K
+
-containing medium,
valinomycin increases the conductance greatly and
when it is added to a suspension of Streptococcus
faecalis cells the high ratio of [K
+
]
i
/ [K
+
]
o
falls rapidly.
383
The loss of K
+
from cells probably explains the anti-
biotic activity. However, under suitable conditions,
with a high external [K
+
], the bacteria will continue
to grow and reproduce in the presence of the anti-
biotic.
384
Uncomplexed valinomycin has a more extended
conformation than it does in the potassium com-
plex.
385,386
The conformational change results in the
breaking of a pair of hydrogen bonds and formation
of new hydrogen bonds as the molecule folds around
the potassium ion. Valinomycin facilitates potassium
transport in a passive manner. However, there are
cyclic changes between two conformations as the
carrier complexes with ions, diffuses across the mem-
brane, and releases ions on the other side. The rate of
transport is rapid, with each valinomycin molecule
being able to carry ~ 10
4
potassium ions per second
across a membrane. Thus, a very small amount of this
ionophore is sufficient to alter the permeability and
the conductance of a membrane.
Because the stability constant of its complex with
potassium is much greater than that with sodium,
valinomycin is a relatively specific potassium ionophore.
In contrast, the mushroom peptide antamanide has a
binding cavity of a different geometry and shows a
strong preference for sodium ions.
388,390
The structure
of the Na
+
antamanide complex is also shown in
Fig. 8-22B. The Streptomyces polyether antibiotic
monensin (Fig. 8-22D),
389,391
a popular additive to
animal feeds, is also an ionophore. However, its mode
of action, which involves disruption of Golgi functions,
is uncertain.
392
Anions of lipid-soluble phenols such as 2,4-dini-
trophenol can serve as effective carriers of protons
(Chapter 18). However, proteins usually serve as the
natural carriers, both of protons and of other ions. A
protein is sometimes pictured as rotating to present
the solute-binding surface first to one side, then to the
other side of a membrane. However, gated pores or
channels are probable for most biological transport.
415 C. The Transport of Molecules through Membranes
permeability coefficient P for D-glucose across a lipid
bilayer is only 10
6
to 10
5
m s
1
. For an intact eryth-
rocyte, P is much greater: ~ 1 m s
1
. This is the result
of facilitated diffusion by a transport protein with a
high specificity for hexose and pentose sugars having
a pyranose ring in a C1 chain conformation.
396
This
human erythrocyte glucose transporter, now known as
GLUT1, has a K
m
of 1.6 mM for D-glucose but of >3 M
for L-glucose. It is a 55-kDa intrinsic membrane glyco-
protein migrating in band 4.5 of Fig. 8-14. From the
sequence of its cloned gene the unglycosylated carrier
was deduced to be a 54-kDa peptide of 492 residues.
3. The 12-Helix Major Facilitator Superfamily
A large family of transmembrane facilitators from
bacteria and eukaryotes appear to consist largely of 12
transmembrane helices with intervening cytosolic and
extracellular loops. Some of these transporters facili-
tate simple uniport diffusion, but others participate in
active transport of the symport or antiport type.
393,394
Several hundred members of the family are known.
395
Entrance of sugars into cells. It is important
that sugars be able to enter cells rapidly. However, the
Figure 8-22 (A) Uncomplexed valinomycin and its complex with K
+
(from Duax et al.
387
). (B) Stereodiagram of the Na
[Phe
4
, Val
6
]antamanide complex. A molecule of C
2
H
5
OH, which forms the fifth ligand to the Na
+
, is omitted for clarity. From
Karle et al.
388
(C) Monensin and its complex with K
+
. From Pangborn et al.
389
N
O
N
O
O
O
O
O
CH
3
H
3
C CH
3
H
3
C CH
3
H
3
C CH
3
H
H
H
H
H H
l-Lactic acid d-Hydroxy-
isovaleric acid
Valinomycin
3
d-Val l-Val
B
A
O O O
O O
COOH
H
3
C
HO
OH
CH
3
CH
3
H
OCH
3
CH
3
H
H
CH
2
OH
H H
CH
3
CH
2
CH
3
CH
3
CH
3
Monensin
C
K
+
K
+
K
+
Na
+
antamanide
416 Chapter 8. Lipids, Membranes, and Cell Coats
Figure 8-23 Predicted topology of the human glucose transporter GLUT1. The 12 predicted helices are numbered and the
single external N-linked oligosaccharide is marked CHO. The sequence RXGRR (marked) is found in many 12-helix transporters.
Its occurrence in these two positions suggests that the transporters may have evolved by duplication of a 6-helix motif. How-
ever, the human glucose transporters otherwise show no sequence similarity to other 12-helix transporters. After Bell et al.
398
See also Muekler.
400
12 11 10 9 8 7 6 5 4 3 2 1
CHO Hydrophobic
Polar
Lys, Arg
Asp, Glu
Cytoplasm
COOH
NH
2
RXGRR
M
e
m
b
r
a
n
e
M
e
m
b
r
a
n
e
The glucose transporter is specifically inhibited by the
fungal metabolite cytochalasin B which can also be
used for photoaffinity labeling of the transporter.
397
There are at least six closely related facilitative
sugar transporters in the human body.
398 400
GLUT1 is
found not only in red cells but also in brain
401
and
other tissues. GLUT2 is the principal liver transporter
and GLUT3 is found along with GLUT1 and GLUT5 in
the brain.
401
GLUT5 is primarily a fructose transport-
er.
402,403
The latter is present in spermatozoa, in which
fructose transport is especially important (Box 20-A),
and also in the small intestine. Both the GLUT1 and
GLUT5 genes are overexpressed in breast cancer.
403
GLUT6 is an unexpressed pseudogene (see Chapter
27) and GLUT7 has been found only in liver micro-
somes. GLUT4 of skeletal and cardiac muscle and
adipose tissue has received a great deal of attention
because of its response to insulin
400,404-405a
and to
exercise
406
(see Chapter 17).
All of the GLUT family appear to be 12-helix
transmembrane-regulated, gated-pore proteins
407,408
with relatively short cytosolic N- and C-terminal ends.
The proposed topology of GLUT1, which has been
supported by much experimental data,
400,406
is shown
in Fig. 8-23. However, the helices are thought to be
bundled with the glucose channel centered within a
helix bundle. The structure is unknown, but it could
resemble the ,-barrel of glucoamylase (Fig. 2-29),
which has 12 helices, a glucose-binding site in the
center, and a structure that could easily be modified to
form a central pore.
Dehydroascorbate, the oxidized form of vitamin C
(Box 18-D) is also transported into cells by GLUT1 and
GLUT3.
409
A related transporter carries L-fucose into
mammalian cells.
410
Another facilitates the uptake of
galactose in yeast.
411
Cotransport of sugars and other nutrients with
H
+
or Na
+
. Epithelial cells of the small intestine or of
kidney tubules must take up glucose at low concentra-
CH
2
H
3
C
H
OH
H
O
CH
3
O
OH
HN
O
Cytochalasin B
417
tions and discharge it into the bloodstream at a higher
concentration.
412
This active transport is accomplished
by cotransport of glucose with Na
+
in a 1:2 ratio, with
the sodium ion concentration gradient across the
membrane providing a usable source of energy
amounting to 5.8 kJ / equivalent of Na
+
.
413
The 662-
residue human, sodium-dependent transporter
SGLT1 may have 14 transmembrane helices, 5 of
which have been proposed to provide the sugar path-
way.
414
Cotransport with Na
+
is also observed for trans-
port of many other sugars, amino acids, neurotrans-
mitters, and cofactors.
415
A confusing variety of
transporter molecules have been identified and are
now being classified into families based on gene se-
quences as well as function.
416,417
Transporters from
intestinal mucosal cells, kidney membranes, and syn-
aptic endings of neurons have been studied most and
have been the source for many of the cloned genes.
418
The Na
+
-dependent transporters of neutral amino
acids from these tissues have long been classified as
system A, system B, and system ASC (alanine, serine,
cysteine) according to substrate specificities.
417,419-420a
Both cationic amino acids and cystine are taken up by
kidney tubules and intestinal epithelial cells by anoth-
er Na
+
-dependent transporter which is defective in
human cystinuria, a common metabolic genetic prob-
lem.
421,422
The brain contains several transporters specialized
for rapid uptake of neurotransmitters glutamate and
aspartate,
423 425
glycine,
426
-aminobutyrate
(Gaba),
427,428
and catecholamines and also for taurine,
L-proline,
429
serotonin,
430
and other substances. Many
of these are not only Na
+
dependent but also require
cotransport of Cl

.
428 430
There are several different
glutamate transporter genes with specialized distribu-
tion in the brain and other tissues.
423,424
Cotransport of sugars with H
+
is especially com-
mon in bacteria
431
but also occurs in eukaryotes. For
example, the alga chlorella employs hexose / H
+
symporters.
432
The most investigated H
+
cotransport-
er is probably the lactose (lac) permease from E. coli
which enables E. coli to take up lactose and other -
galactosides from very dilute solutions.
433 436
From a
variety of measurements it has been possible to pro-
pose a stacking arrangement for the 12 helices
434
and
to identify groups that are essential for function of the
417-residue protein. Glutamates as well as an argin-
ine, and a histidine, all in transmembrane regions, are
essential and may be involved in the gating and trans-
port functions.
436
The 469-residue melibiose permease
of E. coli transports -D-galactopyranosides, including
meliobiose (Galp1 6 Glc) and raffinose (Galp1
6 Glcp1 2 Fruf).
437,438
This permease will couple
sugar uptake to either the H
+
or Na
+
gradient (or to a
gradient of Li
+
but not of K
+
).
437
Cells of E. coli also
contain H
+
symporters for D-galactose, D-xylose, and L-
arabinose that are homologous to the mammalian
GLUT proteins.
439
A series of specific H
+
-linked cotransporters are
found in the brush border membranes of small intes-
tine and in kidney epithelial cells.
440,441
In green plants
H
+
-linked cotransport of amino acids is used in the
distribution of amino acids synthesized in the roots
and leaves to other parts of the plants.
442,443
Antiporter or ion exchange transporters are also
common. For example, E. coli uses a metal iontetra-
cycline / H
+
transporter to carry the antibiotic tetracy-
cline out of cells. This protein, when present, provides
a high level of antibiotic resistance to the bacteria.
444
4. Active Transport Systems
Both bacteria and eukaryotes also possess complex
active transport systems for uptake of sugars, amino
acids, and other nutrients and for pumping out toxic
xenobiotics. One of the most important groups are the
high affinity ABC (ATP-binding cassette) transporters,
also called traffic ATPases.
445 448a
The E. coli genome
contains genes for 80 ABC transporters, 54 of which
had been identified before the genome sequence was
completed.
447
In the human body an ABC transporter
enables eukaryotic cells to pump out a large number of
different drugs and other foreign compounds. This
multidrug resistance protein (or P-glycoprotein)
not only protects cells but also can seriously interfere
with drug treatment. For example, cancer cells with
increased amounts of this transporter often arise dur-
ing chemotherapy. The transporter protein is a single
1280-residue, 170-kDa chain which probably has
12 transmembrane helices and two ATP-binding
domains.
448,449
Other human ABC transporters include
the antigen processing transporter TAP (Chapter 31),
the 1480-residue anion transporter CFTR, which is
defective in cystic fibroses (Box 26-C),
450
the erythro-
cyte glutathione-conjugate exporter (Box 11-B), and
a long-chain fatty acid transporter of peroxisomes.
ABC transporters usually consist of four domains.
Two are hydrophobic intrinsic membrane domains,
each with six membrane-spanning helices and two are
peripheral membrane ATP binding domains. All four
domains may be in a single peptide chain, as in CFTR,
or they may be separate smaller proteins as in bacterial
periplasmic permeases.
431,446
Periplasmic permeases. Gram-negative bacteria
contain numerous ABC transporters with components
located on the periplasmic surfaces of their plasma
membranes. Many of these can be dissociated from
the surfaces by osmotic shock, i.e., by sudden changes
in the osmotic pressure of the medium.
451,452
For
example, cells of E. coli suspended in 0.5 M sucrose,
treated with 10
4
M EDTA for 10 min, and then diluted
C. The Transport of Molecules through Membranes
418 Chapter 8. Lipids, Membranes, and Cell Coats
with cold water release ~ 50 binding proteins that
hold sugars, amino acids, ions, and other substances
tightly with K
f
= 10
6
10
8
M
1
.
453,454
An example is the
L-arabinose binding protein of E. coli, a 306-residue
peptide organized into two large / units with a
deep cleft between them. The sugar is bound into this
cleft
455,456
by an extensive network of hydrogen-bond-
ing interactions between polar groups in the sugar and
in the protein similar to the network that binds either
D-galactose (Fig. 4-18) or D-glucose to another of the
periplasmic binding proteins.
457,458
Proteins with the
same general architecture bind D-ribose, L-arabinose,
458
and maltodextrins.
459
The 1,4-linked maltodextrins,
as well as maltose, are major nutrients for E. coli and
enter the periplasmic space via the previously men-
tioned LamB porin. Other periplasmic proteins bind
histidine,
460
basic amino acids,
461
branched chain
amino acids, only leucine, oligopeptides, polyamines,
462
and the tetrahedral anions phosphate
454
and sulfate.
463
The sulfate
2
anion is held to its binding protein by
seven hydrogen bondsone from a serine OH, one
from an indole ring NH, and five from peptide NH
groups, three of which are at the positive ends of
helices. No permanently charged groups nor
cations nor water molecules come into contact with
the SO
4
2
.
463
The phosphate-binding protein also
binds a tetrahedral dianion HPO
4
2
but it doesnt bind
sulfate. It forms numerous hydrogen bonds with its
ligand and also an ion pair with a guanidinium group.
An aspartate carboxylate hydrogen bonds to the OH
BOX 8-D COLICINS: ANTIBIOTIC PROTEINS
Certain strains of E. coli and related bacteria
synthesize proteins known as colicins that kill cells
of other susceptible strains.
a c
Three kinds of co-
licins, each encoded in its own small DNA plasmid
(colicinogenic factor), are known. Colicin E3 is a
58-kDa ribonuclease (RNase) that attacks 26S ribo-
somal RNA of susceptible bacteria;
d
colicin E2 is a
deoxyribonuclease (DNase) that cleaves the bacteri-
al chromosome.
e
Colicin E1 and its relatives, co-
licins A, B and Ia, Ib and N, attack the bacterial
inner membranes and form lethal pores which allow
K
+
and other ions to flow out of the cell. The pres-
ence of a single channel will kill the bacterium. The
effect is similar to that of valinomycin.
f,g
The small
colicin V is an 88-residue peptide antibiotic that is
secreted by a dedicated ABC export system.
h
Co-
licins are members of a larger group of bacterio-
cins. One of these proteins, megacin Cx from
Bacillus megaterium, kills bacteria of sensitive strains
by blocking protein synthesis.
i
The channel-forming colicins bind to bacterial
surface molecules that serve as their receptors. For
example, colicin N binds to the abundant E. coli
surface protein OmpF. Interaction with a complex
of membrane proteins known as tol Q, R, A, and B
then leads to translocation across both outer and
inner membranes and refolding of the colicin in a
pore-forming conformation.
g
This mechanism is
also used by colicins A, E, and K. The single-strand-
ed bacteriophages M13, fd, and f1 parasitize the
same transport system. Colicins B, D, I, and M enter
bacteria with the aid of a second transport system
consisting of proteins TonB, ExbB, and ExbD which
participate in uptake of chelated iron (Chapter 16)
and of vitamin B
12
. This system is also parasitized
by bacteriophages T1, T5, and 80.
j
a
Luria, S. E. (1975) Sci. Am. 233(Dec), 30 37
b
Parker, M. W., Tucker, A. D., Tsernoglou, D., and Pattus, F.
(1990) Trends Biochem. Sci. 15, 126 129
c
Parker, M. W., and Pattus, F. (1993) Trends Biochem. Sci. 18, 391
395
d
Escuyer, V., Boquet, P., Perrin, D., Montecucco, C., and Mock,
M. (1986) J. Biol. Chem. 261, 10891 10898
e
Dvhsllrt, K., and Nomura, M. (1976) Proc. Natl. Acad. Sci. U.S.A.
73, 3989 3993
f
Wiener, M., Freymann, D., Ghosh, P., and Stroud, R. M. (1997)
Nature (London) 385, 461 464
g
Evans, L. J. A., Labeit, S., Cooper, A., Bond, L. H., and Lakey, J.
H. (1996) Biochemistry 35, 15143 15148
h
Fath, M. J., Zhang, L. H., Rush, J., and Kolter, R. (1994) Biochem-
istry 33, 6911 6917
i
Brusilow, W. S. A., and Nelson, D. L. (1981) J. Biol. Chem. 256,
159164
j
Derouiche, R., Bndetti, H., Lazzaroni, J.-C., Lazdunski, C.,
and Lloubs, R. (1995) J. Biol. Chem. 270, 11078 11084
k
Griko, Y. V., Zakharov, S. D., and Cramer, W. A. (2000) J. Mol.
Biol. 302, 941953
l
Chan, P. T., Ohmori, H., Tomizawa, J.-I., and Lebowitz, J. (1985)
J. Biol. Chem. 260, 8925 8935
The N-terminal portion of the 522-residue
polypeptide chain of colicin E1 appears to be re-
quired for transport into the membrane and the
central part for binding to the receptor; the channel-
forming property is characteristic of the C-terminal
region.
k
A similar organization has been established
for the smaller colicin N:translocation domain,
(residues 1 66), receptor domain, (residues 67182),
and pore-forming domain (residues 183 387).
The colicin E1 plasmid is a 4.43 MDa circular
double stranded DNA molecule consisting of 6646
base pairs.
l
Only one site is susceptible to cleavage
by the restriction endonuclease ECoR1 (Chapter 26)
This feature has led to its widespread use in cloning
of genes.
419
of the phosphate. Since the sulfate ion lacks the neces-
sary H to form this bond it is excluded from the bind-
ing site.
454
All of these binding proteins have similar architec-
tures. The ligands fit into a groove between two do-
mains as is seen in Fig. 8-24 for the histidine binding
protein. The histidine is held by formation of carboxy-
latearginine and NH
3
+
aspartate ion pairs and an
additional hydrogen bond to the imidazole. The
proteins are able to bend in the hinge region between
the two domains to give a better fit to their ligands.
The periplasmic binding proteins function togeth-
er with the other subunits of the ABC transporter
system. One of the best understood systems is encod-
ed by the histidine transport operon of Salmonella
typhimurium.
445,453
There are four genes: hisJ (encoding
the histidine-binding protein), hisQ, hisM, and hisP.
The Q and M proteins are hydrophic integral mem-
brane proteins which interact with two copies of the P
protein, which contains the ATP-binding motif, to
form a His QMP
2
membrane complex. The soluble
HisJ transfers its bound histidine to this complex,
which with the hydrolysis of ATP supplying the driv-
ing force transfers the histidine across the membrane,
presumably via a channel.
445
In E. coli the gene malE
encodes the periplasmic maltose binding protein,
while malF and malG are genes for the integral plasma
membrane components. The malK gene encodes an
ATP-binding protein homologous to the hisP protein.
As with the histidine permease, a multiprotein complex
MalFGK
2
is formed. It accepts maltose or a maltodex-
trin and ATP is hydrolyzed to drive the transport.
459
MalT encodes a positive regulatory protein that stimu-
lates transcription of the mal genes.
464
Membrane
transport systems are made more complex by the fact
that several of the binding proteins serve also as recep-
tors for stimulation of chemotaxis (Chapter 19).
The bacterial phosphotransferase system. A
third system for uptake of sugars is utilized by E. coli
and by many other bacteria. This phosphoenolpyru-
vate-dependent phosphotransferase system converts
glucose, mannose, fructose, other sugars, or mannitol
into their 6-phosphate esters, at the same time trans-
porting the latter across the membrane (a group trans-
location).
431,465
Four proteins form a cascade (Eq. 8-4).
Phosphoenolpyruvate, an intermediate in sugar me-
tabolism whose high energy of hydrolysis provides the
Successive
transfers of
a phospho
group
PEP (phosphoenolpyruvate)
Sugar phosphate (inside)
(Carbohydrate-
specific protein
or domain)
E IIA N PO
3
2
or
E III N PO
3
2
E IIB S PO
3
2
Sugar
(outside)
C
CH
2

OOC O PO
3
2
E IIC (Integral membrane
domain)
Sugar-specific
permease
complex
HPr N PO
3
2
(Phosphate carrier protein)
Enzyme I N PO
3
2
(Phosphohistidine form)
(8-4)
Figure 8-24 (A) MolScript ribbon drawing of the periplasmic histidine-binding protein HisJ, a component of an ABC trans-
porter system of Salmonella. The bound L-histidine is shown as a ball-and-stick model. (B) Stereoscopic view of the histidine-
binding site showing hydrogen-bonding interactions of protein side chains with the histidine. From Oh et al.
460
Courtesy of
Giovanna Ferro-Luzzi Ames.
A B
C. The Transport of Molecules through Membranes
420 Chapter 8. Lipids, Membranes, and Cell Coats
driving force, phosphorylates N
2
of a histidine side
chain on enzyme I, a large 64-kDa soluble membrane-
associated protein.
466-467a
The phospho group is then transferred sequential-
ly to the small 88-residue dimeric phosphate carrier
protein HPr, to the carbohydrate-specific membrane
proteins IIA and IIB, and to the sugar being transport-
ed. A histidine side chain at position 15 of HPr is
phosphorylated by PEP to form N
1
-phosphohistidine.
468
The NH
2
of the same histidine forms a hydrogen
bond to C-terminal glutamate 85, which is also hydro-
gen bonded to an arginine side chain. This chain of
interacting groups may function in the phospho trans-
fer reactions.
469
Both enzyme I and HPr function in
the transport of many ligands but there are specific
enzymes II (EIIs) for each sugar or other ligand.
470
In
E. coli there are at least 13 different PTS transporters.
471
A complete EII usually consists of three domainsEIIA
(or EIII), EIIB, and EIIC. In some cases all three
domains are in a single polypeptide chain, but in other
cases they are individual proteins or some combina-
tion of individual and bifunctional proteins. The 637-
residue mannitol-specific EII contains all three domains.
Domains A and B are cytoplasmic while the N-terminal
segments form the integral membrane C domain. The
two phosphorylation sites are His 554 (EIIA) and Cys
384 (EIIB).
472
The glucose-specific EII from E. coli
consists of two subunits, IIA and IICB.
471,473 475
The
mannose transporter has three subunits representing
domains IIAB, IIC, and an additional integral mem-
brane subunit IID.
476
In addition to their direct trans-
port functions, components of the PTS system play
regulatory roles in chemotaxis, transcription, and
control of other transporters.
477,478
5. Transport of Ions
Cell membranes are impermeable to most ions.
Only a small number of ions can enter cells readily
and these usually do so with the assistance of protein
channels or pores. The principal anion of plasma (Box
5A) is Cl

, which passes through membranes readily


by virtue of the presence of channel-forming proteins.
Chloride ions are often distributed across membranes
passively according to Eq. 8-5, which describes the
Donnan equilibrium.
167,479,480
[K
+
]
i
[Cl

]
i
= [K
+
]
o
[Cl

]
o
Here the subscripts i and o refer to the inside and the
outside of the cell, respectively. The potassium ion
concentration within a cell is maintained at a high
value by the operation of the Na
+
+ K
+
pump and by
the presence of nondiffusible anions within the cell.
According to Eq. 8-5, the internal chloride concentra-
tion must be low, with the product of [K]
i
[Cl]
i
equaling
that of the low exterior [K
+
] and high exterior [Cl

].
The internal [Na
+
] and [Ca
2+
] are both low, while the
internal [K
+
] is high. These differences are also linked
to the membrane potential (Eq. 8-2), which is ordinari-
ly expressed as a negative voltage of the interior of a
cell, mitochondrion, plastid, etc. with respect to a
reference electrode in the external medium.
The maintenance of both the membrane potential
and the steep gradients of ionic concentrations is
essential to cells, both as a means of coupling meta-
bolic energy to transport and other processes and for
electrical signaling. The effects are most pronounced
for mitochondrial membranes for which E
m
may attain
140 to 170 mV and for plasma membranes of excitable
cells such as neurons (E
m
= 70 to 90 mV). For liver
and kidney cells E
m
of plasma membranes may be
approximately 35 mV and for erythrocytes only
9 mV.
480
In excitable cells electrical impulses are initiated by
opening or closing ion channels. They are propagated
along an axon by a complex sequence of opening and
closing of voltaged-gated channels,
481
a process that is
described in Chapter 30. All cells appear to also contain
ATP-driven ion pumps as well as simple channels,
cotransporter proteins, and ion exchangers.
Anions. Cell membranes have long been known
to be relatively permeable to Cl

and other small an-


ions. However, the molecular basis of this permeabili-
ty is quite complex.
482
Voltage-gated selective anion
channels, often called chloride channels, are impor-
tant in electrically excitable membranes, where they
ensure a high resting chloride conductance and stabili-
ty.
483
The gene for one of these channel proteins was
first cloned from the electric ray Torpedo
484,485
and is
designated Clc-0. Similar channels have been found in
organisms ranging from bacteria to yeast, green plants,
and vertebrate animals. The yeast genome contains
just one Clc gene
486
but mammals have at least nine.
One of these, Clc-2, is defective in myotonia congenita,
a human disease of impaired muscle relaxation; and in
similar diseases of mice, goats, and horses.
483,487
A
mutation in the chloride channel Clc-5 causes kidney
problems, including proteinuria, hypercalciuria, and
kidney stones.
483
Erythrocyte membranes permit rapid transport of
anions to allow for the exchange of Cl

within the red


cells for HCO
3

generated by tissue metabolism. The


HCO
3

binds to deoxyhemoglobin (Eq. 7-47) and is


carried to the lungs, where it is released upon oxygen-
ation of the hemoglobin. Then the reverse exchange of
internal HCO
3

for Cl

occurs. This electroneutral ion


exchange is mediated by the band 3 protein (Fig. 8-14)
which contains a channel that allows anions but not
cations to pass.
238,239,488
The band 3 protein, also
known as AE1 (anion exchanger 1), is found principally
in red blood cells but is also present in kidney tubules.
489
(8-5)
421
Related proteins occur in other tissues.
488
The 911-
residue band 3 protein consists of two distinct parts of
nearly equal size. The N-terminal portion is attached
to the membrane skeleton (Fig. 8-16). The C-terminal
part, which is embedded in the membrane, is thought
to form 14 transmembrane helices and to contain the
ion exchange channel or channels.
489a
As previously
mentioned, defects in the N-terminal portion cause
spherocytosis. The mutation Arg 589 His in the
C-terminal half causes renal tubular acidosis in
which the kidneys do not adequately remove acids
from the body.
238,489
Band 3 proteins can also exchange
phosphate, sulfate, and phosphoenolpyruvate for Cl

or bicarbonate.
Another chloride channel, which is regulated by
cyclic AMP (Chapter 11), functions in secretory epithe-
lia. Its regulation is faulty in cystic fibrosis (Box 26-A),
one of the most common human genetic defects,
especially among persons of European descent.
490
As
was previously mentioned, this cystic fibrosis trans-
membrane conductance regulator (CFTR) is a mem-
ber of the ABC superfamily of transporters. The large
1480-residue protein apparently has two 6-helix mem-
brane-spanning domains, two cytoplasmic nucleotide-
binding domains,
491
and another large cytoplasmic
regulatory domain.
In addition to AE1 (band 3 protein), kidneys
depend upon other modes of reabsorption of HCO
3

from the proximal tubules. These include a Na


+
/
HCO
3

cotransporter, which seems to be related to the


AE family of ion exchangers. However, it transfers
three HCO
3

ions per Na
+
and is therefore highly
electrogenic.
492,493
Transporters for phosphate, sul-
fate, and small organic anions are found in many
organisms. Bacterial periplasmic transporters have
already been described. Plants employ H
+
/ phos-
phate,
494
H
+
/ sulfate,
495
and H
+
/ nitrate
496
cotransport-
ers. Phosphate transporters are probably essential to
all organisms. One of the best known is the mitochon-
drial P
i
/ H
+
cotransporter which carries phosphate
ions originating from hydrolysis of ATP to ADP + P
i
back into the mitochondria.
497,498
See also Table 18-8.
A human Na
+
/ P
i
cotransporter in the kidney is also
essential. An X-linked trait leading to inadequate
synthesis of the transporter causes hypophosphatemic
vitamin D-resistant rickets.
499
Monocarboxylates such as lactate and pyruvate
enter animal cells with the aid of monocarboxylate /
H
+
cotransporters of low specificity.
500
A Cl

/ oxalate
transporter is one of several ion exchange proteins in
the kidney.
501
Transport systems for ADP, phosphate,
dicarboxylates, and other anions are very active in
mitochondrial membranes (Chapter 18).
Cation channels. When a nerve impulse passes
along an axon gated pores or channels specifically
permeable to Na
+
and K
+
open for short periods of
time as a result of changes in membrane potential
induced by the advancing wave of the action potential
(Chapter 30). We have already considered the struc-
ture of a potassium ion channel (Fig. 8-21). However,
there are at least 30 types of K
+
channels that can be
distinguished.
502
Many of these appear to have similar
channel structures but to serve a variety of purposes.
While the K
+
channels of neurons are voltage gated
many others are controlled by hormones, neurotrans-
mitters, or mechanical stimuli.
503
One of the most
investigated K
+
channels, known as K
ATP
, sets the
resting potential in the insulin-secreting cells of the
pancreas by facilitating a flow of K
+
into cells. Such
channels, which help equilibrate intracellular and
extracellular K
+
at near equilibrium, are found in cells
with low negative values of E
m
. They are called in-
wardly rectifying. When the internal glucose concentra-
tion in the cells rises it initiates a complex signaling
sequence involving blockage of the K
ATP
channels by
ATP, opening of voltage-sensitive Ca
2+
channels, and
insulin secretion.
504 506
The K
ATP
channel is also
blocked by sulfonylureas. As a result, these com-
pounds induce insulin release and are useful in treat-
ment of diabetes meltitus (Box 17-G). When its gene
was cloned the sulfonylurea receptor was found to
be a transmembrane protein of the ABC transporter
family.
505
The potassium channel protein is another
subunit of the transporter. Similar K
ATP
channels are
found in the kidneys
507
and also in embryonic cells in
which they may participate in regulation of the cell
cycle.
508
Voltage-regulated sodium channels are the major
participants in propagation of nerve impulses. The
large 260-kDa subunit of the sodium channel of nerve
membranes contains four homologous repeat sequences,
each of which may form transmembrane helices and
also contain a loop that may participate in forming a
pore similar to the K
+
pore of Fig. 8-21.
509-510a
How-
ever, the structure is uncertain.
511
The channel complex
also contains 36- or 33-kDa
1
and
2
subunits that
appear to be members of the Ig superfamily.
Epithelial cells contain a quite different Na
+
chan-
nel that participates in reabsorption of urinary Na
+
and in control of blood pressure (Box 22-D). The
channel consists of at least three structurally similar
subunits, each with two predicted transmembrane
helices and a large extracellular domain.
512
These
channels are blocked specifically by the diuretic com-
pound amiloride.
513
C. The Transport of Molecules through Membranes
N
N Cl
H
2
N NH
2
C
N
C
NH
2
NH
2
O
+
H
Amiloride, a diuretic
422 Chapter 8. Lipids, Membranes, and Cell Coats
The best known sodium channel is, in fact, a gen-
eral cation channel which is part of the nicotinic
acetylcholine receptor. Acetylcholine is the principal
excitatory transmitter in the peripheral nervous sys-
tem and upon release from synaptic endings or neuro-
muscular junctions occupies receptors on the
postsynaptic membranes of one or more adjacent
neurons. When acetylcholine binds in the ion pore,
the acetylcholine receptor opens and cations flow out,
depolarizing the membrane. Under favorable circum-
stances, this initiates a nerve impulse (action potential)
in the postsynaptic neuron. The receptor gene was
first cloned from Torpedo and the receptor protein has
been studied extensively.
382
The 290-kDa protein
consists of five similar-sized subunits with an
2

structure. These form a nearly symmetric fivefold


oligomer with a pore in the center. Images of both the
open and the closed states, obtained by electron mi-
croscopy at a resolution of 0.9 nm, suggest that the
inner pore of ~ 2.6 nm diameter is formed by five
helices, one from each subunit. It is open when acetyl-
choline binds to the two subunits and closes to a
much smaller diameter when the acetylcholine leaves
(and is destroyed by hydrolysis).
Calcium channels are a third major group of
cation-selective channels.
514
As pointed out in Box 6-D,
calcium ions are involved in a very wide range of
signaling functions. These are discussed in several
places in this book. Several of these functions depend
upon voltage-gated Ca
2+
channels. Muscle is rich in
L-type or DHP-sensitive channels (Box 6-D) which
play a role in transmission of nerve impulses to muscles
by allowing rapid flow of calcium ions into cells from
outside.
515
The structure appears to be homologous to that
of voltage-gated Na
+
channels with a large 170-kDa
subunit with a fourfold repeat plus smaller subunits.
The ryanodine receptors of muscle control the
release of Ca
2+
from stores in the endoplasmic reticu-
lum
516,517
(see also Chapter 19). These receptors are
ligand gated, being activated by cyclic ADP-ribose
(Chapter 11). Two additional types of voltage-sensi-
tive Ca
2+
channels, N and P, are found in the central
nervous system.
514,518
Another ligand gated calcium
channel has been found in endothelial cells.
519
It is
activated by sphingosylphosphocholine
519
rather
than by cAMP-ribose or inositol triphosphate.
520
A
sodium / calcium ion exchanger and cotransporter
521
utilizes the Na
+
gradient to exchange three external
Na
+
ions for one internal Ca
2+
.
Many aspects of calcium function are poorly un-
derstood. Among these is the role of a group of pro-
teins known as annexins (formerly lipocortins,
calpactins, endonexins, etc.).
522
The ten or more mem-
bers of the annexin family
523 527
share the property of
binding to phospholipid membranes in the presence of
Ca
2+
. One of the several proposed functions of annex-
ins is formation of Ca
2+
channels, a function that is
suggested by the modular three-dimensional struc-
tures.
525,526
Other suggested functions include roles in
membrane fusion, exocytosis, and adhesion.
Active transport of cations. Most organisms
take up ions from their surroundings by active trans-
port. Green plants extract essential nutrients from the
extremely dilute solutions in contact with their roots.
Microorganisms such as yeast and bacteria have the
same ability, and specific concentrating systems for
many ions such as K
+
, Ca
2+
, sulfate, and phosphate
have been identified. The skin of a frog can take up
Na
+
from a 10
5
M solution of NaCl and extrude it into
the internal fluids whose Na
+
concentration may be
greater than 0.1 M. Ions can also be concentrated from
internal fluids and excreted at higher concentrations.
Some seabirds and marine animals rid their bodies of
excess salt by secretion from salt glands. The lining of
the human stomach is able to concentrate hydrogen
ions in gastric juice to ~ 0.16 M.
Organelles within cells have their own ion-concen-
trating mechanisms. Thus, mitochondria can concen-
trate K
+
, Ca
2+
, Mg
2+
, and other divalent metal ions as
well as dicarboxylic acids (Chapter 18). The entrance
and exit of many substances from mitochondria ap-
pear to occur by exchange diffusion, i.e., by secondary
active transport. Such ion exchange processes may
also occur in other membranes.
ATP-driven ion pumps. Within virtually all cells
the sodium concentration is relatively low, while that
of potassium is high (Box 5-A). One theory
528
regards
the cytoplasm as analogous to an ion exchange resin
with fixed charges in a lattice. Highly crosslinked ion
exchange resins exhibit specificity toward binding of
certain ions; e.g., sulfonic acid resins tend to bind K
+
preferentially, while phosphonic acid resins tend to
bind Na
+
. Do proteins also prefer K
+
to Na
+
?
In contrast to the ion exchange theory, much evi-
dence indicates that cells have an active ion pump
that removes Na
+
from cells and introduces K
+
. For
example, the cytoplasm of the giant axons of nerves of
squid can be squeezed out and replaced by ionic solu-
tions. Erythrocyte ghosts can be allowed to reseal with
various materials inside. Ion transport into or out of
cells has been demonstrated with such preparations
and also with intact cells of many types. Such trans-
port is blocked by such inhibitors as cyanide ion,
which prevents nearly all oxidative metabolism. How-
ever, the cyanide block can be relieved by introduction
into the cells of ATP and other phosphate compounds
of high group-transfer potential.
Uptake of K
+
by cells and extrusion of Na
+
from
cells are also specifically blocked by cardiac glyco-
sides such as ouabain (Fig. 22-12). Ouabain labeled
with
3
H binds to the outer surface of cells, and from
423
this binding it was estimated that erythrocytes possess
100 200 ion pumping sites per cell (~ 1 site / m
2
).
529
For the HeLa cell (a widely studied strain of human
cancer cells) 10
5
to 10
6
sites / cell (~ 10 / m
2
) were
found. Further experiments showed that in the pres-
ence of Na
+
within the cell and K
+
on the outside of
the cell, ATP is hydrolyzed. The rate of hydrolysis was
directly related to the concentrations of these two ions
and to the number of ouabain binding sites and also
required the presence of Mg
2+
. These observations led
to the concept of an (Na
+
+ K
+
)- activated ATPase
(often abbreviated Na
+
, K
+
-ATPase) as synonymous
with the membrane-bound ion pump. Within the cell
Na
+
must be located on one side of the membrane and
K
+
on the other to activate this enzyme. However, the
purified enzyme would be expected to hydrolyze ATP
in the test tube in the presence of Na
+
+ K
+
+ Mg
2+
.
Such a protein was isolated from several sources and
has been studied intensively. It is an mixed dimer
with molecular masses of ~ 113 kDa for the chains
and ~ 55 kDa for the glycoprotein chains.
530,531
The proteins may associate to
2

2
tetramers in
membranes. The genes for various isoforms of the
proteins from several sources have been cloned and
sequenced.
531,532
The large subunit may span the
bilayer of the membrane as many as ten times; the
glycoprotein subunit is thought to be largely on
the outer surface and may have only one membrane-
spanning helix.
533,534
A small 68-residue subunit
copurifies with the pump protein. It may be involved
in control of the ATPase, which has complex regulatory
properties.
535,536
Sulfatides (ceramide galactose-3-
sulfate) may also play a role in the enzymatic
activity.
535
The sodiumpotassium pump displays a curious
stoichiometry. Three sodium ions are pumped from the
inside and two potassium ions from the outside of a cell for
each molecule of ATP cleaved. Thus, an excess of positive
ions is pumped out with the result that a negative
charge develops inside the cell and a positive charge
accumulates on the outside. This action of the Na
+
,
K
+
-ATPase is the primary source of the membrane
potential for most eukaryotic cells and is said to be
electrogenic. Because the cell membrane is somewhat
permeable to K
+
, outward diffusion of K
+
through the
leaky membrane along its concentration gradient
helps to maintain the membrane potential as does
inward leakage of Cl

. At the same time, Na


+
diffuses
inward, aided by the membrane potential. Even
though the permeability of Na
+
is low, a steady state
is reached at which the rate of passive inward diffu-
sion of cations just balances the membrane potential
set up by the active transport.
The energy for transport of Na
+
and K
+
by the ion
pump is supplied by ATP. The Na
+
,K
+
- ATPase does
not merely catalyze the hydrolysis of ATP but also
couples its cleavage to the pumping of the ions. The
pumping of sodium and potassium ions is one of the
most important energy-requiring activities of cells. It
is said to account for 23 % of the ATP utilization in a
resting human. Thus, it constitutes an important fraction
of the basal metabolic activity.
The Na
+
,K
+
- ATPase is one of a family of over 50
ion pumps that are characterized by transfer of a
phospho group from ATP to an aspartate side chain
carboxylate in the invariant sequence DKTG to give an
intermediate phosphoenzyme + ADP.
530,537
Phosphoenzyme
intermediate of
Na
+
,K
+
- ATPase
These P-type ATPases are characterized by phospho-
enzyme intermediates, by a conserved consensus
sequence, and through inhibition by vanadate ion.
537 539
The structures are poorly known. Some consist of
single chains (perhaps dimerized) and some have
more than one chain. However, the major subunit
always appears to have about ten transmembrane
helices with a large ~ 430-residue cytoplasmic domain
between the fourth and fifth helices. This domain
contains the ATP binding site and the phosphoaspartyl
group of the phosphoenzyme.
534
This is Asp 369 for
the Na
+
,K
+
- ATPase.
In addition to the Na
+
,K
+
- ATPases there is a very
active Ca
2+
-ATPase which transports two Ca
2+
from
the inside of cells to the outside while returning two
H
+
from outside per ATP.
540 543a
This is the primary
transporter by which cells maintain a low internal
[Ca
2+
]. During its action it becomes phosphorylated
on Asp 351. However, in neurons, in which the mem-
brane potential is maintained at a high negative value
by the sodium pump, an Na
+
/ Ca
2+
ion exchange plays
an even more important role.
540
Other P-type ATPases include the gastric H
+
,
K
+
- ATPase, which acidifies the stomach and has a
high degree of sequence homology with the Na
+
,
K
+
- ATPase.
544
Secretion of HCl into the stomach
apparently involves diffusion of K
+
together with Cl

from the bloodstream through the cells lining the


stomach. The K
+
is then pumped back into these cells
in exchange for H
+
by H
+
,K
+
-ATPase.
545
The chloride
channel may be in the same protein as the (K
+
+ H
+
)
pump.
546
The kidney is the principle acid excretory
organ of the body and as such also contains proton
pumps. An electrogenic H
+
- ATPase pumps H
+
alone
outward through the plasma membranes of fungi and of
green plants.
547,548
The resulting proton gradient may
be used to provide energy for transport of other materials
into cells. A group of metal ion P-type transporters
carry copper and other nutrient ions into cells and
C
CH
2
C
O
O
PO
3
2
N
C O
H
H

C. The Transport of Molecules through Membranes


424 Chapter 8. Lipids, Membranes, and Cell Coats
extrude Cd
2+
and other toxic ions.
539
Some alkaliphilic
bacteria pump Na
+
to create a sodium ion gradient.
539a
All of these ion pumping systems require MgATP as
the source of energy and function via phosphoenzyme
intermediates. The ionic gradients generated can be
used to move other ions or nonionic compounds into
or out of cells by exchange or cotransport processes.
For example, internal H
+
may be exchanged for exter-
nal Na
+
in an exchanger-mediated process that assists
in control of cytoplasmic pH.
549,549a
The reverse process
in E. coli
549b
and many other bacteria provides the
principal mechanism by which those cells export Na
+
.
This exchange is driven by the electrochemical gradi-
ent of the H
+
ion created by oxidative phosphorylation
(Chapter 18). The Na
+
ion, in turn, can be used by
bacterial cells to drive other uptake processes, e.g.,
sugar or amino acid-Na
+
cotransport.
What is the mechanism by which ATPase trans-
porters function? We still do not know.
550
The pump-
ing cycles for the Na
+
,K
+
- ATPase
and the Ca
2+
- ATPase are similar
although different in details. The
ATPases are reversible and with
suitable ionic gradients will work
as ATP synthases.
551
A strictly
hypothetical model for the Na
+
,
K
+
-ATPase is shown in Fig. 8-25.
There are at least two conformations
of the ion pump proteins.
552,552a
In
one conformation the protein binds
three sodium ions tightly, while in
the other conformation it binds two
potassium ions. The ATP operates
the motor that carries out the
conformational changes. In Fig. 8-25
the ion pump, in conformation A, is
shown embedded in a membrane.
In the center, perhaps between three
or more transmembrane helices,
there is a narrow cavity, perhaps
resembling that of the K
+
channel
(Fig. 8-21), into which chelating
groups (e.g., C = O groups of the
peptide chain) protrude. These
groups form the three binding sites
for the 0.19-nm-diameter Na
+
ion.
The spontaneous binding of the
sodium ions triggers a phosphoryla-
tion reaction by which a phospho
group from the MgATP
2
complex is
transferred to the side chain carboxyl
of the active site aspartate. This
phosphorylation in turn triggers a
change to the second conformation
in which the channel to the outside
is open and that to the inside is
closed. At the same time the affinity
for Na
+
is decreased over 100-fold and the sodium ions
dissociate on the outside. The affinity for Na
+
may
decrease because the diameter of the pore is increased
to accommodate the larger 0.27-nm diameter of K
+
ions, perhaps by a twisting motion of the peptide
chains that form the channel.
The next step is loading with two K
+
ions. The
affinity for K
+
in the second conformation is high. The
return to conformation 1 with release of K
+
to the inside
is triggered by hydrolytic removal of the phospho group
as inorganic phosphate (P
i
). It may seem surprising
that a channel could be opened and closed so readily
with synchronous changes in the number and speci-
ficity of ion binding sites. However, recall the type of
structural alteration occurring upon oxygenation of
hemoglobin (Fig. 7-25). Rotation of the hemoglobin
subunits with respect to one another causes small
changes in the geometrical relationships of groups
protruding into the central channel. This strongly
Figure 8-25 A strictly hypothetical model of a Na
+
+ K
+
pump which operates
by ATP-driven opening and closing of a channel at opposite ends and with
alternate tight binding of Na
+
and K
+
.
C
OPO
3
2
O
MgADP

C
OPO
3
2
O
ATP
ATP
Outside Inside
Large
subunit
Glycoprotein
The Na
+
-binding conformation 1.
Projecting groups create 0.2 nm
binding sites in central pore
E
1
ATP
C
O
O

C
O
O

The Na
+
ions are now bound in
the pore
The carboxyl group has been
phosphorylated. The negative
charge may help to bind the
Na
+
ions more tightly
3Na
+
A conformational change
alters geometry
H
2
O
ATP
P
i
MgATP
The K
+
-binding conformation 2.
0.3 nm K
+
-binding sites are now
present; Na
+
ions diffuse out
Two potassium ions are now
bound
Hydrolysis of acyl phosphate
linkage induces a change to
original conformation 1
Potassium ions diffuse into cells
Ouabain
binds and
inhibits
3Na
+
2K
+
2K
+
E
1
ATP
E
1
ATP
E
1
ATP (Na
+
)
3
E
1
P (Na
+
)
3
E
2
P
E
2
P (K
+
)
2
E
2
(K
+
)
2
C
O
O

C
OPO
3
2
O
425
affects the binding of 2,3-bisphosphoglycerate. Very
small movements could open up the Na
+
binding
groups and create new binding sites for the larger K
+
ion, using in part the same chelating groups.
Images of both the Ca
2+
-ATPase (Fig. 8-26)
553
and
the H
+
-ATPase of Neurospora plasma membranes
548
at
0.8 nm resolution reveal similar transmembrane re-
gions and large cytoplasmic domains which are some-
what differently organized. The picture in Fig. 8-26
has been greatly clarified by determination of the
structure by X-ray diffraction to a resolution of
0.26 nm.
553a,b,c
Two calcium-binding sites have been
located in the transmembrane domain between the
helices marked M4, M5, M6, and M8. The Ca
2+
ions
are apparently coordinated by side chains of Asp, Glu,
Gln, and Thr. There are three cytosolic domains. The
site of phosphorylation, Asp 351, lies within a large
~ 27 kDa P (phosphorylation) domain adjacent to the
membrane. The ATP is held by a nucleotide-binding
N domain which must at some point in the cycle move
close to Asp 351 for phosphorylation to occur. The
third cytosolic (A, actuator) domain is thought to be
involved in control of the conformational alterations.
The nucleotide-binding domain lacks the P loop
characteristic of many ATPases and GTPases (see p. 648),
but is homologous in its sequence to L-2-haloacid
dehalogenase (Eq. 12-2)
Two other types of proton-pumping ATPases are
considered in Chapter 18. One is the mitochondrial
F
1
F
0
ATPase, which ordinarily operates in the reverse
direction as the bodys principal ATP synthase. The
other type, which in some ways resembles the mito-
chondrial F
1
F
0
ATPase, is the vacuolar ATPase (V-
ATPase). These are true proton pumps which acidify
vacuoles of plants and also lysosomes and phagocytic
vacuoles.
554,555
They are also considered in Chapter 18.
6. Exocytosis, Endocytosis, and the Flow of
Membrane Constituents
Observation of cells under the microscope with
time-lapse photography reveals that the plasma
membrane as well as the mitochondria and other
organelles are in a constant state of motion. Mitochon-
dria twist and turn and the surface membrane undu-
lates continuously. Vesicles empty their contents to the
outside of the cells, while materials are taken into cells
through endocytosis. In addition, chemical evidence
indicates a directed flow of the materials of which
membranes are constructed from the endoplasmic
reticulum (ER) to the Golgi vesicles, excretion gran-
ules, and plasma membrane (see Fig. 10-8). Along this
route new materials are inserted from the cytoplasmic
side of the membrane, while enzymes within the
vesicles add glycosyl units and make other modifica-
tions. The plasma membrane surface area grows quite
rapidly. In secretory cells fusion of secretion granules
with the plasma membrane also adds additional
material to the membrane.
Counterbalancing this expansion of the plasma
C. The Transport of Molecules through Membranes
Figure 8-26 The structure of the 994-residue Ca
2+
-ATPase
of the endoplasmic reticulum of rabbit muscle at 0.8-nm
resolution. (A) Predicted topology diagram organized to
correspond to the electron density map prepared by electron
crystallography of frozen-hydrated tubular crystals. The
number of amino acid residues in each connecting loop is
marked. (B) The electron density map with the predicted
structure embedded. The relationships of the helices in (B)
to those in (A) are not unambiguous. The helices marked B,
D, E, and F in (B) may form the Ca
2+
channel. The large
cytoplasmic loops, which are black in (A), were not fitted.
From Zhang et al.
553
Courtesy of David L. Stokes.
B
A
cytoplasm
membane
lumen
9
11
32
21
6
11 9
56
11
ATP
400
110
N
C
M
1
M
2
M
3
M
4
M
5
M
6
M
7
M
8
M
9
M
1
0
S
2
S1
S
3
S
4
S
5
426 Chapter 8. Lipids, Membranes, and Cell Coats
membrane is active endocytosis of fluids and of solid
materials from outside the cell. This not only brings
new materials into the cell but also accomplishes
removal of material from the plasma membrane and
partial recycling of its components. One form of
endocytosis is seen with the ameba.
555a
The cytoplasm
flows around a smaller organism or other particle of
food enclosing it in an internal membrane-bound
compartment (endocytic vacuole or endosome).
This vacuole then fuses with lysosomes which supply
the necessary enzymes to digest the food. In a similar
way phagocytic cells of our bodies engulf micro-
organisms or other particles to form phagosomes
which, over a period of more than 24 hours, undergo
extensive biochemical changes.
556
They acquire
digestive enzymes, vacuolar ATPase,
557
other proteins
needed to kill bacteria, and other parasites.
Uptake of smaller particles including protein
molecules occurs by micropinocytosis, a process that
can be seen only by electron microscopy. This often
takes place via coated pits, indentations of ~ 0.3 m
diameter underlain by a thickened membrane.
558 559b
The pit membrane is also coated with protein mol-
ecules and appears to have many short bristles or
spikes protruding into the cytoplasm (Fig. 8-27A).
After endocytosis the coated pits become coated
vesicles of 0.15 0.25 nm (Fig. 8-27B). Within a few
seconds, however, these vesicles lose their coat and
become endosomes.
The major protein making up the coat is the 180-
kDa clathrin, but smaller 33 - to 36-kDa peptides of
several types also contribute.
561
The coat forms a
basket with pentagonal and hexagonal faces sur-
rounding the lipid bilayer of the vesicle. At each
vertex of the basket is a triskelion, a trimer of
clathrin together with an equal number of the smaller
chains. The smallest baskets consist of 12 pentagons
plus 4, 8 or more hexagons, a relationship that allows
formation of a variety of larger baskets.
562,563
Addi-
tional 50- and 100-kDa accessory proteins form a shell
around the clathrin cage. That clathrin is essential for
normal cell growth has been established by the
observation that deletion of its structural gene from
yeast is lethal.
564
The addition of more trimer units
from a reserve of soluble clathrin in the cytoplasm
allows the vesicles to develop and break off from the
membrane. From studies with inhibitors it is evident
that metabolic energy is required to drive the process.
Other vesicles are surrounded by nonclathrin mem-
brane coats. Some of these originate from caveolae
(little caves), which act in endocytosis, exocytosis, and
transmembrane signaling.
564a,b,c
A coatomer complex
of eight subunits with molecular masses from 20 to 60
kDa coats vesicles involved in transport between
compartments of the Golgi.
565 567
What is inside a coated vesicle? Cells take up a
variety of peptide hormones and proteins. This usual-
ly occurs with the aid of specific receptor proteins
located in or on the outside of the plasma membrane.
Some of these, e.g., receptors for the low-density lipo-
protein of plasma (Chapter 22), are clustered in coated
pits. Other receptors, such as those for insulin or epi-
dermal growth factor, are spread more evenly across the
membrane but collect in coated pits when the hormone
binds. Endocytosis provides a means for the cell to
take up and in some cases destroy the hormone or the
receptor or both.
Transmembrane proteins, including hormone
receptors, are incorporated into coated vesicles with
the help of clathrin adapter proteins (APs). These
Figure 8-27 (A) Region of a coated membrane from fibro-
blasts at an intermediate stage of the budding process,
demonstrated by deep etching and rotary replication (by
J. E. Heuser
559b
). From Pearse and Bretscher.
560
Courtesy of
Barbara Pearse. (B) Three structures identified among the
smallest coated vesicles. Structure (a) contains 12 pentagons
and four hexagons, the latter lying at the vertices of a tetra-
hedron; (b) has a barrel shape built of 12 pentagons and eight
hexagons; structure (c) also has twelve pentagons and eight
hexagons, but the latter are arranged in two arcs of four,
related in the same way as the two parts of a tennis ball.
Larger coats seem to be constructed on similar principles,
with the addition of further hexagons. From Pearse and
Bretscher.
560
A
B (a) (b) (c)
~ 0.1 m
427
complex oligomeric proteins bind to recognition or
sorting sequences such as dileucine on YXX (Y = Tyr,
X = any amino acid, = bulky hydrophobic).
567a d
The
adapter proteins also bind to clathrin, the N-terminal
-propeller domain associating with the sequence
LXD / E of some AP adapters.
567c
Completion of a
coated vesicle requires membrane fusion as the vesicle
is pinched off from the membrane surface. A GTPase
(Chapter 11) called dynamin as well as another pro-
tein endophilin I are required. Endophilin I is an
acyltransferase able to transfer the fatty acyl group
of arachidonoyl-coenzyme A to lysophosphatidic acid
in the cytosolic surface of the membrane. This may
change the curvature of the membrane, assisting in
vesicle formation.
567e
Once inside a cell the vesicles lose their coats to
become endosomes which may then fuse with lysosomes
or with Golgi membranes. The removal of a clathrin
coat requires ATP as well as the chaperonin Hsp 70
(Chapter 10) and a coat protein called auxilin.
568
Triskelion is distorted and displaced from the clathrin
cage. The interior of the newly formed endosome is
quickly acidified by the action of a proton pump in the
vesicle walls.
554,569
This sometimes leads to dissociation
of enclosed receptors from their ligands and permits
recycling of receptors and lipids of the vesicle mem-
branes to the cell surface. This is the case for the low-
density lipoprotein receptor.
570,571
Exocytosis, by which the content of a secretion
vesicle is released to the outside of a cell, is just as
important as endocytosis. The process is sometimes
very specialized. For example, the release of a nema-
tocyst from Hydra (Fig. 1-13) can occur in about 3 ms.
572
Exocytosis involves fusion of membranes,
562,573
a
process also occurring during the movement of endo-
somes along the endocytic pathway,
574
during vesicular
transport between Golgi compartments, and in many
other biological processes. Exocytosis is often triggered
by the binding of Ca
2+
to specific proteins of the
vesicle wall and of the cytoskeleton (Chapter 7).
575
The fusion of membranes at several stages in the
vesicle-mediated transport of materials between
Golgi compartments requires a specific protein known
as the N-ethylmaleimide-sensitive fusion protein
(NSF).
576,577
A host of other specialized proteins are
also involved
562,578
and are discussed in Chapter 10. See
also Chapters 20 and 29.
D. Communication
External coats and cell walls help to control the
access of materials to a cell. However, it is the outer
surface of the plasma membrane that makes the cells
first contact with nutrients, hormones, and other
important chemicals. The membrane must often not
only detect these materials but also send signals to the
E. The Extracellular Matrix and Cell Walls
interior of the cell and sometimes to adjacent cells.
These signals may be about changes in pH or nutrient
concentration or the presence of hormones, neuro-
transmitters, or harmful materials. For these reasons
cell membranes contain many embedded receptors
and signaling complexes. These are discussed in
Chapter 11 and later sections of the book.
The plasma membrane also contains many mark-
ers of the individuality of the species or of an indi-
vidual. These are chemical groupings that, in higher
animals, can be recognized by the immune system as
self rather than as a foreign invader. These surface
markers may also be used by parasites as camouflage
to evade the immune response of the host. Such
chemical groupings on cell surfaces are often de-
scribed as antigenic determinants and the molecules
that carry them as antigens. Each antigenic determi-
nant elicits the production of antibodies that will bind
specifically to it. Over 250 different antigenic groups
have already been described for the surface of the red
blood cell. They determine the blood type. Groups on
the surfaces of other cells determine whether a trans-
planted tissue will be rejected. Various proteins from
plant and other sources act as agglutinins by binding
to surface groups much as do antibodies. Viruses that
attack cells may also become adsorbed onto specific
surface molecules, which act as receptors.
Immunoglobulins can also be receptors. For
example, molecules of IgE bound to basophils and the
related mast cells of tissues serve as receptors for
allergens. Binding of an allergen to the IgE molecules
stimulates the release of granules containing histamine
and other substances (Chapter 31).
E. The Extracellular Matrix and Cell Walls
The surroundings of cells are extremely complex
and vary from one organism to another and from one
tissue to another. The principal function of cell walls
and other surface coats is to protect cells against attack
by organisms and against physical disruption.
1. The Structure of Bacterial Cell Walls
The plasma membrane of bacterial cells, other
than the wall-less mycoplasmas and some
archaebacteria, is surrounded by a multilayered wall
which may be separated from the membrane by a thin
periplasm (or periplasmic space). This can be seen
most clearly in suitably prepared thin sections of cells
of E. coli or other gram-negative bacteria as a relatively
empty space of 11- to 25-nm thickness (Fig. 8-28).
579 581
The volume of this space (which may be filled with
gelled material) depends upon the osmotic pressure of
the medium. In E. coli it contains 20 40% of the total
428 Chapter 8. Lipids, Membranes, and Cell Coats
cell water. In gram-negative bacteria the innermost
layer of the walls lies within the periplasm (Fig. 8-28).
It is a porous network of a highly crosslinked material
known as peptidoglycan or murein. The backbone of
the peptidoglycan is a -1,4-linked polymer of alter-
nating N-acetylglucosamine and N-acetylmuramic
acid residues. Alternate units of the resulting chitin-
like molecule carry unusual peptides attached to the
lactyl groups of the N-acetylmuramic acid units (Fig.
8-29). The peptide side chains are crosslinked as
indicated in the figure. The peptides vary consider-
ably in structure and crosslinkages.
582,583
In E. coli and
other gram-negative bacteria the peptidoglycan forms
a thin (2-nm) continuous network around the cell, but
in gram-positive bacteria the highly crosslinked
peptidoglycan forms a layer as much as 10 nm thick.
584
The peptidoglycan layer is surrounded by other layers
whose structures vary from one organism to another,
with the outermost antigenic layers being the most
variable.
The outer membrane of gram-negative bacteria.
Outside the murein layer of E. coli and other gram-
negative organisms is a phospholipid-containing
outer membrane which has the thickness and some-
thing of the structure of a typical biological membrane
(Fig. 8-28).
585,586
This membrane is attached to the
peptidoglycan layer with the aid of a small hydropho-
bic 58-residue lipoprotein whose N terminus contains
a glycerylcysteine which carries three fatty acids
connected by ester and amide linkages:
The fatty acid chains are evidently embedded in the
outer membrane as an anchor. About one-third of the
lipoprotein molecules are attached covalently to the
peptidoglycan through an amide linkage between the
side chain amino group of the C-terminal lysine of the
protein and a diaminopimelic acid residue of the
peptidoglycan (Fig. 8-29). Thus, the protein replaces
one of the terminal D-alanine residues of about one in
ten of the murein peptides. There are ~ 2.5 x 10
5
molecules of the bound form of the lipoprotein per cell
spread over a surface area of peptidoglycan of ~ 3 m
2
.
They appear to be associated as trimers located
primarily in the periplasmic space.
589
About 10
5
copies per cell of the previously men-
tioned (Section C,2) larger 325-residue structural
protein, OmpA protein,
350
after its gene symbol ompA
Figure 8-28 Schematic
molecular structure of the
E. coli envelope. Sugar
residues are represented by
ovals and rectangles.
Circles represent polar head
groups of phospholipids.
MDO, membrane-derived
oligosaccharides; KDO, 3-
deoxy-manno-octulosonic
acid (structures for KDO
and heptose are in Fig. 4-
15). From Raetz and
Dowhan.
587
O-Antigen
Outer core
Heptose
KDO
Lipid A
Porin
Peptidoglycan
MDO
Phospholipids
Proteins
Cytoplasm
Inner
membrane
Periplasm
Lipoprotein
Outer
membrane
Lipopoly-
saccharide
C
N
C
N
Peptide
S
O
H
H
O C R
2
O
C R
1
O
O
R
3
O
H
COO
H

Glycerylcysteine at N-terminus
of lipoprotein and linked to
3 acyl groups
429
Figure 8-29 (A) Repeating unit of structure of a bacterial peptidoglycan
(murein). Some connecting bridges are pentaglycine (Staphylococcus aureus),
trialanylthreonine (Micrococcus roseum), and polyserine (S. epidermis).
(B) Schematic drawing of the peptidoglycan of S. aureus. From Osborn.
588
HO
C
NH
HN
NH
N
O
O
O
COO
H
3
C
H
H
NH
H
O
H
OOC
O
CH
3
H
N
O
H
3
C
H
H
O
CH
2
OH
O
NH
O
CH
2
OH
NH
O
O
CH
3
O

l-Ala; sometimes replaced


by l-Ser or Gly
d-Glu; sometimes replaced
by d-Gln or 3-hydroxy-d-Gln
CH
3
O
d-Ala
The COOH of this d-Ala is
linked to the free NH
2
of the
diamino acid in another chain
Note peptide
linkage to
-COOH of
glutamic acid
HO

[-d-GlcNAc-(14)--d-MurNAc-(14)](n = 1070)
n
Polysaccharide
backbone
meso-Diaminopimelic
acid; sometimes replaced
by l,l-diaminopimelic,
Lys, ornithine,
diaminobutyric
acid, or homoserine
In some cases converted
to CONH
2
This NH
2
group is linked to COOH
of d-Ala in another chain. In gram
negative bacteria the linkage is direct
but in many bacteria a short chain
(up to 5) of amino acids intervenes
HO
d
(Gly)
5
GlcNAc
MurNAc
l-Ala
d-GluNH
2
l-Lys
d-Ala
(Gly)
5
GlcNAc
MurNAc
l-Ala
d-GluNH
2
l-Lys
d-Ala
GlcNAc
MurNAc
l-Ala
d-GluNH
2
l-Lys
d-Ala
(Gly)
5
GlcNAc
MurNAc
l-Ala
d-GluNH
2
l-Lys
d-Ala
(Gly)
5
d-Ala
(Gly)
5
GlcNAc
A
B
(outer membrane protein A), are also
present. The outer membrane contains
almost the same number of molecules of
two porins. Together with phospholip-
ids or with the lipopolysaccharide
discussed in the next paragraphs, the
porins and OmpA protein associate in
hexagonal arrays which provide the
basic framework structure of the outer
membrane.
585
Two of the outer mem-
brane proteins have been shown to
contain some of the modified lysine -
aminoadipic acid 5-semialdehyde
(allysine).
590
The aldehyde groups of
allysine are able to form crosslinks to
other proteins as has been well estab-
lished for collagen and elastin in the
human body.
A characteristic feature of the outer
surface of gram-negative bacteria is a
lipopolysaccharide
586
that is anchored
in the outer membrane. It was discussed
briefly in Chapter 4 where the structures
of the repeating oligosaccharides known
as O-antigens are given. Figures 8-28
and 8-30 show the manner in which the
oligosaccharide bearing the O antigen is
attached to a lipophilic anchoring group
that is embedded in the outer membrane
of the bacteria.
591
The anchor, which is
called lipid A, is a -1,6-linked disaccha-
ride of N-acetylglucosamine. It is also
linked both to phosphate groups and to
the fatty acyl groups that fit into the
lipid bilayer of the membrane. In E. coli
and Salmonella typhimurium four mol-
ecules of 3-D-hydroxymyristic acid are
joined by ester and amide linkages to the
two GlcN units (Fig. 8-30). Other fatty
acids, including lauric, myristic, and
palmitic acids, are esterified to the
hydroxyl groups of two or three residues
of hydroxymyristic acid (Fig. 8-30).
Lipid A from other gram-negative
bacteria is similar but with variations in
the fatty acyl group composition and
linkages to the carbohydrate.
592 594
E. The Extracellular Matrix and Cell Walls
430 Chapter 8. Lipids, Membranes, and Cell Coats
Figure 8-30 Structures of the lipopolysac-
charides of the outer membrane of E. coli
and S. typhimurium including the bilayer
anchor lipid A. For structures of L-glycero-
D-mannoheptulose, KDO, and colitose, see
Fig. 4-15.
O
O
O
P HO
HO
O
O
O
O
O
NH
O
O
O
O
O
HO
O
HO
NH
O
HO
O P
O
OH
OH
O
EtNHPKDO
2,4
KDO
KDO
2,7
EtNHPP 4Hep
1,5
7Hep
1,3
Hep1
P
Glc
4
1,3
1,3
3Gal
GlcNH1 1,2
Glc
1,2
Glc
1,
Gal
1,4
Glc
Col 1,4
1,6
GlcNAc
1,3
1,3
Col
n = 040
O-Antigen
Core oligosaccharide
Hep = l-glycero-d-mannoheptose
KDO = 3-deoxy-d-manno-2-octulosonic acid
(Fig. 4-15)
Lipid A
(Enterotoxin)
Many differences are also found in the core oligo-
saccharide (Fig. 8-30) and in the O-antigens.
594 596
Why do cells of Salmonella have a thousand
distinguishable surface antigens, many based on
differences in the O-antigens? The ends of these
carbohydrate clusters are the groups to which the
antibodies in animals clamp themselves if the bacteria
enter the bloodstream. Mutants known as R forms
(because of the growth as rough colonies on agar
plates) completely lack the outer O-antigen. The R
mutants of Salmonella are nonpathogenic, whereas the
smooth strains with intact O-antigen often cause
illness. Perhaps, if the O-antigen has the right cluster
of sugar rings at the ends, the host organism does not
recognize it as dangerous. This is only part of a con-
tinuous battle between the immune system of the
body and camouflaged surfaces of attacking pathogens.
O O
P
O
HO
H H
H
O OH
O O
H
In Bacillus subtilus d-alanine is attached to one of
the green oxygen atoms in at least half of the units
In B. subtilus
-d-glucose is
attached here
Ribitolteichoic acids

n
HO
OH
O
P
O
O H
O O

n
In Lactobacillus arabinosus d-alanine in ester
linkage occupies this position but is replaced
by -d-glucose on about one unit in nine
Glycerolteichoic acids
H
431
The lipopolysaccharides of bacterial surfaces have
been identified as the endotoxins that cause many
of the worst effects of gram-negative bacterial infec-
tions and that are often lethal.
597
Although the name
endotoxin came from the assumption that there was
an internal heat-resistant toxin that was released from
the bacteria, the toxin is part of the bacterial cell wall.
While the O-antigens provide for innumological
recognition, it is the lipid A part that is responsible for
the toxicity and unusually strong fever-inducing
properties of the lipopolysaccharide.
592 594,597
Teichoic and teichuronic acids. The cell walls of
gram-positive bacteria are composed of a thick pepti-
doglycan layer which also contains proteins and
additional polymers known as teichoic acids and
teichuronic acids. In some species these account for
50% of the dry weight of the cells.
598,599
Teichoic acids
are high polymers of the following general types:
They are often attached through phosphodiester
linkages to N-acetylglucosamine or a disaccharide
which, in turn, is attached to muramic acid residues of
the peptidoglycan. Since the teichoic acid is uniformly
distributed in its attachment to the peptidoglycan, it
must be intimately associated with peptidoglycan
throughout the wall.
Teichoic acids are often covalently attached to
glycolipids which are part of the plasma membrane.
For example, the glycerolteichoic acid of Streptococcus
faecium contains about 28 monomer units of glycerol
phosphate, approximately 60% of which carry residues
of kojibiose (Glu1 2Glu) as a phosphatidylkojibiosyl
diacylglycerol membrane anchor.
600
Teichuronic acids
contain uronic acids:
[4(N-acetyl)ManA16Glc1]
n
From Micrococcus luteus
601
From Bacillus megaterium
602
Nearly 800 of the foregoing tetrasaccharide units
are joined to give a large, densely packed, ~ 500-kDa
polymer. The reducing end of the polymer is
covalently attached to the peptidoglycan layer.
Mycobacteria. Most mycobacterial species seem
to be harmless saprophytic soil bacteria of the gram-
positive group. However, tuberculosis (caused by
Mycobacterium tuberculosis) may infect one-third of the
inhabitants of the earth
603
and kills about three million
E. The Extracellular Matrix and Cell Walls
4Glc13Rha14Rha1
3

GlcA1
n
people per year.
604
Leprosy, caused by M. leprae,
affects 10 million or more.
605
Most antibiotics are not
effective against mycobacteria because of the unusual
nature of their multilayered cell walls. The muramic
acid of their peptidoglycans contains glycolyl groups
instead of acetyl groups, perhaps providing extra
hydrogen bonding within the wall.
604
Most character-
istically, they are rich in lipid materials. Among these
are a unique polysaccharide, a highly branched
arabinogalactan, that is covalently attached to
muramic acid residues of the peptidoglycan.
Clusters of arabinofuranosyl units at the nonreducing
ends of the chains are esterified with mycolic acids
(Section A,1).
10,604,605
Mycobacteria also contain
lipoarabinomannans that act as major antigens.
603,606
Some mycobacterial species synthesize small
glycopeptidolipids that may disrupt cell membranes
of hosts.
607
Mycobacteria dont produce toxins of
usual types but cause slow, long-lasting infections.
Other bacterial coats. Archaebacteria not only
have unusual plasma membranes that contain
phytanyl and diphytanyl groups (Section A,3)
608
but
also have special surface layers (S-layers) that may
consist of many copies of a single protein that is
anchored in the cell membrane.
609
The surface protein
of the hypothermic Staphylothermus marius consists of a
complex structure formed from a tetramer of 92-kDa
rods with an equal number of 85-kDa arms.
610,611
S-
layers are often formed not only by archaebacteria but
also by eubacteria of several types and with quite
varied structures.
612 614
While many bacteria carry
adhesins on pili, in others these adhesive proteins are
also components of surface layers.
615
Additional
sheaths, capsules, or slime layers, often composed of
dextrans (Chapter 4) and other carbohydrates, sur-
round some bacteria.
2. The Surroundings of Animal Cells
Cells in the external epithelial layers are always
surrounded by a protective covering of some kind.
Our own skin is made up of specialized cells which
become filled with microfibrils of keratin as they move
outward and become the relatively dry nonliving
external surface (Box 8-F). Internal epithelial cells
secrete protein and carbohydrate materials that form a
thin basement membrane around the exposed parts
of the cells. The connective tissue that lies between
organs and which also includes tendons, cartilage, and
bone consists of a relatively small number of cells
surrounded by a matrix consisting of the protein
fibers collagen and elastin in a ground substance
rich in proteoglycans (Chapter 4).
616 618
In bone, the
calcium phosphate is deposited within this matrix.
432 Chapter 8. Lipids, Membranes, and Cell Coats
ensures that the three chains remain in proper register
in the triple helix while the procollagen is converted
into collagen, a process involving additional
crosslinking.
Before this maturation can occur there must be
other modifications to procollagen. These begin while
the peptide chains are still attached to ribosomes of the
rough ER. Hydroxylases (Chapter 18) localized in the
membranous vesicles of the ER convert some of the
proline and lysine residues of the procollagen chains
into 4-hydroxyproline
625,629,630
and hydroxylysine
(Eqs. 8-6 and 8-7). Lesser amounts of 3-hydroxypro-
line are formed. About 100 molecules of 4-hydrox-
yproline and 50 of 5-hydroxylysine are created in each
1 chain.
Galactosyl units are then transferred onto some of
the hydroxyl groups of the hydroxylysine side chains,
and glucosyl groups are transferred onto some of the
galactosyl groups. This glycosylation may prevent
incorrect association of the procollagen molecules.
The collagens. The most abundant proteins in
the body are the collagens,
619 624
a family of closely
related materials that account for 20% of the total
protein in higher animals. Collagens make up much
of the organic mass of skin, tendons, blood vessels,
bone, the cornea, vitreous humor of the eye, and
basement membranes. They are found in every
metazoan phylum studied. There are at least 16 types
in the human body.
625
Type I collagen, which accounts
for 90% of the total, is the major form occurring in
skin, tendon, and bone. It is synthesized by the
fibroblasts and is excreted into the extracellular space
where it is polymerized into a durable long-lived
material. Collagen II is found exclusively in cartilage
and the vitreous humor of the eyes. Form III is located
in blood vessels and intestines and is prominent in
embryonic tissues. Collagen IV is the major form
found in basement membranes. Some other collagens
and characteristic features are listed in Table 8-4.
All collagens contain the triple-helical structure
shown in Fig. 2-23. Collagen I consists of two chains
of one kind (1) and one of another (2), while most
other collagens have three identical chains. In these
chains over 1000 residues have the characteristic
GlyXY sequence.
624,626
At each end of the rodlike
molecules short segments of the peptides fold into
globular domains. For type I collagen 16 residues at
the N termini and 25 at the C termini form these
domains. Collagens are synthesized as intracellular
precursors known as procollagens. The three chains
of procollagens are much longer than in the mature
proteins and have larger non-triple-helical ends (Fig.
8-31). The C-terminal extensions are crosslinked by
SS bridges.
625,627
Synthesis of the collagen chains
requires at least six minutes after which they are
released into the cisternal space of the ER, associate,
and become crosslinked (Fig. 8-32). This crosslinking
Figure 8-31 Schematic representation of type
I procollagen. The molecule is composed of
two identical pro1 chains (solid lines) and
one pro2 chain (dashed line). In addition to
the central triple-helical region that gives rise
to the collagen molecule, as portrayed in Fig.
2-23(C), the precursor contains amino- and
carboxyl-terminal non-triple-helical domains.
The amino-terminal domain is composed of a
presumably globular region, a short collagen-
like segment, and a non-triple-helical region in
which cleavages by the amino-terminal
protease occur. Interchain disulfide bonds are
limited to the carboxyl-terminal domain. The
short telopeptides at the ends of collagen
chains represent the residual sequences of the
linkage regions between the collagen helix and
the terminal domains. After Prockop
628
and
Byers.
625
N
C
N
C
O O
H
N
C
N
C
O O
H
H
H OH
O
2
H
Proline residue 4-Hydroxyproline residue
NH
3

NH
3
+
Lysine side chain
OH H
O
2
+
(8-6)
(8-7)
Amino-terminal
propeptide
Triple-helical domain Carboxyl-
terminal
propeptide
Collagen-like segment
Nonhelical segment
~1.4 nm
300 nm
Globular
domain
S
S
S
S
0.93 nm
433
Figure 8-32 Scheme summarizing the biosynthesis of a fibrillar collagen by a fibroblast. (A) Assembly of pro- chains in cisternae
of the rough endoplasmic reticulum; posttranslational hydroxylations and glycosylations; association and disulfide bonding of
C-propeptides; and zipper-like folding of the triple helix by nucleated growth. (B) Proteolytic processing of procollagen to collagen;
self-assembly of fibrils by nucleated growth; and covalent crosslinking of the fibrils. The collagen molecule is first shown as a triple
helix and then either as a wavy line to depict a molecule assembling on the surface of a fibril or as a rectangle to depict the quarter-
staggered assay of monomers in a fibril. The proteolytic processing of procollagen and assembly of fibrils may occur within crypts
of fibrils as shown here or perhaps at some distance from the cell. After D. J. Prockop.
628
Within the extracellular space two procollagen
peptidases act to cleave a 35-kDa peptide from the C
terminus
631
and a 20-kDa peptide from the N-terminal
end of each of the three chains of the secreted
procollagen. The amino acid composition of the
peptides removed is quite unlike that of the remaining
collagen monomer (also called tropocollagen) which
contains one-third glycine and much proline.
The three-stranded monomers of collagens I III
are rods of dimensions ~ 1.4 x 300 nm (Fig. 8-23).
When they reach their final location they associate and
become crosslinked to form strong fibers with diam-
eters ranging from 8 nm to 0.5 m. Tendons tend to
contain large fibrils, while those in bone are small.
The smallest 8-nm fibrils must contain about 20 triple
helices in a single cross section but the successive
monomers are staggered by 6.4 6.7 nm (234 residues
for type I collagen) in such a way that 3-nm gaps are
E. The Extracellular Matrix and Cell Walls
left between the ends (Fig. 8-32). These gaps give rise
to the characteristic banding pattern seen in the fibrils
in Fig. 2-23D. Finer analysis of the bands together
with the known sequences shows that the bands also
reflect the locations of residues with charged side
chains.
632
These ionized groups are thought to pro-
vide electrostatic stabilization to the fibrils.
633
The
exact packing of the collagen molecules into sheetlike
or microfibrillar crystalline arrays is still uncertain.
634
However, there is agreement on the staggered
arrangement
634a
and upon the fact that precisely
formed crosslinks with neighboring rods prevent
the gaps from weakening the fibrils.
Crosslinking of collagen is initiated by oxidation
of some of the lysyl and hydroxylysyl side chains from
amino groups to aldehyde groups under the action of
a copper-containing oxidase (Eq. 8-8, Chapter 18). The
aldehyde groups enter into a variety of reactions that
A B
OH OH
O OH
OH OH
OH
OH
O Gal Glc
Gal
A B
Gal
O
Glc
OH
OH
OH
OH
OH
OH
Glc Nac
(Man)
n
OH
Gal
O
S
S
S
S
S
S
S
S
S
S
S
S
434 Chapter 8. Lipids, Membranes, and Cell Coats
lead to crosslinking of the collagen monomers and to
the formation of insoluble fibers.
635
One reaction is
an aldol condensation followed by elimination of
water (Eq. 8-9, step a). If one of the two aldehydes
involved in the condensation is derived from hydroxy-
lysine and the other from lysine, two isomeric conden-
sation products are formed. The aldol condensation
product can react further: An imidazole group from
a histidine side chain can add to the carboncarbon
double bond and (either before or after this reaction)
another lysine side chain can form a Schiff base with
the free aldehyde. The results of these two processes
are summarized in Eq. 8-9, step b. The final product
histidinohydroxymerodesmosine links four different
side chain groups. In other instances, simple Schiff
bases (aldimines) are formed between aldehyde and
-amino groups. If there is an adjacent hydroxyl group
these can isomerize to ketoamines (Eq. 8-10). Two
residues of hydroxylysine, one of which is glycosylated
as shown in Eq. 8-10, are often involved. Borohydride
reduction (Eq. 4-2) and hydrolysis leads to isolation of
dihydroxylysinonorleucine, the predominant product
of such treatment of bone or cartilage.
Crosslinkages reducible with borohydride are
characteristic of newly formed collagen but these
disappear and are replaced by more stable crosslinks as
collagen matures. For example, a 3-hydroxypyridine
that joins three triple helices may be formed from the
reaction of two ketoamine groupings (with elimination
of one glycosylated hydroxylysine residue).
636 639
Similar chemistry can also produce pyrrole crosslinks.
639
C
O
H
H
C
H O
C
H
O
N
N H
C
H
N
N
N
H
2
N
H
b
c
Addition with protonation
on carbon
Formation of Schiff base
a
Aldol condensation,
elimination of water
His
Histidinohydroxymerodesmosine
(8-9)
N
O
O Gal Glu
+

Hydroxypyridine
(pyridinoline)
crosslink
NH
3
+
C
O
(OH)

H
Lysine side chain
O
2,
Cu
(OH)
Lysyl oxidase
(8-8)
The crosslinkages in collagen are not located at
random but are found in certain positions, often toward
the ends of the collagen monomers. Thus, histidine
residues are found only at positions 89, 929, and 1034
in the 1(I) peptide chains. Residue 9 in the N-terminal
globular portion of one chain is linked to residue 946
of another while residue 103 is linked to 1047 in the
globular C-terminal peptide.
640
The variety and number
of crosslinkages vary among different species. As
collagen ages through a lifetime, glycation (Eq. 4-8)
leads to crosslinkages in which two ketoamines are
formed via glycation cyclize.
641
Collagens I, II, and III form fibrils with similar
structures. However, other collagens are longer or
shorter and aggregate in different ways. A pepsin-
resistant part of the collagen V molecules resembles
collagens I, II, and III but it may contain an additional
non-collagenous segment at the N terminus. Collagens
V and XI are quantitatively minor components of the
extracellular matrix but are thought to provide a core
around which the fibrils of collagens I and III may
form.
642
Types XII, XIV, IX, and XVI collagens contain
interruptions in the helix which create bends, flexible
sites, and sites of increased proteolytic susceptibility.
They may link the fibrils to other components of the
surrounding matrix.
643
435
Forming quarter-staggered fibrils
I
a,b
Skin, bone, tendon, dentin Most abundant, banded quarter-staggered fibrils 7, 17
III
a,c
Skin, blood vessels Abundant, small banded fibrils 2
V
a,d
Most interstitial tissues; cartilage, bone Abundant, small fibrils 2
Predominant in cartilage and bone
II
a
Hyaline cartilage, vitreous humor Very abundant, small banded fibrils 12
XI
a
Hyaline cartilage
With interrupted triple helices
XII
a,e
Embryonic tendon, periodental ligaments Fibril associated
XIV
a,e
Fetal skin, tendon Fibril associated
IX
a,e
Cartilage, vitreous humor Minor, contains attached glycosamino-
glycan, fibril associated
XVI
f
Cartilage Fibril associated
Forming sheets and networks
IV
a,g
All basement membranes Nonfibrillar network 13
X
a
Mineralizing cartilage, growth plate Short chain
VIII
a,h
Endothelial cells; Descemets Small helices linked in hexagonal arrays
membrane of the cornea
Forming beaded filaments
VI
a
Most interstitial tissues, intervertebral discs Beaded microfilaments
Forming anchoring fibrils
VII
a,i
Basement membranes Long-chain, antiparallel dimers, anchoring fibrils
a
Martin, G. R., Timpl, R., Mller, P. K., and Khn, K. (1985) Trends
Biochem. Sci. 10, 285-287
van der Rest, M., and Garrone, R. (1991) FASEB J. 5, 2814-2823
Prockop, D. J., and Kivirikko, K. I. (1995) Ann. Rev. Biochem. 64,
403-434
b
Prockop, D. J. (1990) J. Biol. Chem. 265, 15349-15352
c
Nah, H.-D., Niu, Z., and Adams, S. L. (1994) J. Biol. Chem. 269,
16443-16448
d
Myers, J. C., Loidl, H. R., Stolle, C. A., and Seyer, J. M. (1985)
J. Biol. Chem. 260, 5533-5541
TABLE 8-4
Types of Vertebrate Collagen
Type
number Location Characteristics
Gene location:
human
chromosome
e
Shaw, L. M., and Olsen, B. R. (1991) Trends Biochem. Sci. 16, 191-194
f
Myers, J. C., Yang, H., DIppolito, J. A., Presente, A., Miller, M. K.,
and Dion, A. S. (1994) J. Biol. Chem. 269, 18549-18557
g
Hudson, B. G., Reeders, S. T., and Tryggvason, K. (1993) J. Biol.
Chem. 268, 26033-26036
h
Benya, P. D., and Radilla, S. R. (1986) J. Biol. Chem. 261, 4160-4169
i
Lunstrum, G. P., Sakai, L. Y., Keene, D. R., Morris, N. P., and
Burgeson, R. E. (1986) J. Biol. Chem. 261, 9042-9048
E. The Extracellular Matrix and Cell Walls
436 Chapter 8. Lipids, Membranes, and Cell Coats
The basement membrane collagen IV forms a non-
fibrillar network.
644,645
The 400-nm-long molecules
aggregate via their identical ends. Four molecules are
held together through their triple-helical N termini,
while the C-terminal globular domains connect pairs
of molecules. Type IV collagen is also a proteoglycan
with a glycoaminoglycan chain attached to one of its
nonhelical domains. It may become covalently attached
to type II collagen via a hydroxypyridine linkage.
646
Dimers of the microfibrillar collagen VI are formed
from 105-nm-long monomers by antiparallel and
staggered alignment, with the 75-nm overlapping
helical segments twisting around each other to form
coiled dimers. After a symmetrical association of dimers
to tetramers, fibrillar structures are formed by end-to-
end aggregation. The connection of monomers to
dimers, tetramers, and polymers occurs by disulfide
bonds between triple-helical segments and globular
domains. The 450-nm-long collagen VII molecules
associate into antiparallel dimeric structures which show
a 60-nm overlap and which subsequently assemble
laterally with their ends in register.
622
At least 32 genes encode the peptide chains of
vertebrate collagens.
647,648
These chains are assembled
into the 19 known types of collagen. Alternative splicing
of the mRNAs provides additional isoforms.
649
The
collagen 2(I) gene from both the chick and human
DNA is ~ 38 kb in length and consists of 52 exons
separated by introns ranging in length from 80 to 2000
base pairs.
647,650
At least nine of the exons that encode
the triple-helical regions have exactly 54 bp. All are
multiples of 9 bp, i.e., the length needed to encode one
Gly-X-Y triplet (Chapter 2, Section D,4). The signifi-
cance of these observations is unclear but the presence
of so many introns does suggest ways in which col-
lagen sequences could have been transferred into the
genes for such proteins as acetylcholineesterase and the
C1q component of complement.
647
The human 1(I)
collagen gene also consists of 51 segments but they lie
within a shorter 18-kb length of
DNA.
651
A collagen gene from
Drosophila is much less fragmented.
Collagens are found in all meta-
zoan organisms.
652
Invertebrate
collagens play a variety of special-
ized roles.
653
For example, minicol-
lagens from Hydra strengthen the
walls of their nematocysts.
572
Elastic fibers. The elastic
properties of lung, skin, and large
blood vessels are provided by elastic
fibers in the extracellular matrix.
654
The fibers consist of amorphous
material together with the insoluble
protein elastin, which is rich in
glycine, proline, and hydrophobic
amino acids. Its special structure (Chapter 2) provides
elasticity to the fibers. A 72-kDa precursor tropoelastin
is secreted into the extracellular space where it is acted
upon by lysyl oxidase (Eq. 8-7) and crosslinked into
a rubber-like network.
654
Remarkable crosslinkages
are formed. Three aldehyde groups derived by oxi-
dation of lysine side chains combine with one lysine
amino group through aldol condensations, dehydration,
and oxidation to form residues of desmosine and
isodesmosine.
Cartilage and basement membranes. Tendons
consist largely of collagen, but in most tissues the
collagen fibrils are embedded in a matrix of proteogly-
cans and various other proteins.
655 657
Both the core
proteins of the proteoglycans and the attached
N
OOC COO
COO
OOC
H
H
3
N
H NH
3
NH
3
H
H
3
N H
2
Desmosine
This group
shifted to C-2
in isodesmosine

+
+

+
+
N
O
H
OH
Gal Glu
NH
O
OH
Gal Glu
NH
O
O
Gal Glu
Aldimine Ketoamine
(8-10)
437
Figure 8-33 (A) Electron micrograph
of polygonal network formed by associ-
ation of collagen type IV monomers.
(B) Electron micrographs of single
molecules of laminin. (A) and (B) from
Yurchenko and Schittny.
663,663a
Courtesy
of Peter Yurchenko (C) Structural model
of the three-chain laminin molecule
with domain designations. From Beck
et al.
670
Domains I and II are a triple-
helical coiled coil rod which, in the B1
chain, is interrupted by a small cysteine-
rich domain . Sites of cleavage by
cathepsin are marked by arrows.
A
B
C
G1
G2
G3
G4
G5
NH
2
(B1)
NH
2
(B2)
III IV V VI III IV V VI
IIIa
IVa
IIIb
V
VI
NH
2
(A)

I
IVb
polysaccharides interact with collagen fibrils and with
fibronectin and other (previously discussed) cell sur-
face proteins. The cartilage matrix
658,659
consists large-
ly of proteoglycans (Fig. 4-16)
660
and of several
difficult to study, insoluble proteins. One of these is
the 148-kDa cartilage matrix protein, which yields
52-kDa subunits upon reduction.
661,662
It interacts with
both proteoglycans and collagen and may help to
integrate the cartilage matrix.
Basement membranes (Fig. 1-6)
663
function in part
as an exoskeleton that helps keep cells positioned.
However, the thick basement membranes of the capil-
lary walls of the glomeruli of the kidney provide the
ultrafilters that prevent most proteins from entering
the urine. Basement membranes contain large
amounts of collagen IV, which forms a polygonal
network (Fig. 8-33A). A second macromolecular net-
work is formed by the very large 950-kDa cross-
shaped multisubunit protein called laminin (Fig.
8-33B).
664 666
Laminin is one of a series of extracellular
proteins which appear to have arisen by shuffling of
structural modules during evolution.
667
It contains
sites for binding to heparin, to integrins,
668
to the
heparin sulfate proteoglycan agrin (see also Fig. 4-11),
and to the 150-kDa sulfated glycoprotein nidogen
(entactin).
669
At least seven isoforms of laminin, with
varying tissue distributions, are formed, in part as a
result of alternative splicing of the mRNA tran-
scripts.
666
Laminin is rich in EGF-like modules.
665
An
X-ray structure of three of them shows that they form
a continuous rod of complex structure.
321
A smaller
100 kDa basement membrane fibulin also contains
multiple EGF-like repeats.
671
As with other extracellu-
lar structures, crosslinking of laminin and other com-
ponents of basement membranes by transglutaminase
provides additional stability.
672
E. The Extracellular Matrix and Cell Walls
200 nm
100 nm
438 Chapter 8. Lipids, Membranes, and Cell Coats
BOX 8-E GENETIC DEFECTS OF COLLAGEN STRUCTURE
Any major protein of the body is likely to be
associated with a number of genetic problems. In the
case of collagen, the possibility for harmful mutations
is enhanced by the existence of a large number of
genes that encode the more than 16 types of collagen
which are expressed differently in different tissues.
There are known human disorders resulting from
defects in synthesis, secretion, or structure of types I,
II, III, IV, and VII collagens. Other defects involve
lysyl hydroxylase and procollagen N-proteinase.
a d
In the severe lethal form of osteogenesis
imperfecta (brittle bone syndrome) the victims
collagen I may contain an 1 chain lacking as many as
100 residues or a shortened 2 chain. In other cases a
cysteine, arginine, or other amino acid has been sub-
stituted for glycine in the triple-helical region of an 1
chain.
e,f
The cloning of collagen genes has permitted
an exact description and precise location of the defects
in these genes. Alterations toward the N-terminus of
the 1 chain or in the 2 chain often cause a milder
type of osteogenesis imperfecta.
g
Some patients have
deletions in the pro-2 chains of collagen I causing the
chain to be incorporated into the collagen without
removal of the N-terminal or C-terminal peptide to
give a protein with poor stability.
h
In other cases
amino acid substitutions in the 2 chain cause forma-
tion of chains with excessive posttranslational modifi-
cation. Sometimes the 2 chain is not incorporated
into the triple helix and the collagen I formed contains
three 1 chains.
Another well established abnormality of collagen
is found in cattle suffering from dermatosparaxis, a
disease in which the skin is extremely brittle. The
collagen chains are disorganized and have poor fiber-
forming properties. The procollagen peptidase that
cleaves a peptide from the N termini of the chains of
procollagen is apparently defective. A similar human
collagen disease is the EhlersDanlos syndrome,
which in some instances is accompanied with recur-
rent joint dislocations and curvature of the spine. At
least ten different types of the disorder are known.
d
The procollagen peptidase is sometimes lacking.
i
In
other cases a person synthesizes an abnormal pro-2
chain that is resistant to cleavage by the peptidase
because of deletion of the normal cleavage site. In
others collagen is formed in only small amounts or
is degraded rapidly. Some individuals lack lysyl
hydroxylase and others have a defect in mRNA
splicing which causes loss of an exon from the mRNA
and synthesis of shortened pro-2 chains.
j
Somewhat similar symptoms are observed in
lathyrism, a disease which arises when animals ingest
seeds of Lathyris odoratus, the common sweetpea.
Since lathyris peas form part of the diet of some peo-
ples, the condition is also known in humans and often
causes curvature of the spine and rupture of the aorta.
The biochemical problem has been traced to the pres-
ence in the seeds of -cyanoalanine and of its decar-
boxylation product -aminopropionitrile.
Although the mode of action is not certain, this com-
pound is an inhibitor of lysyl oxidase essential to the
crosslinking of both collagen and elastin. A hereditary
defect with a similar effect in the mouse involves a
defect in lysyl oxidase.
k,l
Collagen defects account for a variety of other
skeletal problems
m
including some cases of the com-
mon osteoarthritis.
n
Mice lacking the 1 chain of
collagen IX develop a degenerative joint disease
resembling human osteoarthritis.
o
An inherited defect
in the basement membrane collagen IV is responsible
for the inherited Alport disease in which kidney
filtration is defective.
p,q
Similar symptoms are observed
with the acute autoimmune disease Goodpasture
syndrome and in diabetic nephropathy.
p
a
Prockop, D. J. (1990) J. Biol. Chem. 265, 15349 15352
b
Kuivaniemi, H., Tromp, G., and Prockop, D. J. (1991) FASEB J.
5, 2052 2060
c
Prockop, D. J., and Kivirikko, K. I. (1995) Ann. Rev. Biochem. 64,
403 434
d
Byers, P. H. (1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 3 (Scriver, C. R., Beaudet, A. L.,
Sly, W. S., and Valle, D., eds), pp. 4029 4077, McGraw-Hill,
New York
e
Cohen-Solal, L., Zylberberg, L., Sangalli, A., Gomez Lira, M.,
and Mottes, M. (1994) J. Biol. Chem. 269, 14751 14758
f
Lightfoot, S. J., Atkinson, M. S., Murphy, G., Byers, P. H., and
Kadler, K. E. (1994) J. Biol. Chem. 269, 30352 30357
g
Marini, J. C., Lewis, M. B., Wang, Q., Chen, K. J., and Orrison,
B. M. (1993) J. Biol. Chem. 268, 2667 2673
h
Mundlos, S., Chan, D., Weng, Y. M., Sillence, D. O., Cole, W. G.,
and Bateman, J. F. (1996) J. Biol. Chem. 271, 21068 21074
i
Holmes, D. F., Watson, R. B., Steinmann, B., and Kadler, K. E.
(1993) J. Biol. Chem. 268, 15758 15765
j
Weil, D., DAlessio, M., Ramirez, F., Steinmann, B., Wirtz, M. K.,
Glanville, R. W., and Hollister, D. W. (1989) J. Biol. Chem. 264,
16804 16809
k
Pope, F. M., Martin, G. R., Lichtenstein, J. R., Penttinen, R.,
Gerson, B., Rowe, D. W., and McKusick, V. A. (1975) Proc. Natl.
Acad. Sci. U.S.A. 72, 1314 1316
l
Rowe, D. W., McGoodwin, E. B., Martin, G. R., and Grahn, D.
(1977) J. Biol. Chem. 252, 939 942
m
Freisinger, P., Ala-Kokko, L., LeGuellec, D., Franc, S., Bouvier,
R., Ritvaniemi, P., Prockop, D. J., and Bonaventure, J. (1994)
J. Biol. Chem. 269, 13663 13669
n
Ala-Kokko, L., Baldwin, C. T., Moskowitz, R. W., and Prockop,
D. J. (1990) Proc. Natl. Acad. Sci. U.S.A. 87, 6565 6568
o
Fssler, R., Schnegelsberg, P. N. J., Dausman, J., Shinya, T.,
Muragaki, Y., McCarthy, M. T., Olsen, B. R., and Jaenisch, R.
(1994) Proc. Natl. Acad. Sci. U.S.A. 91, 5070 5074
p
Zhou, J., Hertz, J. M., Leinonen, A., and Tryggvason, K. (1992)
J. Biol. Chem. 267, 12475 12481
q
Gunwar, S., Ballester, F., Noelken, M. E., Sado, Y., Ninomiya, Y.,
and Hudson, B. G. (1998) J. Biol. Chem. 273, 8767 8775
N C CH
2
CH
2
NH
3
+
439
BOX 8-F SKIN
Mammalian skin must be tough, water-resistant,
self-renewing, and rapidly healing. The outer
layers of cells or epidermis consist principally of
keratinocytes, epithelial cells specialized for for-
mation of keratin (Fig. 7-31). In the inner layer of
the epidermis the basal stem cells divide, provid-
ing a constant outward flow of cells which become
progressively flattened, dehydrated, and filled with
keratin fibrils.
a
The outer layers contain only dead
cells which are finally sloughed or abraded from the
surface. Human epidermis is completely renewed
in about 28 days!
About 25 different human genes encode the
keratins of skin and other soft tissues. Others specify
the keratins of hair and nails.
b
Both of these hard
tissues as well as claws, hoofs, beaks, horns, scales,
quills, and feathers are largely keratin. However,
there are additional constituents. During the final
stages of keratinocyte differentiation a 15-nm-thick
crosslinked sheath of protein, the cornified cell
envelope (CE), forms beneath the plasma membrane.
c
Crosslinkages between keratin and other proteins
are formed by the action of transglutaminases.
d f
A specialized protein involucrin, which contains
glutamine-rich repeating sequences, provides many
of the side chain amide groups for the crosslinking
reaction (Eq. 2-23).
g
Loricin, a protein
containing glycine-rich flexible loops,
h
is
also a major partner in these cross-
linkages.
c,h
The histidine-rich filaggrin
undergoes complex processing before
binding to keratin fibrils to provide
another form of crosslinkage.
i,j
Small
proline-rich proteins, desmosonal pro-
teins, and others are also present in the
CE.
c
As the final outer stratum corneum
is formed the phospholipid bilayer
deteriorates and intercellular lipid layers
are formed.
k,l
These contain principally
ceramides, cholesterol, and free fatty
acids. Some sphingolipids are covalently
attached to proteins.
a
Also present in the epidermis are
embedded macrophage-like Langerhans
cells as well as pigmented melanocytes,
cells with highly branched dendrites,
which lie just above the basal stem cell
layer. Each melanocyte contains hun-
dreds of pigmented organelles called
melanosomes. They contain not only
the black or reddish melanin pigments
but also the enzymes needed to form
them (Chapter 25).
n,o
The dendrites of a melanocyte contact about 36
keratinocytes and are able to transfer melanosomes
to these adjacent cells. The numbers and sizes of the
melanosomes as well as melanin structure determine
differences in skin color.
m
Similar cells in amphibians,
the melanophores, also contain light receptors.
p
Their melanosomes are not transferred to other cells
but may be either clustered near the center of the
cell or dispersed. The location can be changed
quickly by transport of the melanosomes along a
network of microtubules allowing the animals to
change in response to changes in light color.
q
Various stimuli, including ultraviolet irradiation of
melanocytes, cause increased synthesis of melanin
with a resultant tanning
o
and added protection
against sunburn.
Beneath the basement membrane of the epidermis
is the dermis, a thick, tough, collagen-rich connec-
tive tissue. Blood vessels and nerve endings are
found in this layer, as are roots of hairs and oil and
sweat glands.
r
Skin suffers from a variety of ailments includ-
ing serious hereditary diseases.
a
One group of
keratinization disorders, known as ichthyoses,
are characterized by thickened, scaly skin. Some
hereditary ichthyoses result from defects in type II
Diagram of a dendritic melanocyte surrounded by satellite kerati-
nocytes. The Golgi area (G), where the melanosomes are synthesized, is
shown around the nucleus. The other branched cell, higher in the
epidermis, is a Langerhans cell with its tennis racquet-shaped granules.
Courtesy of Dr. W. Quevedo, Jr. From Montagna et al.
m
Stratum corneum
Langerhans cell
Basal layer
Basement
membrane
Dermis
Melanocyte
E. The Extracellular Matrix and Cell Walls
440 Chapter 8. Lipids, Membranes, and Cell Coats
Fibrillin and Marfans syndrome. Most connec-
tive tissues contain insoluble, beaded microfibrils 10
12 nm in diameter. A component of some of these
microfibrils, which are often found in elastic tissue,
was purified from media used to culture human fibro-
blasts in 1986. This protein, called fibrillin, is a single-
chain 350-kDa glycoprotein which contains ~14%
cysteine.
673674
Using a DNA probe based on the par-
tially cloned fibrillin gene, the location of the gene was
established on the long arm of chromosome 15 at a site
previously identified as that of a gene defective in
Marfans syndrome. This disorder often causes dislo-
cation of lenses of the eyes and aortic aneurysm as
well as elongated limbs and fingers. Point mutations
in the fibrillin gene have been identified in both
Marfans patients and family members that carry the
defective gene.
675,676
The cuticles of invertebrates. The tough elastic
cuticle of the nematode Caenorhabditis is largely col-
lagen. However, the molecules are smaller than in
vertebrates and there are ~ 100 different genes whose
transcription gives rise to a large variety of similar
proteins.
652
Cuticles of some annelids have unusually
long collagens.
653,677
In contrast, the epithelial cells of
insects and other arthropods secrete chitin which
serves as the framework for development of a thick
and often hard cuticle or exoskeleton. The cuticle also
contains a variety of proteins.
678,679
In some instances
mineralization by calcium carbonate occurs. During
the later phases of the cuticle development extensive
crosslinking of the proteins takes place. This is largely
by reactions between modified aromatic side chains
and resembles the chemistry of formation of melanin
and lignin (Chapter 25).
680
Bones, teeth, and shells. Living organisms are
able to induce the formation of over 60 inorganic com-
pounds.
681
Most of these are formed by animals. Two
forms of calcium carbonate, calcite and aragonite,
predominate.
682
These minerals form shells, exoskeleton
bones, bones, teeth, and other specialized structures.
BOX 8-F SKIN (continued)
a
Roop, D. (1995) Science 267, 474 475
b
Takahashi, K., Paladini, R. D., and Coulombe, P. A. (1995)
J. Biol. Chem. 270, 18581 18592
c
Robinson, N. A., Lapic, S., Welter, J. F., and Eckert, R. L. (1997)
J. Biol. Chem. 272, 12035 12046
d
Kim, S.-Y., Chung, S.-I., and Steinert, P. M. (1995) J. Biol. Chem.
270, 18026 18035
e
Huber, M., Rettler, I., Bernasconi, K., Frenk, E., Lavrijsen, S. P.
M., Ponec, M., Bon, A., Lautenschlager, S., Schorderet, D. F.,
and Hohl, D. (1995) Science 267, 525 528
f
Candi, E., Melino, G., Lahm, A., Ceci, R., Rossi, A., Kim, I. G.,
Ciani, B., and Steinert, P. M. (1998) J. Biol. Chem. 273, 13693
13702
keratin.
b
Lamellar ichthyosis reflects defects in
the crosslinking enzyme transglutaminase.
e,f
Epidermolysis bullosa is a heterogeneous group
of disorders characterized by easy formation of
blisters. One form has been traced to a defect in the
anchoring fibrils of type VII collagen, which tie cells
of the basal layer to the basement membrane.
s,t
Others are defects in keratins of the basal or inter-
mediate layers.
a
Yet others involve the lipid metab-
olism of skin, e.g. a steroid sulfatase deficiency.
a
The most frequent skin disorder, which affects
about 2% of the worlds population is psoriasis.
The thickened, scaly patches can cover much of the
skin and become disabling. The inflammation and
excessive epidermal growth are usually a T-cell
mediated immunologic response to antigenic
stimuli.
p,u,v
However, there is a hereditary form.
u
Other common skin disorders include actinic kera-
tosis induced by light and cancer.
g
Yaffe, M. B., Beegen, H., and Eckert, R. L. (1992) J. Biol. Chem.
267, 12233 12238
h
Hohl, D., Mehrel, T., Lichti, U., Turner, M. L., Roop, D. R., and
Steinert, P. M. (1991) J. Biol. Chem. 266, 6626 6636
i
Resing, K. A., Walsh, K. A., Haugen-Scofield, J., and Dale, B. A.
(1989) J. Biol. Chem. 264, 1837 1845
j
Mack, J. W., Steven, A. C., and Steinert, P. M. (1993) J. Mol. Biol.
232, 50 66
k
ten Grotenhuis, E., Demel, R. A., Ponec, M., Boer, D. R., van
Miltenburg, J. C., and Bouwstra, J. A. (1996) Biophys. J. 71,
1389 1399
l
Bouwstra, J. A., Thewalt, J., Gooris, G. S., and Kitson, N. (1997)
Biochemistry 36, 7717 7725
m
Montagna, W., Prota, G., and Kenney, J. A., Jr. (1993) Black Skin
Structure and Function, Academic Press, San Diego, California
n
Potterf, S. B., Muller, J., Bernardini, I., Tietze, F., Kobayashi, T.,
Hearing, V. J., and Gahl, W. A. (1996) J. Biol. Chem. 271, 4002
4008
o
Romro-Graillet, C., Aberdam, E., Biagoli, N., Massabni, W.,
Ortonne, J.-P., and Ballotti, R. (1996) J. Biol. Chem. 271, 28052
28056
p
Greaves, M. W., and Weinstein, G. D. (1995) N. Engl. J. Med. 332,
581 588
q
Rogers, S. L., Tint, I. S., Fanapour, P. C., and Gelfand, V. I. (1997)
Proc. Natl. Acad. Sci. U.S.A. 94, 3720 3725
r
Martin, P. (1997) Science 276, 75 81
s
Byers, P. H. (1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 3 (Scriver, C. R., Beaudet, A. L.,
Sly, W. S., and Valle, D., eds), pp. 4029 4077, McGraw-Hill,
New York
t
Christiano, A. M., Ryynnen, M., and Uitto, J. (1994) Proc. Natl.
Acad. Sci. U.S.A. 91, 3549 3553
u
Tomfohrde, J., Silverman, A., Barnes, R., Fernandez-Vina, M. A.,
Young, M., Lory, D., Morris, L., Wuepper, K. D., Stastny, P.,
Menter, A., and Bowcock, A. (1994) Science 264, 1141 1145
v
Boehncke, W.-H., Dressel, D., Zollner, T. M., and Kaufmann, R.
(1996) Nature (London) 379, 777
441
While some organisms promote mineral deposition
completely outside of their cell coats, the mineraliza-
tion is usually controlled by the proteins and polysac-
charides lying around and between cells.
683 687
Bone deposition begins in the proteoglycan and
collagen II matrix of cartilage. Later these polymers
are largely replaced by collagen I which accounts for
90% of the organic material in mature bone. Bone
collagen has a distinctive pattern of crosslink-
ing.
639,688,689
Some borohydride-reducible crosslinks
remain throughout adult life.
639
However, complex
hydroxypyridine and pyrrole linkages are more char-
acteristic of bone collagen. Embedded in the bone
matrix are spidery cells, the osteocytes. Among
these are osteoblasts,
689a
which secrete the collagen
and other proteins that promote the laying down of
calcium phosphate. Also present are large multinucleate
osteoclasts
689b
which dissolve bone and reabsorb
calcium and phosphate. Cells of both types remain
active in mature bone, which is both a structural
material and a store of calcium and phosphorus.
How do osteoblasts induce calcium phosphate
deposition? The first crystals laid down within the
cartilage matrix are of carbonate apatite only 2 3 nm
thick and tens of nanometers in length. They are
intimately associated with the collagen and other
components of the matrix
690
and may be formed with-
in matrix vesicles.
691
From observations with large
biologically formed calcium carbonate crystals it is
known that proteins or other organic initiators of
crystallization can be found embedded in mature
crystals. If the same is true in bone what are the initia-
tors? The answer is uncertain but it is known that
without osteoblasts the partially mineralized cartilage
will not become bone.
692
A key to the development of osteoblasts appears
to be an osteoblast-specific transcription factor
OS/2 or Cbfa1.
693 695
Mutations in human Cbfa1 are
linked to a series of skeletal defects.
696
A unique
change accompanying conversion of a precursor cell
into an osteoblast is the formation of a 49-residue
-carboxyglutamate (Gla)-containing protein called
osteocalcin.
697
(See also Box 15-F). It is the most
abundant noncollagenous protein of bone. Its three
Gla residues doubtless help to bind calcium ions and
osteocalcin may be an initiator of crystallization.
Osteocalcin has also been found in fish scales.
698
Also
present in bone is a 74-residue matrix Gla protein
which has 5 Gla residues.
699
Other phosphoproteins, glycoproteins, and
proteoglycans may also be required for mineral-
ization. For example, the 286-residue glycoprotein
osteonectin
323,700,701
accounts for 3% of total bone
protein. It contains two Ca
2+
-binding motifs and
inhibits growth of hydroxylapatite crystals, but its role
in bone development is not clear. One of the phos-
phoproteins of developing bone has been identified as
E. The Extracellular Matrix and Cell Walls
the 24 kDa propeptide cut from the N-terminal end of
the 1 chain of type I procollagen.
702
Another bone
protein, the sialic acid-containing osteopontin has
the cell-binding sequence GRGDS identical to that in
fibronectin.
703,704
Since it also binds to hydroxylapatite
this protein may form a bridge between cell surfaces
and the mineral matrix.
The mineral phase of bone is largely hydroxylapatite,
Ca
10
(PO
4
)
6
(OH)
2
(Fig. 8-34), which is essentially in
chemical equilibrium with the calcium and phosphate
ions present in the blood serum. Thus, bone cells can
easily promote either the deposition or dissolution of
the mineral phase by localized change in pH, concen-
trations of Ca
2+
or HPO
4
2
, or of chelating compounds
such as ATP or inorganic pyrophosphate. Small 100
nm vesicles rich in acidic phospholipids and contain-
ing both Ca
2+
and the enzymes alkaline phosphatase
and pyrophosphatase, may play an essential role in
calcification. Perhaps they release the calcium and
enzymes that generate inorganic phosphate.
691
Bone is noted for its continual remodeling
caused by the action of both osteoblasts and osteo-
clasts.
704a,b
The latter are large multinuclear cells
derived from the same precursors as give rise to macro-
phages.
689b,704
Osteoclasts generate a sealed, acidic
compartment on a bone surface, using a vacuolar
ATPase to pump protons from the cytosol of the osteo-
clast, using H
2
CO
3
as a source of protons.
706
The HCO
3
-
then leaves cells in exchange for Cl
-
which then also enters
the acidified compartment. Not only is the calcium
phosphate dissolved but lysomal enzymes digest the
organic materials. Each osteoclast creates a long tun-
Figure 8-34 An end-on view of a crystallite of hydroxy-
lapatite. The shaded atoms of Ca, P, and O represent an
underlying layer. The OH

groups form a longitudinal


H-bonded array in the center. From J. A. Weatherell and
C. Robinson,
705
p. 55. A small amount of Mg
2+
is present in
place of Ca
2+
and a very small fraction of the OH

is replaced
by fluoride ion which has a bone strengthening effect.
Ca Ca
Ca
Ca Ca
Ca
Ca
Ca
Ca
OH
P
P
P
a
b
442 Chapter 8. Lipids, Membranes, and Cell Coats
BOX 8-G THE BIOCHEMISTRY OF TEETH
Like other bones, mammalian teeth are com-
posed largely of collagen and hydroxylapatite. The
much studied rat incisor is 65% mineral; about 85%
of the organic material is type I collagen. A tooth
consists of three mineralized tissues together with
the internal blood vessels and nerves of the pulp.
a
The outer layer of enamel is formed by secretion
from a thin layer of epithelial cells, the ameloblasts.
The enamel matrix is devoid of collagen but con-
tains two characteristic groups of proteins, the
amelogenins and the enamelins.
The amelogenins are hydrophobic but are also rich
in proline, histidine, and glutamic acid.
a,b
They
account for 90% of the matrix proteins but are
replaced by the initially less abundant enamelins
c
in fully mineralized teeth.
a,d
Another protein
present in developing enamel is ameloblastin,
which appears to be unique to amyloblasts.
a
As
mineralization of the enamel progresses the matrix
is lost and 98% of the enamel is hydroxylapatite.
a
Dentin is of epithelial-mesenchymal origin.
The odontoblasts secrete an extracellular matrix
that is rich in collagen I and which also contains all
of the other major bone proteins as well as a dentin
sialoprotein
e
and poorly characterized phosphate
carriers, the phosphophoryns.
The third hard tissue of teeth is cementum,
which binds teeth to the periodental ligaments. The
ligaments contain fibrillar collagen which inserts
into the cementum.
f,g
Except for the common cold, tooth decay (caries)
is the most prevalent disease in the United States.
h,i
Caries is initiated by attack of acids produced by
bacterial fermentations on the enamel. The saliva
contains calcium and phosphate and is supersatu-
rated with respect to these ions. As a result the
enamel surface is continuously recalcified. The 43-
residue salivary protein statherin retards precipita-
tion of calcium phosphates from the saliva,
preventing excessive build-up of calcified deposits
on the teeth.
j
Other proline-rich proteins may play a
role in recalcification repair.
k
A freshly cleaned tooth surface quickly becomes
coated with a thin pellicle of salivary proteins. This
provides a surface for growth of dental plaque,
which contains many bacteria and adhesive
polysaccharides such as dextrans.
l
The latter are
generated from dietary sucrose by such bacteria as
Streptococcus mutans. (Chapter 20) and others.
m
Many factors affect the probability of tooth decay.
A high sucrose diet promotes decay.
n
While most
people have some trouble with tooth decay, 1 or 2
per thousand remain totally free of caries and seem
to be immune. Many factors must affect resistance
to caries. For example, individuals vary in the kinds
and numbers of bacteria present on teeth and in the
structure of tooth enamel.
o
Addition of fluoride ion
to water supplies at a level of 1ppm (0.05 mM) is
generally believed to reduce the incidence of tooth
decay. However, caries has been declining in many
developed countries at rates that are the same for
water with or without fluoride.
p,q
If teeth escape
caries periodental disease, caused by bacteria, is
often a major problem for older people.
r
a
Krebsbach, P. H., Lee, S. K., Matsuki, Y., Kozak, C. A., Yamada,
K. M., and Yamada, Y. (1996) J. Biol. Chem. 271, 4431 4435
b
Renugopalakrishnan, V., Strawich, E. S., Horowitz, P. M., and
Glimcher, M. J. (1986) Biochemistry 25, 4879 4887
c
Deutsch, D., Palmon, A., Fisher, L. W., Kolodny, N., Termine,
J. D., and Young, M. F. (1991) J. Biol. Chem. 266, 16021 16028
d
Robinson, C., Weatherell, J. A., and Hhling, H. J. (1983) Trends
Biochem. Sci. 8, 284 287
e
Ritchie, H. H., Hou, H., Veis, A., and Butler, W. T. (1994) J. Biol.
Chem. 269, 3698 3702
f
Arzate, H., Olson, S. W., Page, R. C., Gown, A. M., and
Narayanan, A. S. (1992) FASEB J. 6, 2990 2995
g
Yamauchi, M., Katz, E. P., and Mechanic, G. L. (1986)
Biochemistry 25, 4907 4913
h
Sanders, H. J. (1980) Chem. Eng. News (Feb. 25) 58(8) 30 39
i
Shaw, J. H. (1987) N. Engl. J. Med. 317, 996 1004
j
Schlesinger, D. H., and Hoy, D. I. (1977) J. Biol. Chem. 252, 1689
1695
k
Clements, S., Mehansho, H., and Carlson, D. M. (1985) J. Biol.
Chem. 260, 13471 13477
l
Kolenbrander, P. E., Ganeshkumar, N., Cassels, F. J., and
Hughes, C. V. (1993) FASEB J. 7, 406 408
m
Abeygunawardana, C., and Bush, C. A. (1990) Biochemistry 29,
234248
n
Newbrun, E. (1982) Science 217, 418 423
o
Cevc, G., Cevc, P., Schara, M., and Skaleric, U. (1980) Nature
(London) 286, 425 426
p
Diesendorf, M. (1986) Nature (London) 322, 125 129
q
Hileman, B. (1988) Chem. Eng. News Aug. 1, 26 42
r
Williams, R. C. (1990) N. Engl. J. Med. 322, 373 382
Enamel
Pellicle
Plaque
Dentin
Gum
Pulp
Bone
Pulp canal
Cementum
Periodontal
membrane
Root
This cross section of a human molar
shows deposits of pellicle and plaque
near the gum line
h
Crown
443
nel into the interior of the bone. However, the tunnel
is soon lined with new osteoblasts and new bone is
laid down.
707
In osteoporosis, a very common disease
of older persons, the rate of resorption of bone by
osteoclasts exceeds that of bone formation by osteo-
blasts. Women typically lose 50% and men ~30% of
their calcium phosphate from vertebrae and ends of
long bones as they age.
708
In osteopetrosis (marble
bone disease) excessive calcification of bone and other
tissues occurs as a result of a deficiency of carbonic
anhydrase.
709
See also Chapter 13. A disease of pro-
gressive calcification of soft tissues
710
results from an
excess of bone morphogenic factor-4, one of a group
of protein factors acting on bone development (Chapter
32).
631
Pagets disease is another disorder of bone
remodeling that leads to overproduction of bone of
poor quality.
711
As discussed in Box 22-C, the circulating
level of Ca
2+
as well as the cellular uptake of this ion
are controlled by vitamin D and its metabolites and
by calcitonin and the parathyroid hormones.
Lack of a pyrophosphate ion pump in cartilage
cells may cause a deficit in pyrophosphate in the
surroundings of the forming bone. In mice with a
defect in this pump bony spurs, similar to those in
human osteoarthritis, are formed.
711a
Over half of the
worlds population of persons over 65 years of age are
afflicted by arthritis, over 100 types being known.
711b
Calcium carbonate in several different crystalline
forms arises biologically.
705,712
Sometimes the mineral
is deposited within vesicles inside the cell. Thus some
species of the protozoan Hymenomonas use Golgi vesicles
as sites for construction of segments of plantlike cell
walls complete with crystalline calcium carbonate
plates attached to the outer surface.
713
These wall
segments are then transported to their final locations.
The intricately sculptured spicules of sea urchins and
sponges are single crystals of calcium carbonate which
have grown within intracellular vesicles.
687,714
Diatoms
also form their shells of nearly pure, hydrated SiO
2
(See
Box 4-B) entirely within membrane-bound vesicles.
715
On the other hand, shells of molluscs are usually
deposited outside the cells of the organism but again
under the influence of a protein matrix.
716,717
The
animals apparently actively pump bicarbonate outward
where it reacts with Ca
2+
(Eq. 8-11).
718
Ca
2+
+ HCO
3

CaCO
3
+ H
+
The protons released then react with more bicarbonate
to form carbonic acid which is converted to CO
2
and
water (Eq. 8-12) by the enzyme carbonic anhydrase.
A problem that arises within organisms is avoiding
crystallization under supersaturating conditions.
For example, normal urine is supersaturated with
calcium oxalate. To prevent formation of renal calculi
(stones)
719
an inhibitory glycoprotein is present and
slows the formation and growth of crystals.
720
Under
some disease conditions calcium carbonate stones may
form in pancreatic ducts. A 17 kDa lectinlike glycopro-
tein called lithostatine has been proposed to inhibit
stone formation by binding to certain planes on CaCO
3
microcrystals just as antifreeze proteins (Box 4-D) inhibit
ice formation.
721
However, this proposed function for
lithostatine is doubtful.
722,723
Pathological deposits of
crystalline calcium pyrophosphate and basic calcium
phosphates are sometimes present in joints,
724
even in
Neanderthal skeletons.
725
3. Cell Walls of Fungi and Green Plants
The cell walls of yeasts and other fungi are made
up largely of glucans, chitin, and a mannan-protein
complex. Yeasts contain predominately glucans but
chitin is the major polysaccharide in many other fungi.
The most abundant glucan is a 1,3 linked polymer
with about 3% 1,6 linkages to the branches and a
molecular mass of ~ 240 kDa. In addition there is about
15% of a highly branched 1,6 linked glucan containing
1,3 linked branches. At the outer surface of the wall
are mannoproteins
726 728
which carry small serine- or
threonine-linked oligomannans as well as large highly
branched mannans linked to asparagine through the
usual N-acetylglucosamine-containing core structure.
Some of these highly branched mannan chains serve as
species-specific antigens.
727
Like those of the bacterial
and animal cell surfaces, the antigens vary in structure,
a fact with important medical implications. Curiously,
many fungi have surface fimbriae which are composed
of collagen, which is usually regarded as exclusively
an animal protein.
729
Cell walls of higher plants (Fig. 1-7, 4-14) are com-
posed largely of polysaccharides. They are discussed
briefly in Chapter 20.
E. The Extracellular Matrix and Cell Walls
H
2
CO
3
CO
2
+ H
2
O
Carbonic
anhydrase
(8-11)
(8-12)
444 Chapter 8. Lipids, Membranes, and Cell Coats
63. Quinn, P. J., and Cherry, R. J., eds. (1992)
Structural and Dynamic Properties of Lipids and
Membranes, Portland Press, London
64. Disalvo, E. A., and Simon, S. A., eds. (1995)
Permeability and Stability of Lipid Bilayers, CRC
Press, Boca Raton, Florida
65. Jacobson, K., Sheets, E. D., and Simson, R.
(1995) Science 268, 14411442
66. Pascher, I., Lundmark, M., Nyholm, P.-G., and
Sundell, S. (1992) Biochim. Biophys. Acta. 1113,
339373
67. Cameron, D. G., Gudgin, E. F., and Mantsch,
H. H. (1981) Biochemistry 20, 44964500
68. Zasadzinski, J. A. N., Helm, C. A., Longo, M.
L., Weisenhorn, A. L., Gould, S. A. C., and
Hansma, P. K. (1991) Biophys. J. 59, 755760
69. Wiener, M. C., and White, S. H. (1992) Biophys.
J. 61, 428433
70. Smith, D. P. E., Bryant, A., Quate, C. F., Rabe,
J. P., Gerber, C. H., and Swalen, J. D. (1987)
Proc. Natl. Acad. Sci. U.S.A. 84, 969972
71. Yang, J., Tamm, L. K., Tillack, T. W., and Shao,
Z. (1993) J. Mol. Biol. 229, 286290
72. Nagle, J. F., Zhang, R., Tristram-Nagle, S.,
Sun, W., Petrache, H. I., and Suter, R. M.
(1996) Biophys. J. 70, 14191431
73. Mueller, P., Rudin, D. O., Tien, H. T., and
Wescott, W. C. (1962) Nature (London) 194,
979980
74. Howard, K. P., and Prestegard, J. H. (1995)
J. Am. Chem. Soc. 117, 50315040
75. Ostro, M. J. (1987) Sci. Am. 256(Jan), 103 111
76. Philippot, J. R., and Schuber, F., eds. (1995)
Liposomes as Tools in Basic Research and
Industry, CRC Press, Boca Raton, Florida
77. Harwood, J. L. (1992) Trends Biochem. Sci. 17,
203204
77a. Schrder, M. (1980) Chem. Rev. 80, 187213
77b. Mohapatra, S. K., and Behrman, E. J. (1982) J.
Inorg. Biochem 16, 8589
77c. Deetz, J. S., and Behrman, E. J. (1981) Intl. J.
Peptide Prot. Res. 17, 495500
78. Norrby, P.-O., Becker, H., and Sharpless, K. B.
(1996) J. Am. Chem. Soc. 118, 3542
79. Montal, M., and Mueller, P. (1972) Proc. Natl.
Acad. Sci. U.S.A. 69, 35613566
80. Ladha, S., Mackie, A. R., Harvey, L. J., Clark,
D. C., Lea, E. J. A., Brullemans, M., and
Duclohier, H. (1996) Biophys. J. 71, 13641373
81. Jones, M. N., and Chapman, D. (1995)
Micelles, Monolayers, and Biomembranes, Wiley-
Liss, New York
81a. Knobler, C. M. (1990) Adv. Chem. Phys. 77,
397449
82. Bryce, M. R., and Petty, M. C. (1995) Nature
(London) 374, 771776
83. Zasadzinski, J. A., Viswanathan, R., Madsen,
L., Garnaes, J., and Schwartz, D. K. (1994)
Science 263, 17261733
84. Fettiplace, R., Andrews, D. M., and Haydon,
D. A. (1971) J. Membr. Biol. 5, 277296
85. White, S. W., and King, G. I. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 65326536
86. Snyder, R. G., Maroncelli, M., Qi, S. P., and
Strauss, H. L. (1981) Science 214, 188190
87. Di, L., and Small, D. M. (1995) Biochemistry 34,
1667216677
88. Tu, K., Tobias, D. J., Blasie, J. K., and Klein, M.
L. (1996) Biophys. J. 70, 595608
89. Katsaras, J., Raghunathan, V. A., Dufourc, E.
J., and Dufourcq, J. (1995) Biochemistry 34,
46844688
90. Snyder, R. G., Liang, G. L., Strauss, H. L., and
Mendelsohn, R. (1996) Biophys. J. 71, 3186
3198
91. Chen, S. C., Sturtevant, J. M., and Gaffney, B.
J. (1980) Proc. Natl. Acad. Sci. U.S.A. 77, 5060
5063
1. Christie, W. W. (1973) Lipid Analysis; Isolation,
Preparation, Identification and Structural
Analysis of Lipids, Pergamon, Oxford
2. Hitchcock, C., and Nichols, B. W. (1971) Plant
Lipid Biochemistry, Academic Press, New York
3. Kates, M. (1972) in Laboratory Techniques in
Biochemistry and Molecular Biology, Vol. 3, Part
II (Work, T. S., and Work, E., eds), North-
Holland Publ., Amsterdam
4. Weete, J. W. (1980) Lipid Biochemistry of Fungi
and Other Organisms, Plenum, New York
5. Gurr, M. I., and James, A. T. (1975) Lipid
Biochemistry, an Introduction, 2nd ed., Cornell
Univ. Press, Ithaca, New York
6. Porter, K. R., and Bonneville, M. A. (1973) Fine
Structure of Cells and Tissues, 4th ed., Lea &
Febiger, Philadelphia, Pennsylvania
7. Aveldao, M. I., and Sprecher, H. (1987) J.
Biol. Chem. 262, 11801186
8. Gunstone, F. D. (1967) An Introduction to the
Chemistry and Biochemistry of Fatty Acids and
Their Glycerides, 2nd ed., Chapman and Hall,
London (Chapter 6)
9. Steck, P. A., Schwartz, B. A., Rosendahl, M. S.,
and Gray, G. R. (1978) J. Biol. Chem. 253, 5625
5629
10. McNeil, M., Daffe, M., and Brennan, P. J.
(1991) J. Biol. Chem. 266, 1321713223
11. Qureshi, N., Takayama, K., and Schnoes, H.
K. (1980) J. Biol. Chem. 255, 182189
11a. Adlof, R. O., Duval, S., and Emken, E. A.
(2000) Lipids 35, 131 135
12. Cravatt, B. F., Prospero-Garcia, O., Siuzdak,
G., Gilula, N. B., Henriksen, S. J., Boger, D. L.,
and Lerner, R. A. (1995) Science 268, 15061509
13. Hummel, H. E., Gaston, L. K., Shorey, H. H.,
Kaae, R. S., Byrne, K. J., and Silverstein, R. M.
(1973) Science 181, 873874
14. Klun, J. A., Chapman, O. L., Mattes, K. C.,
Wojtkowski, P. W., Beroza, M., and Sonnet, P.
E. (1973) Science 181, 661663
15. Moolenaar, W. H. (1995) J. Biol. Chem. 270,
1294912952
16. Wells, W. W., and Eisenberg, F., Jr., eds. (1978)
Cyclitols and Phosphoinositides, Academic
Press, New York
17. DeBuch, H. (1978) Trends Biochem. Sci. 3, 4445
18. Chilton, F. H., and Connell, T. R. (1988) J. Biol.
Chem. 263, 52605265
19. Ford, D. A., and Gross, R. W. (1994)
Biochemistry 33, 12161222
20. Friedman, S. M., ed. (1978) Biochemistry of
Thermophily, Academic Press, New York
21. Kushwaha, S. C., Kates, M., Sprott, G. D., and
Smith, I. C. P. (1981) Science 211, 11631164
22. Luzzati, V., Gambacorta, A., DeRosa, M., and
Gulik, A. (1987) Ann. Rev. Biophys. Biophys.
Chem. 16, 2547
23. Goldfine, H., and Langworthy, T. A. (1988)
Trends Biochem. Sci. 13, 217221
24. Ferrante, G., Ekiel, I., and Sprott, G. D. (1986)
J. Biol. Chem. 261, 1706217066
25. Pond, J. L., Langworthy, T. A., and Holzer, G.
(1986) Science 231, 11341136
26. Florin-Christensen, J., Florin-Christensen, M.,
Knudson, J., and Rasmussen, L. (1986) Trends
Biochem. Sci. 11, 354355
27. Cooney, R. V., Mumma, R. O., and Benson, A.
A. (1978) Proc. Natl. Acad. Sci. U.S.A. 75, 4262
4264
28. Renou, J.-P., Giziewicz, J. B., Smith, I. C. P.,
and Jarrell, H. C. (1989) Biochemistry 28, 1804
1814
29. Gounaris, K., and Barber, J. (1983) Trends
Biochem. Sci. 8, 378381
30. Knowles, F. C., and Benson, A. A. (1983)
Trends Biochem. Sci. 8, 178180
References
31. Tupper, S., Wong, P. T. T., Kates, M., and
Tanphaichitr, N. (1994) Biochemistry 33,
1325013258
32. Andersson, A.-S., Rilfors, L., Bergqvist, M.,
Persson, S., and Lindblom, G. (1996)
Biochemistry 35, 1111911130
32a. Vikstrm, S., Li, L., and Wieslander, . (2000)
J. Biol. Chem. 275, 92969302
33. Nishihara, M., Utagawa, M., Akutsu, H., and
Koga, Y. (1992) J. Biol. Chem. 267, 1243212435
34. Powles, T. J., Easty, D. M., Easty, G. C., Bondy,
P. K., and Munro-Neville, A. (1973) Nature
(London), New Biol. 245, 83
35. Lynch, D. V. (1993) in Lipid Metabolism in
Plants (Moore, T. S., Jr., ed), pp. 285308, CRC
Press, Boca Raton, Florida
36. Araki, S., Satake, M., Ando, S., Hayashi, A.,
and Fujii, N. (1986) J. Biol. Chem. 261, 5138
5144
37. Araki, S., Abe, S., Odani, S., Ando, S., Fujii,
N., and Satake, M. (1987) J. Biol. Chem. 262,
1414114145
38. Song, W., and Rintoul, D. A. (1989)
Biochemistry 28, 41944200
39. Hsieh, T. C.-Y., Lester, R. L., and Laine, R. A.
(1981) J. Biol. Chem. 256, 77477755
40. Barr, K., and Lester, R. L. (1984) Biochemistry
23, 55815588
41. Dickson, R. C., Nagiec, E. E., Wells, G. B.,
Nagiec, M. M., and Lester, R. L. (1997) J. Biol.
Chem. 272, 2962029625
42. Yamakawa, T., and Nagai, Y. (1978) Trends
Biochem. Sci. 3, 128131
43. Hansson, G. C. (1983) J. Biol. Chem. 258, 9612
9615
44. Clausen, H., Holmes, E., and Hakomori, S.-I.
(1986) J. Biol. Chem. 261, 13881392
45. Farooqui, A. A. (1981) Adv. Lipid Res. 18, 159
202
46. Godchaux, W., III, and Leadbetter, E. R. (1984)
J. Biol. Chem. 259, 29822990
47. Abbanat, D. R., Leadbetter, E. R., Godchaux,
W., III, and Escher, A. (1986) Nature (London)
324, 367369
48. Jones, M. N., and Chapman, D., eds. (1994)
Micelles, Monolayers and Biomembranes, Wiley,
New York
49. Jain, M. (1988) Introduction to Biological
Membranes, 2nd ed., Wiley, New York
50. Lofan, R., and Nicolson, G. L. (1981) in
Advanced Cell Biology (Schwartz, L. M., and
Azar, M. M., eds), Van Nostrand Reinhold
Co., New York
51. Storch, J., and Kleinfeld, A. M. (1985) Trends
Biochem. Sci. 10, 418 421
52. Robertson, R. N. (1983) The Lively Membranes,
Cambridge Univ. Press, Cambridge
53. Starzak, M. (1984) The Physical Chemistry of
Membranes, Academic Press, San Diego,
California
54. Gennis, R. B. (1989) Biomembranes: Molecular
Structure and Function, Springer-Verlag, New
York
55. Morell, P., and Norton, W. T. (1980) Sci. Am.
242(May), 88116
56. Martenson, R. E., ed. (1992) Myelin: Biology
and Chemistry, CRC Press, Boca Raton, Florida
57. Weimbs, T., and Stoffel, W. (1994) Biochemistry
33, 1040810415
58. Korn, E. D. (1966) Science 153, 14911498
59. Tanford, C. (1980) The Hydrophobic Effect:
Formation of Micelles and Biological Membranes,
2nd ed., Wiley, New York
60. Danielli, J. F., and Davsen, H. (1985) J. Cell.
Comp. Physiol. 5, 495508
61. Singer, S. J., and Nicolson, G. L. (1972) Science
175, 720731
62. Bretscher, M. S., and Raff, M. C. (1975) Nature
(London) 258, 4349
445
References
92. Sun, W.-J., Tristram-Nagle, S., Suter, R. M.,
and Nagle, J. F. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 70087012
93. Mou, J., Yang, J., and Shao, Z. (1994)
Biochemistry 33, 44394443
94. Wang, Z.-q, Lin, H.-n, Li, S., and Huang, C.-h.
(1995) J. Biol. Chem. 270, 20142023
95. Wang, G., Lin, H.-n, Li, S., and Huang, C.-h.
(1995) J. Biol. Chem. 270, 2273822746
96. Wang, Z.-q, Lin, H.-n, Li, S., and Huang, C.-h.
(1994) J. Biol. Chem. 269, 2349123499
97. Dufourc, E. J., Parish, E. J., Chitrakorn, S., and
Smith, I. C. P. (1984) Biochemistry 23, 60626071
98. Maulik, P. R., and Shipley, G. G. (1996)
Biochemistry 35, 80258034
99. McMullen, T. P. W., and McElhaney, R. N.
(1997) Biochemistry 36, 49794986
100. Finegold, L., ed. (1993) Cholesterol in
Membrane Models, CRC Press, Boca Raton,
Florida
101. Recktenwald, D. J., and McConnell, H. M.
(1981) Biochemistry 20, 45054510
102. Needham, D., McIntosh, T. J., and Evans, E. E.
(1988) Biochemistry 27, 46684673
103. Goldfine, H., Johnston, N. C., and Phillips, M.
C. (1981) Biochemistry 20, 29082916
104. Lewis, R. V. A. H., Pohle, W., and McElhaney,
R. N. (1996) Biophys. J. 70, 27362746
105. Shimshick, E. J., and McConnell, H. M. (1973)
Biochemistry 12, 23512360
106. Almeida, P. F. F., Vaz, W. L. C., and Thompson,
T. E. (1992) Biochemistry 31, 71987210
107. Truble, H., and Eibl, H. (1974) Proc. Natl.
Acad. Sci. U.S.A. 71, 214219
108. Basnez, G., Nieva, J. L., Rivas, E., Alonso, A.,
and Goni, F. M. (1996) Biophys. J. 70, 22992306
109. Smith, R. L., and Oldfield, E. (1984) Science
225, 280288
109a. Smith, I. C. P. (1983) in NMR of Newly
Accessible Nuclei, Vol. 2 (Laslo, P., ed), pp. 126,
Academic Press, New York
110. Marassi, F. M., and Macdonald, P. M. (1992)
Biochemistry 31, 1003110036
111. Oldfield, E., Bowers, J. L., and Forbes, J.
(1987) Biochemistry 26, 69196923
112. Bruzik, K. S., and Nyholm, P.-G. (1997)
Biochemistry 36, 566575
113. Hong, M., Schmidt-Rohr, K., and Nanz, D.
(1995) Biophys. J. 69, 19391950
114. Thurmond, R. L., Niemi, A. R., Lindblom, G.,
Wieslander, ., and Rilfors, L. (1994)
Biochemistry 33, 1317813188
115. Boden, N., Jones, S. A., and Sixl, F. (1991)
Biochemistry 30, 21462155
116. Davis, J. H. (1993) in Cholesterol in Membrane
Models (Finegold, L., ed), pp. 67157, CRC
Press, Boca Raton, Florida
117. Baenziger, J. E., Jarrell, H. C., and Smith, I. C.
P. (1992) Biochemistry 31, 33773385
118. De Boeck, H., and Zidovetzki, R. (1992)
Biochemistry 31, 623630
119. de Kruijft, B., Cullis, P. R., and Verkleij, A. J.
(1980) Trends Biochem. Sci. 5, 7981
120. Peng, X., and Jonas, J. (1992) Biochemistry 31,
63836390
121. Hong, M., Schmidt-Rohr, K., and
Zimmermann, H. (1996) Biochemistry 35,
83358341
122. Lu, D., Singh, D., Morrow, M. R., and Grant,
C. W. M. (1993) Biochemistry 32, 290297
123. Salmon, A., and Hamilton, J. A. (1995)
Biochemistry 34, 1606516073
124. Ruocco, M. J., Siminovitch, D. J., Long, J. R.,
Das Gupta, S. K., and Griffin, R. G. (1996)
Biophys. J. 71, 17761788
125. Bruzik, K. S., and Harwood, J. S. (1997) J. Am.
Chem. Soc. 119, 66296637
126. Casal, H. L., and McElhaney, R. N. (1990)
Biochemistry 29, 54235427
127. Hbner, W., Mantsch, H. H., Paltauf, F., and
Hauser, H. (1994) Biochemistry 33, 320326
128. Lger, C. L., Daveloose, D., Christon, R., and
Viret, J. (1990) Biochemistry 29, 72697275
129. Jakobsson, E. (1997) Trends Biochem. Sci. 22,
339344
130. Berger, O., Edholm, O., and Jhnig, F. (1997)
Biophys. J. 72, 20022013
131. Chiu, S.-W., Clark, M., Balaji, V.,
Subramaniam, S., Scott, H. L., and Jakobsson,
E. (1995) Biophys. J. 69, 12301245
132. Browning, J. L. (1981) Biochemistry 20, 7144
7151
133. Cevc, G., Watts, A., and Marsh, D. (1981)
Biochemistry 20, 49554965
134. Harlos, K., and Eibl, H. (1980) Biochemistry 19,
895899
135. Koynova, R. D., Kuttenreich, H. L., Tenchov,
B. G., and Hinz, H.-J. (1988) Biochemistry 27,
46124619
136. Hayakawa, E., Naganuma, M., Mukasa, K.,
Shimozawa, T., and Araiso, T. (1998) Biophys.
J. 74, 892898
137. Katsaras, J., Jeffrey, K. R., Yang, D. S.-C., and
Epand, R. M. (1993) Biochemistry 32, 10700
10707
138. Srisiri, W., Sisson, T. M., OBrien, D. F.,
McGrath, K. M., Han, Y., and Gruner, S. M.
(1997) J. Am. Chem. Soc. 119, 48664873
139. Siegel, D. P., and Epand, R. M. (1997) Biophys.
J. 73, 30893111
140. Thurmond, R. L., Lindblom, G., and Brown,
M. F. (1993) Biochemistry 32, 53945410
141. Siegel, D. P., Banschbach, J., and Yeagle, P. L.
(1989) Biochemistry 28, 50105019
142. Siegel, D. P., and Banschbach, J. L. (1990)
Biochemistry 29, 59755981
143. Landau, E. M., and Rosenbusch, J. P. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1453214535
144. Delacroix, H., Gulik-Krzywicki, T., and
Seddon, J. M. (1996) J. Mol. Biol. 258, 88103
144a. Tenchov, B., Koynova, R., and Rapp, G. (1998)
Biophys. J. 75, 853866
145. Russell, N. J. (1984) Trends Biochem. Sci. 9,
108112
146. deMendoza, D., and Cronan, J. E., Jr. (1983)
Trends Biochem. Sci. 8, 4952
147. Mantsch, H. H., Madec, C., Lewis, R. N. A. H.,
and McElhaney, R. N. (1987) Biochemistry 26,
40454049
148. Bechtel, D. B., Mueller, D. D., Whaley, T. W.,
and Bulla, L. A., Jr. (1985) J. Biol. Chem. 260,
97849792
149. Brub, L. R., and Hollingsworth, R. I. (1995)
Biochemistry 34, 1200512011
150. Huster, D., Jin, A. J., Arnold, K., and
Gawrisch, K. (1997) Biophys. J. 73, 855864
151. Truble, H. (1971) J. Membr. Biol. 4, 193208
152. Wilson, M. A., and Pohorille, A. (1996) J. Am.
Chem. Soc. 118, 65806587
153. Ho, C., and Stubbs, C. D. (1997) Biochemistry
36, 1063010637
154. Bretscher, M. S. (1980) Trends Biochem. Sci. 5,
VI VII
155. Devaux, P., and McConnell, H. M. (1972) J.
Am. Chem. Soc. 94, 44754481
156. Kell, D. B. (1984) Trends Biochem. Sci. 9, 8688
157. Picard, F., Paquet, M.-J., Dufourc, E. J., and
Auger, M. (1998) Biophys. J. 74, 857868
158. Schmidt, T., Schtz, G. J., Baumgartner, W.,
Gruber, H. J., and Schindler, H. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 29262929
159. Sheets, E. D., Lee, G. M., Simson, R., and
Jacobson, K. (1997) Biochemistry 36, 12449
12458
160. Hopkins, C. R. (1992) Trends Biochem. Sci. 17,
2732
161. Kleinfeld, A. M., Chu, P., and Storch, J. (1997)
Biochemistry 36, 57025711
162. Zhao, J., Zhou, Q., Wiedmer, T., and Sims, P. J.
(1998) Biochemistry 37, 63616366
163. Wolff, D., Canessa-Fischer, M., Vargas, F., and
Diaz, G. (1971) J. Membr. Biol. 6, 304314
164. Sherbet, G. V. (1978) The Biophysical
Characterisation of the Cell Surface, Academic
Press, London
165. Jones, M. N. (1975) Biological Interfaces,
Elsevier, Amsterdam
166. McLaughlin, S. (1989) Ann. Rev. Biophys.
Biophys. Chem. 18, 113136
167. Aidley, D. J. (1971) The Physiology of Excitable
Cells, Cambridge Univ. Press, London and
New York
168. Nystrom, R. A. (1973) Membrane Physiology,
Prentice-Hall, Englewood Cliffs, New Jersey
169. Nicholls, D. G., and Ferguson, S. J. (1992)
Bioenergetics 2, Academic Press, London
170. Wood, P. M. (1985) Trends Biochem. Sci. 10,
106107
171. Teissi, J., Prats, M., LeMassu, A., Stewart, L.
C., and Kates, M. (1990) Biochemistry 29, 5965
172. Teissi, J., Gabriel, B., and Prats, M. (1993)
Trends Biochem. Sci. 18, 243246
173. Heberle, J., Riesle, J., Thiedemann, G.,
Oesterhelt, D., and Dencher, N. A. (1994)
Nature (London) 370, 379 382
174. Gabriel, B., and Teissi, J. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 1452114525
175. Devaux, P. F. (1991) Biochemistry 30, 11631173
176. Victorov, A. V., Janes, N., Taraschi, T. F., and
Hoek, J. B. (1997) Biophys. J. 72, 25882598
177. Shiffer, K. A., Rood, L., Emerson, R. K., and
Kuypers, F. A. (1998) Biochemistry 37, 3449
3458
178. Michel, H. (1983) Trends Biochem. Sci. 8, 5659
179. Cowan, S. W., and Rosenbusch, J. P. (1994)
Science 264, 914916
180. von Heijne, G. (1994) Annu. Rev. Biophys.
Biomol. Struct. 23, 167192
181. Bloom, M. (1995) Biophys. J. 69, 16311632
182. Weimbs, T., and Stoffel, W. (1992) Biochemistry
31, 1228912296
183. Yamaguchi, Y., Ikenaka, K., Niinobe, M.,
Yamada, H., and Mikoshiba, K. (1996) J. Biol.
Chem. 271, 2783827846
184. Jennings, M. L. (1989) Ann. Rev. Biochem. 58,
9991027
185. Bowie, J. U. (1997) J. Mol. Biol. 272, 780789
186. Gafvelin, G., Sakaguchi, M., Andersson, H.,
and von Heijne, G. (1997) J. Biol. Chem. 272,
61196127
187. Jhnig, F. (1990) Trends Biochem. Sci. 15, 9395
188. Jones, D. T., Taylor, W. R., and Thornton, J. M.
(1994) Biochemistry 33, 30383049
189. Persson, B., and Argos, P. (1996) Protein Sci. 5,
363371
190. Fasman, G. D., and Gilbert, W. A. (1990)
Trends Biochem. Sci. 15, 8992
191. Milpetz, F., Argos, P., and Persson, B. (1995)
Trends Biochem. Sci. 20, 204205
192. Shai, Y. (1995) Trends Biochem. Sci. 20, 460464
192a. Killian, J. A., and von Heijne, G. (2000) Trends
Biochem. Sci. 25, 429434
192b. Lew, S., Ren, J., and London, E. (2000)
Biochemistry 39, 96329640
193. Popov, M., Tam, L. Y., Li, J., and Reithmeier, R.
A. F. (1997) J. Biol. Chem. 272, 1832518332
194. Lee, G. F., Burrows, G. G., Lebert, M. R.,
Dutton, D. P., and Hazelbauer, G. L. (1994) J.
Biol. Chem. 269, 2992029927
195. Lee, G. F., Dutton, D. P., and Hazelbauer, G. L.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 5416
5420
196. Yu, H., Kono, M., McKee, T. D., and Oprian,
D. D. (1995) Biochemistry 34, 1496314969
197. Sansom, M. S. P., Son, H. S.,
Sankararamakrishnan, R., Kerr, I. D., and
Breed, J. (1995) Biophys. J. 68, 12951310
446 Chapter 8. Lipids, Membranes, and Cell Coats
265. Wolfe, L. C., John, K. M., Falcone, J. C., Byrne,
A. M., and Lux, S. E. (1982) N. Engl. J. Med.
307, 13671374
266. Diakowski, W., and Sikorski, A. F. (1995)
Biochemistry 34, 1325213258
267. Marfatia, S. M., Lue, R. A., Branton, D., and
Chishti, A. H. (1995) J. Biol. Chem. 270, 715719
268. Krauss, S. W., Chasis, J. A., Rogers, C.,
Mohandas, N., Krockmalnic, G., and Penman,
S. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 7297
7302
269. Khodadad, J. K., Waugh, R. E., Podolski, J. L.,
Josephs, R., and Steck, T. L. (1996) Biophys. J.
70, 10361044
270. Hansen, J. C., Skalak, R., Chien, S., and Hoger,
A. (1996) Biophys. J. 70, 146166
271. Rice-Evans, C. A., and Dunn, M. J. (1982)
Trends Biochem. Sci. 7, 282286
272. Mohandas, N., and Evans, E. (1994) Annu.
Rev. Biophys. Biomol. Struct. 23, 787818
273. Agre, P., Casella, J. F., Zinkham, W. H.,
McMillan, C., and Bennett, V. (1985) Nature
(London) 314, 380383
274. Baines, A. J., and Bennett, V. (1985) Nature
(London) 315, 410413
275. Clark, E. A., and Brugge, J. S. (1995) Science
268, 233239
276. Dejana, E., Corada, M., and Lampugnani, M.
G. (1995) FASEB J. 9, 910918
277. Malik, R. K., and Parsons, J. T. (1996) J. Biol.
Chem. 271, 2978529791
278. Lo, S. H., An, Q., Bao, S., Wong, W.-K., Liu, Y.,
Janmey, P. A., Hartwig, J. H., and Chen, L. B.
(1994) J. Biol. Chem. 269, 2231022319
279. Horwitz, A. F. (1997) Sci. Am. 276(May), 6875
280. de Pereda, J. M., Wiche, G., and Liddington,
R. C. (1999) EMBO J. 18, 40874095
281. Springer, T. A. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 6572
282. Bazzoni, G., and Hemler, M. E. (1998) Trends
Biochem. Sci. 23, 3034
283. Emsley, J., King, S. L., Bergelson, J. M., and
Liddington, R. C. (1997) J. Biol. Chem. 272,
2851228517
284. Luscinskas, F. W., and Lawler, J. (1994) FASEB
J. 8, 929938
285. Simon, K. O., Otey, C. A., Pavelko, F. M., and
Burridge, K. (1991) Curr. Top. Membr. Transp.
38, 5764
286. Edelman, G. M. (1983) Science 219, 450457
287. Becker, J. W., Erickson, H. P., Hoffman, S.,
Cunningham, B. A., and Edelman, G. M.
(1989) Proc. Natl. Acad. Sci. U.S.A. 86, 1088
1092
288. Cunningham, B. A., Hemperly, J. J., Murray,
B. A., Prediger, E. A., Brackenbury, R., and
Edelman, G. M. (1987) Science 236, 799806
289. Kudo, M., Kitajima, K., Inoue, S., Shiokawa,
K., Morris, H. R., Dell, A., and Inoue, Y. (1996)
J. Biol. Chem. 271, 3266732677
290. Newman, P. J., Berndt, M. C., Gorski, J.,
White, G. C., II, Lyman, S., Paddock, C., and
Muller, W. A. (1990) Science 247, 12191222
291. DeLisser, H. M., Yan, H. C., Newman, P. J.,
Muller, W. A., Buck, C. A., and Albelda, S. M.
(1993) J. Biol. Chem. 268, 1603716046
291a. Chen, H. A., Pfuhl, M., McAlister, M. S. B.,
and Driscoll, P. C. (2000) Biochemistry 39,
68146824
292. Casasnovas, J. M., Stehle, T., Liu, J.-h, Wang,
J.-h, and Springer, T. A. (1998) Proc. Natl. Acad.
Sci. U.S.A. 95, 41344139
293. Wang, J.-H., Pepinsky, R. B., Stehle, T., Liu, J.-
H., Karpusas, M., Browning, B., and Osborn,
L. (1995) Proc. Natl. Acad. Sci. U.S.A. 92,
57145718
294. Springer, T. A. (1990) Nature (London) 346,
425434
198. Stopar, D., Jansen, K. A. J., Pli, T., Marsh, D.,
and Hemminga, M. A. (1997) Biochemistry 36,
82618268
199. Nikaido, H. (1994) J. Biol. Chem. 269, 3905
3908
200. Mishra, V. K., and Palgunachari, M. N. (1996)
Biochemistry 35, 1121011220
201. Mishra, V. K., Palgunachari, M. N., Lund-
Katz, S., Phillips, M. C., Segrest, J. P., and
Anantharamaiah, G. M. (1995) J. Biol. Chem.
270, 16021611
202. Casey, P. J. (1995) Science 268, 221225
203. McIlhinney, R. A. J. (1990) Trends Biochem. Sci.
15, 387391
204. Buss, J. E., Mumby, S. M., Casey, P. J., Gilman,
A. G., and Sefton, B. M. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 74937497
205. Johnson, D. R., Bhatnagar, R. S., Knoll, L. J.,
and Gordon, J. I. (1994) Ann. Rev. Biochem. 63,
869914
206. Peseckis, S. M., and Resh, M. D. (1994) J. Biol.
Chem. 269, 3088830892
207. McLaughlin, S., and Aderem, A. (1995) Trends
Biochem. Sci. 20, 272276
208. Milligan, G., Parenti, M., and Magee, A. I.
(1995) Trends Biochem. Sci. 20, 181186
209. Wilson, P. T., and Bourne, H. R. (1995) J. Biol.
Chem. 270, 96679675
210. Duncan, J. A., and Gilman, A. G. (1996) J. Biol.
Chem. 271, 2359423600
211. Marshall, C. J. (1993) Science 259, 18651866
212. Glomset, J. A., Gelb, M. H., and Farnsworth,
C. C. (1990) Trends Biochem. Sci. 15, 139142
213. Casey, P. J., and Seabra, M. C. (1996) J. Biol.
Chem. 271, 52895292
214. Shipton, C. A., Parmryd, I., Swiezewska, E.,
Andersson, B., and Dallner, G. (1995) J. Biol.
Chem. 270, 566572
215. Wedegaertner, P. B., Wilson, P. T., and Bourne,
H. R. (1995) J. Biol. Chem. 270, 503506
216. Kikuchi, A., Sagami, H., and Ogura, K. (1999)
J. Biol. Chem. 274, 1801118016
217. Turner, A. J. (1994) in Essays in Biochemistry,
Vol. 28 (Tipton, K. F., ed), pp. 113128,
Portland Press, London
218. Low, M. G. (1987) Biochem. J. 244, 113
219. Lisanti, M. P., and Rodriguez-Boulan, E.
(1990) Trends Biochem. Sci. 15, 113118
220. Thomas, J. R., Dwek, R. A., and Rademacher,
T. W. (1990) Biochemistry 29, 54135422
221. Brewis, I. A., Ferguson, M. A. J., Mehlert, A.,
Turner, A. J., and Hooper, N. M. (1995) J. Biol.
Chem. 270, 2294622956
222. Previato, J. O., Jones, C., Xavier, M. T., Wait,
R., Travassos, L. R., Parodi, A. J., and
Mendona-Previato, L. (1995) J. Biol. Chem.
270, No 13, 72417250
223. Heinz, D. W., Ryan, M., Smith, M. P., Weaver,
L. H., Keana, J. F. W., and Griffith, O. H.
(1996) Biochemistry 35, 94969504
224. Williams, A. F., and Gagnon, J. (1982) Science
216, 696703
225. Mehlert, A., Richardson, J. M., and Ferguson,
M. A. J. (1998) J. Mol. Biol. 277, 379392
225a. Eisenhaber, B., Bork, P., Yuan, Y., Lffler, G.,
and Eisenhaber, F. (2000) Trends Biochem. Sci.
25, 340341
226. Reynolds, J. A., and Trayer, H. (1971) J. Biol.
Chem. 246, 73377342
227. Pryde, J. G. (1986) Trends Biochem. Sci. 11,
160163
228. Bennett, V. (1985) Ann. Rev. Biochem. 54, 273
304
229. Justice, J. M., Murtagh, J. J., Jr., Moss, J., and
Vaughan, M. (1995) J. Biol. Chem. 270, 17970
17976
230. Neville, D. M., Jr., and Glossmann, H. (1971)
J. Biol. Chem. 246, 63356338
References
231. Inaba, M., and Maede, Y. (1988) J. Biol. Chem.
263, 1776317771
232. Steck, T. L. (1974) J. Cell Biol. 62, 119
233. Luna, E. J., Kidd, G. H., and Branton, D.
(1979) J. Biol. Chem. 254, 25262532
234. Viitala, J., and Jrnefelt, J. (1985) Trends
Biochem. Sci. 10, 392395
235. Marchesi, V. T., Furthmayr, H., and Tomita,
M. (1976) Ann. Rev. Biochem. 45, 667698
236. Jay, D., and Cantley, L. (1986) Ann. Rev.
Biochem. 55, 511538
237. Murthy, S. N. P., Wilson, J., Zhang, Y., and
Lorand, L. (1994) J. Biol. Chem. 269, 22907
22911
238. Tanner, M. J. A. (1996) Nature (London) 382,
209210
239. Mller-Berger, S., Karbach, D., Knig, J.,
Lepke, S., Wood, P. G., Appelhans, H., and
Passow, H. (1995) Biochemistry 34, 93159324
240. Erickson, H. K. (1997) Biochemistry 36, 9958
9967
241. Fukuda, M., Lauffenburger, M., Sasaki, H.,
Rogers, M. E., and Dell, A. (1987) J. Biol. Chem.
262, 1195211957
242. MacKenzie, K. R., Prestegard, J. H., and
Engelman, D. M. (1997) Science 276, 131133
243. Challou, N., Goormaghtigh, E., Cabiaux, V.,
Conrath, K., and Ruysschaert, J.-M. (1994)
Biochemistry 33, 69026910
244. Huang, C.-H., Reid, M., Daniels, G., and
Blumenfeld, O. O. (1993) J. Biol. Chem. 268,
2590225908
245. Marchesi, V. T. (1985) Ann. Rev. Cell Biol. 1,
531561
246. Fujita, T., Ralston, G. B., and Morris, M. B.
(1998) Biochemistry 37, 264271
247. Yan, Y., Winograd, E., Viel, A., Cronin, T.,
Harrison, S. C., and Branton, D. (1993) Science
262, 20272030
248. Cherry, L., Menhart, N., and Fung, L. W.-M.
(1999) J. Biol. Chem. 274, 20772084
249. Begg, G. E., Harper, S. L., Morris, M. B., and
Speicher, D. W. (2000) J. Biol. Chem. 275, 3279
3287
250. Fujita, T., Ralston, G. B., and Morris, M. B.
(1998) Biochemistry 37, 272280
250a. Rief, M., Pascual, J., Saraste, M., and Gaub, H.
E. (1999) J. Mol. Biol. 286, 553561
251. Fowler, V. M. (1986) Nature (London) 322, 777
778
252. Carraway, K. L. (1975) Biochim. Biophys. Acta.
415, 379410
253. Byers, T. J., and Branton, D. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 61536157
254. Elgsaeter, A., Stokke, B. T., Mikkelsen, A., and
Branton, D. (1986) Science 234, 12171223
255. Pantazatos, D. P., and MacDonald, R. I. (1997)
J. Biol. Chem. 272, 2105221059
256. Luna, E. J., and Hitt, A. L. (1992) Science 258,
955964
257. Reichstein, E., and Blostein, R. (1975) J. Biol.
Chem. 250, 62566263
258. Bennett, V. (1992) J. Biol. Chem. 267, 8703
8706
259. Michaely, P., and Bennett, V. (1993) J. Biol.
Chem. 268, 2270322709
260. Kordeli, E., Lambert, S., and Bennett, V. (1995)
J. Biol. Chem. 270, 23522359
261. An, X.-L., Takakuwa, Y., Nunomura, W.,
Manno, S., and Mohandas, N. (1996) J. Biol.
Chem. 271, 3318733191
262. Workman, R. F., and Low, P. S. (1998) J. Biol.
Chem. 273, 61716176
263. Malik, S., Sami, M., and Watts, A. (1993)
Biochemistry 32, 1007810084
264. Azim, A. C., Knoll, J. H. M., Beggs, A. H., and
Chishti, A. H. (1995) J. Biol. Chem. 270, 17407
17413
447
References
295. Wagner, G. (1995) Science 267, 342
296. Nagar, B., Overduin, M., Ikura, M., and Rini,
J. M. (1996) Nature (London) 380, 360364
297. Shapiro, L., Fannon, A. M., Kwong, P. D.,
Thompson, A., Lehmann, M. S., Grbel, G.,
Legrand, J.-F., Als-Nielsen, J., Colman, D. R.,
and Hendrickson, W. A. (1995) Nature
(London) 374, 327336
298. Overduin, M., Harvey, T. S., Bagby, S., Tong,
K. I., Yau, P., Takeichi, M., and Ikura, M.
(1995) Science 267, 386389
298a. Nollet, F., Kools, P., and van Roy, F. (2000) J.
Mol. Biol. 299, 551572
298b. Pertz, O., Bozic, D., Koch, A. W., Fauser, C.,
Brancaccio, A., and Engel, J. (1999) EMBO J.
18, 17381747
299. Bella, J., Kolatkar, P. R., Marlor, C. W., Greve,
J. M., and Rossmann, M. G. (1998) Proc. Natl.
Acad. Sci. U.S.A. 95, 41404145
300. Patel, D. J., and Gumbiner, B. M. (1995) Nature
(London) 374, 306307
301. Berx, G., Cleton-Jansen, A.-M., Nollet, F., de
Leeuw, W. J. F., van de Vijver, M. J.,
Cornelisse, C., and van Roy, F. (1995) EMBO J.
14, 61076115
302. Vartio, T., and Vaheri, A. (1983) Trends
Biochem. Sci. 8, 442444
303. Hynes, R. O. (1986) Sci. Am. 254(Jun), 42 51
304. Ruoslahti, E. (1988) Ann. Rev. Biochem. 57,
375413
305. Engel, J., Odermatt, E., Engel, A., Madri, J. A.,
Furthmayr, H., Rohde, H., and Timpl, R.
(1981) J. Mol. Biol. 150, 97120
306. Ingham, K. C., Brew, S. A., Huff, S., and
Litvinovich, S. V. (1997) J. Biol. Chem. 272,
17181724
307. Johnson, K. J., Sage, H., Briscoe, G., and
Erickson, H. P. (1999) J. Biol. Chem. 274,
1547315479
308. Grant, R. P., Spitzfaden, C., Altroff, H.,
Campbell, I. D., and Mardon, H. J. (1997) J.
Biol. Chem. 272, 61596166
309. Hynes, R. O. (1990) Fibronectins, Springer-
Verlag, New York
310. Moyano, J. V., Carnemolla, B., Domnguez-
Jimnez, C., Garca-Gila, M., Albar, J. P.,
Snchez-Aparicio, P., Leprini, A., Querz, G.,
Zardi, L., and Garcia-Pardo, A. (1997) J. Biol.
Chem. 272, 2483224836
311. Greenberg, C. S., Birckbichler, P. J., and Rice,
R. H. (1991) FASEB J. 5, 30713077
312. Jeong, J.-M., Murthy, S. N. P., Radek, J. T., and
Lorand, L. (1995) J. Biol. Chem. 270, 56545658
313. An, S. S. A., Jimnez-Barbero, J., Petersen, T.
E., and Llins, M. (1992) Biochemistry 31,
99279933
314. Spitzfaden, C., Grant, R. P., Mardon, H. J., and
Campbell, L. D. (1997) J. Mol. Biol. 265, 565
579
315. Kornblihtt, A. R., Pesce, C. G., Alonso, C. R.,
Cramer, P., Srebrow, A., Werbajh, S., and
Muro, A. F. (1996) FASEB J. 10, 248257
316. Yoneda, A., Ogawa, H., Kojima, K., and
Matsumoto, I. (1998) Biochemistry 37, 6351
6360
317. Fischer, D., Chiquet-Ehrismann, R.,
Bernasconi, C., and Chiquet, M. (1995) J. Biol.
Chem. 270, 33783384
318. Yokosaki, Y., Matsuura, N., Higashiyama, S.,
Murakami, I., Obara, M., Yamakido, M.,
Shigeto, N., Chen, J., and Sheppard, D. (1998)
J. Biol. Chem. 273, 1142311428
319. Denda, S., Mller, U., Crossin, K. L., Erickson,
H. P., and Reichardt, L. F. (1998) Biochemistry
37, 54645474
320. Oberhauser, A. F., Marszalek, P. E., Erickson,
H. P., and Fernandez, J. M. (1998) Nature
(London) 393, 181185
321. Stetefeld, J., Mayer, U., Timpl, R., and Huber,
R. (1996) J. Mol. Biol. 257, 644657
322. Colognato, H., MacCarrick, M., ORear, J. J.,
and Yurchenco, P. D. (1997) J. Biol. Chem. 272,
2933029336
323. Lane, T. F., and Sage, E. H. (1994) FASEB J. 8,
163173
324. Venstrom, K. A., and Reichardt, L. F. (1993)
FASEB J. 7, 9961003
325. Gherzi, R., Carnemolla, B., Siri, A., Ponassi,
M., Balza, E., and Zardi, L. (1995) J. Biol. Chem.
270, 34293434
326. Ruoslahti, E., and Pierschbacher, M. D. (1987)
Science 238, 491497
327. DSouza, S. E., Ginsberg, M. H., and Plow, E.
F. (1991) Trends Biochem. Sci. 16, 246 250
328. Leahy, D. J., Hendrickson, W. A., Aukhil, I.,
and Erickson, H. P. (1992) Science 258, 987991
329. Stanley, K. K., Newby, A. C., and Luzio, J. P.
(1982) Trends Biochem. Sci. 7, 145147
330. Roberts, M. F. (1996) FASEB J. 10, 11591172
331. Han, S. K., Yoon, E. T., Scott, D. L., Sigler, P. B.,
and Cho, W. (1997) J. Biol. Chem. 272, 3573
3582
332. Caruthers, A., and Melchior, D. L. (1986)
Trends Biochem. Sci. 11, 331335
333. Gutknecht, J. (1987) Proc. Natl. Acad. Sci.
U.S.A. 84, 64436446
334. Paula, S., Volkov, A. G., and Deamer, D. W.
(1998) Biophys. J. 74, 319327
335. Stein, W. D. (1990) Channels, Carriers and
Pumps: an Introduction to Membrane Transport,
Academic Press, San Diego, California
335a. Paulsen, I. T., Nguyen, L., Sliwinski, M. K.,
Rabus, R., and Saier, M. H., Jr. (2000) J. Mol.
Biol. 301, 75100
336. Fox, R. O., Jr., and Richards, F. M. (1982)
Nature (London) 300, 325330
337. Kempf, C., Klausner, R. D., Weinstein, J. N.,
Van Renswoude, J., Pincus, M., and
Blumenthal, R. (1982) J. Biol. Chem. 257, 2469
2476
338. Klebba, P. E., Hofnung, M., and Charbit, A.
(1994) EMBO J. 13, 46704675
339. Schirmer, T., Keller, T. A., Wang, Y.-F., and
Rosenbusch, J. P. (1995) Science 267, 512514
340. Meyer, J. E. W., Hofnung, M., and Schulz, G.
E. (1997) J. Mol. Biol. 266, 761775
341. Jordy, M., Andersen, C., Schlein, K., Ferenci,
T., and Benz, R. (1996) J. Mol. Biol. 259, 666678
341a. Dumas, F., Koebnik, R., Winterhalter, M., and
Van Gelder, P. (2000) J. Biol. Chem. 275, 19747
19751
342. Samartzidou, H., and Delcour, A. H. (1998)
EMBO J. 17, 93100
343. Pauptit, R. A., Schirmer, T., Jansonius, J. N.,
Rosenbusch, J. P., Parker, M. W., Tucker, A. D.,
Tsernoglou, D., Weiss, M. S., and Schulz, G. E.
(1991) J. Structural Biol. 107, 136145
344. Kreusch, A., Neubser, A., Schiltz, E.,
Weckesser, J., and Schulz, G. E. (1994) Protein
Sci. 3, 5863
344a. Mller, D. J., and Engel, A. (1999) J. Mol. Biol.
285, 13471351
345. Bainbridge, G., Mobasheri, H., Armstrong, G.
A., Lea, E. J. A., and Lakey, J. H. (1998) J. Mol.
Biol. 275, 171176
346. Van Gelder, P., Saint, N., Phale, P., Eppens, E.
F., Prilipov, A., van Boxtel, R., Rosenbusch, J.
P., and Tommassen, J. (1997) J. Mol. Biol. 269,
468472
347. Lyer, R., Wu, Z., Woster, P. M., and Delcour, A.
H. (2000) J. Mol. Biol. 297, 933945
348. Watanabe, M., Rosenbusch, J., Schirmer, T.,
and Karplus, M. (1997) Biophys. J. 72, 20942102
349. Cowan, S. W., Schirmer, T., Rummel, G.,
Steiert, M., Ghosh, R., Pauptit, R. A.,
Jansonius, J. N., and Rosenbusch, J. P. (1992)
Nature (London) 358, 727733
350. Stathopoulos, C. (1996) Protein Sci. 5, 170173
351. Mannella, C. A. (1992) Trends Biochem. Sci. 17,
315320
352. Li, A. Z., Huang, H., Re, X., Qi, L. J., and
Marx, K. A. (1998) Biophys. J. 74, 964973
353. Reumann, S., Maier, E., Benz, R., and Heldt,
H. W. (1995) J. Biol. Chem. 270, 1755917565
354. Chrispeels, M. J., and Agre, P. (1994) Trends
Biochem. Sci. 19, 421425
355. Knepper, M. A. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 62556258
355a. Borgnia, M., Nielsen, S., Engel, A., and Agre,
P. (1999) Ann. Rev. Biochem. 68, 425458
356. Ringler, P., Borgnia, M. J., Stahlberg, H.,
Maloney, P. C., Agre, P., and Engel, A. (1999) J.
Mol. Biol. 291, 11811190
357. de Groot, B. L., Heymann, J. B., Engel, A.,
Mitsuoka, K., Fujiyoshi, Y., and Grubmller,
H. (2000) J. Mol. Biol. 300, 987994
358. Cheng, A., van Hoek, A. N., Yeager, M.,
Verkman, A. S., and Mitra, A. K. (1997) Nature
(London) 387, 627630
359. Ma, T., Song, Y., Gillespie, A., Carlson, E. J.,
Epstein, C. J., and Verkman, A. S. (1999) J.
Biol. Chem. 274, 2007120074
359a. Zeuthen, T., and Klaerke, D. A. (1999) J. Biol.
Chem. 274, 2163121636
360. Barone, L. M., Shih, C., and Wasserman, B. P.
(1997) J. Biol. Chem. 272, 3067230677
361. Calamita, G., Bishai, W. R., Preston, G. M.,
Guggino, W. B., and Agre, P. (1995) J. Biol.
Chem. 270, 2906329066
362. Deen, P. M. T., Verdijk, M. A. J., Knoers, N. V.
A. M., Wieringa, B., Monnens, L. A. H., van
OS, C. H., and van Oost, B. A. (1994) Science
264, 9295
363. Bai, L., Fushimi, K., Sasaki, S., and Marumo,
F. (1996) J. Biol. Chem. 271, 51715176
364. Jan, L. Y., and Jan, Y. N. (1994) Nature (London)
371, 119122
365. Armstrong, C. (1998) Science 280, 5657
366. Doyle, D. A., Cabral, J. M., Pfuetzner, R. A.,
Kuo, A., Gulbis, J. M., Cohen, S. L., Chait, B.
T., and MacKinnon, R. (1998) Science 280, 6977
367. Heginbothan, L., Odessey, E., and Miller, C.
(1997) Biochemistry 36, 1033510342
368. MacKinnon, R., Cohen, S. L., Kuo, A., Lee, A.,
and Chait, B. T. (1998) Science 280, 106109
369. Aidley, D. J., and Stanfield, P. R. (1996) Ion
Channels, Molecules in Action, Cambridge
Univ. Press, New York
370. Kreusch, A., Pfaffinger, P. J., Stevens, C. F.,
and Choe, S. (1998) Nature (London) 392, 945
948
370a. Gulbis, J. M., Zhou, M., Mann, S., and
MacKinnon, R. (2000) Science 289, 123127
370b. Kobertz, W. R., Williams, C., and Miller, C
(2000) Biochemistry 39, 1034710352
370c. Roux, B., Bernche, S., and Im, W. (2000)
Biochemistry 39,
371. Klingenberg, M. (1981) Nature (London) 290,
449454
372. Prince, R. C., Gunson, D. E., and Scarpa, A.
(1985) Trends Biochem. Sci. 10, 99
373. Smith, R., Separovic, F., Milne, T. J.,
Whittaker, A., Bennett, F. M., Cornell, B. A.,
and Makriyannis, A. (1994) J. Mol. Biol. 241,
456466
374. Dathe, M., Schmann, M., Wieprecht, T.,
Winkler, A., Beyermann, M., Krause, E.,
Matsuzaki, K., Murase, O., and Bienert, M.
(1996) Biochemistry 35, 1261212622
375. Kiyota, T., Lee, S., and Sugihara, G. (1996)
Biochemistry 35, 1319613204
376. Monette, M., and Lafleur, M. (1996) Biophys. J.
70, 21952202
377. Ojcius, D. M., and Young, J. D.-E. (1991)
Trends Biochem. Sci. 16, 225229
378. Langs, D. A. (1988) Science 241, 188191
448 Chapter 8. Lipids, Membranes, and Cell Coats
441. Brandsch, M., Thunecke, F., Kllertz, G.,
Schutkowski, M., Fischer, G., and Neubert, K.
(1998) J. Biol. Chem. 273, 38613864
442. Boorer, K. J., Frommer, W. B., Bush, D. R.,
Kreman, M., Loo, D. D. F., and Wright, E. M.
(1996) J. Biol. Chem. 271, 22132220
443. Boorer, K. J., and Fischer, W.-N. (1997) J. Biol.
Chem. 272, 1304013046
444. Someya, Y., Niwa, A., Sawai, T., and
Yamaguchi, A. (1995) Biochemistry 34, 712
445. Nikaido, K., Liu, P.-Q., and Ames, G. F.-L.
(1997) J. Biol. Chem. 272, 2774527752
446. Ltoff, S., Delepelaire, P., and Wandersman,
C. (1996) EMBO J. 15, 58045811
447. Blattner, F. R., Plunkett, G., III, Bloch, C. A.,
Perna, N. T., Burland, V., Riley, M., Collado-
Vides, J., Glasner, J. D., Rode, C. K., Mayhew,
G. F., Gregor, J., Davis, N. W., Kirkpatrick, H.
A., Goeden, M. A., Rose, D. J., Mau, B., and
Shao, Y. (1997) Science 277, 14531462
448. Decottignies, A., Lambert, L., Catty, P.,
Degand, H., Epping, E. A., Moye-Rowley, W.
S., Balzi, E., and Goffeau, A. (1995) J. Biol.
Chem. 270, 1815018157
448a. Driessen, A. J. M., Rosen, B. P., and Konings,
W. N. (2000) Trends Biochem. Sci. 25, 397401
449. Sharom, F. J., DiDiodato, G., Yu, X., and
Ashbourne, K. J. D. (1995) J. Biol. Chem. 270,
1033410341
450. Ko, Y. H., and Pedersen, P. L. (1995) J. Biol.
Chem. 270, 2209322096
451. Nossal, N. G., and Heppel, L. A. (1966) J. Biol.
Chem. 241, 30553062
452. Ames, G. F.-L. (1986) Ann. Rev. Biochem. 55,
397425
453. Ames, G. F.-L., and Higgins, C. F. (1983)
Trends Biochem. Sci. 8, 97100
454. Yao, N., Ledvina, P. S., Choudhary, A., and
Quiocho, F. A. (1996) Biochemistry 35, 2079
2085
455. Quiocho, F. A., and Vyas, N. K. (1984) Nature
(London) 310, 381386
456. Gilliland, G. L., and Quiocho, F. A. (1981) J.
Mol. Biol. 146, 341362
457. Vyas, N. K., Vyas, M. N., and Quiocho, F. A.
(1988) Science 242, 12901295
458. Vyas, N. K., Vyas, M. N., and Quiocho, F. A.
(1991) J. Biol. Chem. 266, 52265237
459. Shilton, B. H., Shuman, H. A., and Mowbray,
S. L. (1996) J. Mol. Biol. 264, 364376
460. Oh, B.-H., Kang, C.-H., De Bondt, H., Kim, S.-
H., Nikaido, K., Joshi, A. K., and Ames, G. F.-
L. (1994) J. Biol. Chem. 269, 41354143
461. Oh, B.-H., Ames, G. F.-L., and Kim, S.-H.
(1994) J. Biol. Chem. 269, 2632326330
462. Sugiyama, S., Vassylyev, D. G., Matsushima,
M., Kashiwagi, K., Igarashi, K., and
Morikawa, K. (1996) J. Biol. Chem. 271, 9519
9525
463. Pflugrath, J. W., and Quiocho, F. A. (1988)
J. Mol. Biol. 200, 163180
464. Manson, M. D., Boos, W., Bassford, P. J., Jr.,
and Rasmussen, B. A. (1985) J. Biol. Chem. 260,
97279733
465. Reizer, J., Michotey, V., Reizer, A., and Saier,
M. H., Jr. (1994) Protein Sci. 3, 440450
466. Garrett, D. S., Seok, Y.-J., Liao, D.-I.,
Peterkofsky, A., Gronenborn, A. M., and
Clore, G. M. (1997) Biochemistry 36, 25172530
467. Chauvin, F., Brand, L., and Roseman, S. (1994)
J. Biol. Chem. 269, 2026320269
467a. Zhu, P.-P., Szczepanowski, R. H., Nosworthy,
N. J., Ginsburg, A., and Peterkofsky, A. (1999)
Biochemistry 38, 1547015479
468. Jones, B. E., Rajagopal, P., and Klevit, R. E.
(1997) Protein Sci. 6, 21072119
469. El-Kabbani, O. A. L., Waygood, E. B., and
Delbaera, L. T. J. (1987) J. Biol. Chem. 262,
1292612929
379. Doyle, D. A., and Wallace, B. A. (1997) J. Mol.
Biol. 266, 963977
380. Iqbal, M., and Balaram, P. (1981) Biochemistry
20, 72787284
381. Malashkevich, V. N., Kammerer, R. A.,
Efimov, V. P., Schulthess, T., and Engel, J.
(1996) Science 274, 761765
382. Unwin, N. (1995) Nature (London) 373, 3743
383. Harrold, F. M., and Baarda, J. R. (1967) J.
Bacteriol. 94, 5360
384. Harold, F. M., and Van Brunt, J. (1977) Science
197, 372373
385. Pressman, B. C. (1976) Ann. Rev. Biochem. 45,
501530
386. Hamilton, J. A., Sabesan, M. N., and Steinrauf,
L. K. (1981) J. Am. Chem. Soc. 103, 58805885
387. Duax, W. L., Hauptman, M., Weeks, C. M.,
and Norton, D. A. (1972) Science 176, 911914
388. Karle, I. L., Karle, J., Wieland, T.,
Burgemeister, W., Faulstich, H., and Witkop,
B. (1973) Proc. Natl. Acad. Sci. U.S.A. 70, 1836
1840
389. Pangborn, W., Duax, W., and Langs, D. (1987)
J. Am. Chem. Soc. 109, 21632165
390. Karle, I. L. (1985) Proc. Natl. Acad. Sci. U.S.A.
82, 71557159
391. Inabayashi, M., Miyauchi, S., Kamo, N., and
Jin, T. (1995) Biochemistry 34, 34553460
392. Ledger, P. W., and Tanzer, M. L. (1984) Trends
Biochem. Sci. 9, 313314
393. Marger, M. D., and Saier, M. H., Jr. (1993)
Trends Biochem. Sci. 18, 1320
394. Goswitz, V. C., and Brooker, R. J. (1995)
Protein Sci. 4, 534537
395. Seidel, H. M., and Knowles, J. R. (1994)
Biochemistry 33, 56415646
396. Cloherty, E. K., Sulzman, L. A., Zottola, R. J.,
and Carruthers, A. (1995) Biochemistry 34,
1539515406
397. Shanahan, M. F., and DArtel-Ellis, J. (1984) J.
Biol. Chem. 259, 1387813884
398. Bell, G. I., Burant, C. F., Takeda, J., and Gould,
G. W. (1993) J. Biol. Chem. 268, 1916119164
399. Silverman, M. (1991) Ann. Rev. Biochem. 60,
757794
400. Mueckler, M. (1994) Eur. J. Biochem. 219, 713
725
401. Maher, F., Vannucci, S. J., and Simpson, I. A.
(1994) FASEB J. 8, 10031011
402. Burant, C. F., Takeda, J., Brot-Laroche, E., Bell,
G. I., and Davidson, N. O. (1992) J. Biol. Chem.
267, 1452314526
403. Zamora-Len, S. P., Golde, D. W., Concha, I.
I., Rivas, C. I., Delgado-Lpez, F., Baselga, J.,
Nualart, F., and Vera, J. C. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 18471852
404. Coderre, L., Kandror, K. V., Vallega, G., and
Pilch, P. F. (1995) J. Biol. Chem. 270, 27584
27588
405. Fischer, Y., Thomas, J., Sevilla, L., Munoz, P.,
Becker, C., Holman, G., Kozka, I. J., Palacn,
M., Testar, X., Kammermeier, H., and Zorzano,
A. (1997) J. Biol. Chem. 272, 70857092
405a. Shepherd, P. R., and Kahn, B. B. (1999) N.
Engl. J. Med. 341, 248257
406. Mueckler, M., and Makepeace, C. (1997) J.
Biol. Chem. 272, 3014130146
407. Cloherty, E. K., Diamond, D. L., Heard, K. S.,
and Carruthers, A. (1996) Biochemistry 35,
1323113239
408. Seatter, M. J., De La Rue, S. A., Porter, L. M.,
and Gould, G. W. (1998) Biochemistry 37,
13221326
409. Rumsey, S. C., Kwon, O., Xu, G. W., Burant, C.
F., Simpson, I., and Levine, M. (1997) J. Biol.
Chem. 272, 1898218989
410. Wiese, T. J., Dunlap, J. A., and Yorek, M. A.
(1994) J. Biol. Chem. 269, 2270522711
References
411. Kasahara, M., Shimoda, E., and Maeda, M.
(1997) J. Biol. Chem. 272, 1672116724
412. Turk, E., Kerner, C. J., Lostao, M. P., and
Wright, E. M. (1996) J. Biol. Chem. 271, 1925
1934
413. Chen, X.-Z., Coady, M. J., Jackson, F.,
Berteloot, A., and Lapointe, J.-Y. (1995)
Biophys. J. 69, 24052414
414. Panayotova-Heiermann, M., Eskandari, S.,
Turk, E., Zampighi, G. A., and Wright, E. M.
(1997) J. Biol. Chem. 272, 2032420327
415. Prasad, P. D., Wang, H., Kekuda, R., Fujita, T.,
Fei, Y.-J., Devoe, L. D., Leibach, F. H., and
Ganapathy, V. (1998) J. Biol. Chem. 273, 7501
7506
416. Malandro, M. S., and Kilberg, M. S. (1996)
Ann. Rev. Biochem. 65, 305336
417. Utsunomiya-Tate, N., Endou, H., and Kanai,
Y. (1996) J. Biol. Chem. 271, 1488314890
418. Kong, C.-T., Yet, S.-F., and Lever, J. E. (1993)
J. Biol. Chem. 268, 15091512
419. Su, T.-Z., Wang, M., Syu, L.-J., Saltiel, A. R.,
and Oxender, D. L. (1998) J. Biol. Chem. 273,
31733179
420. Yao, D., Mackenzie, B., Ming, H., Varoqui, H.,
Zhu, H., Hediger, M. A., and Erickson, J. D.
(2000) J. Biol. Chem. 275, 2279022797
420a. Reimer, R. J., Chaudhry, F. A., Gray, A. T., and
Edwards, R. H. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 77157720
421. Mosckovitz, R., Udenfriend, S., Felix, A., Heimer,
E., and Tate, S. S. (1994) FASEB J. 8, 10691074
422. Chillarn, J., Estvez, R., Mora, C., Wagner, C.
A., Suessbrich, H., Lang, F., Gelp, J. L., Testar,
X., Busch, A. E., Zorzano, A., and Palacn, M.
(1996) J. Biol. Chem. 271, 1776117770
423. Trotti, D., Rossi, D., Gjesdal, O., Levy, L. M.,
Racagni, G., Danbolt, N. C., and Volterra, A.
(1996) J. Biol. Chem. 271, 59765979
424. Peghini, P., Janzen, J., and Stoffel, W. (1997)
EMBO J. 16, 38223832
425. Lebrun, B., Sakaitani, M., Shimamoto, K.,
Yasuda-Kamatani, Y., and Nakajima, T. (1997)
J. Biol. Chem. 272, 2033620339
426. Ponce, J., Biton, B., Benavides, J., Avenet, P.,
and Aragn, C. (2000) J. Biol. Chem. 275,
1385613862
427. Pantanowitz, S., Bendahan, A., and Kanner, B.
I. (1993) J. Biol. Chem. 268, 32223225
428. Bennett, E. R., and Kanner, B. I. (1997) J. Biol.
Chem. 272, 12031210
429. Velaz-Faircloth, M., Guadano-Ferraz, A.,
Henzi, V. A., and Fremeau, R. T., Jr. (1995) J.
Biol. Chem. 270, 1575515761
430. Chen, J.-G., Liu-Chen, S., and Rudnick, G.
(1998) J. Biol. Chem. 273, 1267512681
431. Nikaido, H., and Saier, M. H., Jr. (1992) Science
258, 936942
432. Will, A., Grassl, R., Erdmenger, J., Caspari, T.,
and Tanner, W. (1998) J. Biol. Chem. 273,
1145611462
433. Kaback, H. R., Bibi, E., and Roepe, P. D. (1990)
Trends Biochem. Sci. 15, 309314
434. Frillingos, S., Wu, J., Venkatesan, P., and Kaback,
H. R. (1997) Biochemistry 36, 64086414
435. Green, A. L., Anderson, E. J., and Brooker, R.
J. (2000) J. Biol. Chem. 275, 2324023246
436. Zhao, M., Zen, K.-C., Hubbell, W. L., and Kaback,
H. R. (1999) Biochemistry 38, 74077412
437. Zani, M. L., Pourcher, T., and Leblanc, G.
(1994) J. Biol. Chem. 269, 2488324889
438. Pourcher, T., Bibi, E., Kaback, H. R., and
Leblanc, G. (1996) Biochemistry 35, 41614168
439. McDonald, T. P., Walmsley, A. R., and
Henderson, P. J. F. (1997) J. Biol. Chem. 272,
1518915199
440. Fei, Y.-J., Liu, W., Prasad, P. D., Kekuda, R.,
Oblak, T. G., Ganapathy, V., and Leibach, F. H.
(1997) Biochemistry 36, 452460
449
References
470. Reizer, J., Hoischen, C., Reizer, A., Pham, T.
N., and Saier, M. H., Jr. (1993) Protein Sci. 2,
506521
471. Eberstadt, M., Grdadolnik, S. G., Gemmecker,
G., Kessler, H., Buhr, A., and Erni, B. (1996)
Biochemistry 35, 1128611292
472. Van Dijk, A. A., Scheek, R. M., Dijkstra, K.,
Wolters, G. K., and Robillard, G. T. (1992)
Biochemistry 31, 90639072
473. Gemmecker, G., Eberstadt, M., Buhr, A., Lanz,
R., Golic Grdadolnik, S., Kessler, H., and Erni,
B. (1997) Biochemistry 36, 74087417
474. Pelton, J. G., Torchia, D. A., Remington, S. J.,
Murphy, K. P., Meadow, N. D., and Roseman,
S. (1996) J. Biol. Chem. 271, 3344633456
475. Lanz, R., and Erni, B. (1998) J. Biol. Chem. 273,
1223912243
476. Nunn, R. S., Markovic-Housley, Z.,
Gnovsio-Taverne, J.-C., Flkiger, K.,
Rizkallah, P. J., Jansonius, J. N., Schirmer, T.,
and Erni, B. (1996) J. Mol. Biol. 259, 502511
477. Roseman, S., and Meadow, N. D. (1990) J. Biol.
Chem. 265, 29932996
478. Hurley, J. H., Faber, H. R., Worthylake, D.,
Meadow, N. D., Roseman, S., Pettigrew, D. W.,
and Remington, S. J. (1993) Science 259, 673677
479. Cantor, C. R., and Schimmel, P. R. (1980)
Biophysical Chemistry, Freeman, San Francisco,
California
480. Masuda, T., Dobson, G. P., and Veech, R. L.
(1990) J. Biol. Chem. 265, 2032120334
481. Catterall, W. A. (1995) Ann. Rev. Biochem. 64,
493531
482. Valverde, M. A., Hardy, S. P., and Seplveda,
F. V. (1995) FASEB J. 9, 509515
483. Schmidt-Rose, T., and Jentsch, T. J. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 76337638
484. Jentsch, T. J., Steinmeyer, K., and Schwarz, G.
(1990) Nature (London) 348, 510514
485. Middleton, R. E., Pheasant, D. J., and Miller,
C. (1994) Biochemistry 33, 1318913198
486. Schwappach, B., Stobrawa, S., Hechenberger,
M., Steinmeyer, K., and Jentsch, T. J. (1998) J.
Biol. Chem. 273, 1511015118
487. Gronemeier, M., Condie, A., Prosser, J.,
Steinmeyer, K., Jentsch, T. J., and Jockusch, H.
(1994) J. Biol. Chem. 269, 59635967
488. Pucat, M., Korichneva, I., Cassoly, R., and
Vassort, G. (1995) J. Biol. Chem. 270, 13151322
489. Jarolim, P., Shayakul, C., Prabakaran, D.,
Jiang, L., Stuart-Tilley, A., Rubin, H. L.,
Simova, S., Zavadil, J., Herrin, J. T.,
Brouillette, J., Somers, M. J. G., Seemanova,
E., Brugnara, C., Guay-Woodford, L. M., and
Alper, S. L. (1998) J. Biol. Chem. 273, 63806388
489a. Fujinaga, J., Tang, X.-B., and Casey, J. R. (1999)
J. Biol. Chem. 274, 66266633
490. Welsh, M. J., and Smith, A. E. (1995) Sci. Am.
273(Dec), 5259
491. Seibert, F. S., Linsdell, P., Loo, T. W.,
Hanrahan, J. W., Riordan,
J. R., and Clarke, D. M. (1996) J. Biol. Chem.
271, 2749327499
492. Romero, M. F., Hediger, M. A., Boulpaep, E.
L., and Boron, W. F. (1997) Nature (London)
387, 409413
493. Burnham, C. E., Amlal, H., Wang, Z., Shull, G.
E., and Soleimani, M. (1997) J. Biol. Chem. 272,
1911119114
494. Muchhal, U. S., Pardo, J. M., and
Raghothama, K. G. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1051910523
495. Smith, F. W., Ealing, P. M., Hawkesford, M. J.,
and Clarkson, D. T. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 93739377
496. Zhou, J.-J., Theodoulou, F. L., Muldin, I.,
Ingemarsson, B., and Miller, A. J. (1998) J. Biol.
Chem. 273, 1201712023
497. Schroers, A., Burkovski, A., Wohlrab, H., and
Krmer, R. (1998) J. Biol. Chem. 273, 14269
14276
498. Phelps, A., Briggs, C., Mincone, L., and Wohlrab,
H. (1996) Biochemistry 35, 1075710762
499. Collins, J. F., and Ghishan, F. K. (1996) FASEB
J. 10, 751759
500. Garcia, C. K., Brown, M. S., Pathak, R. K., and
Goldstein, J. L. (1995) J. Biol. Chem. 270, 1843
1849
501. Kuo, S.-M., and Aronson, P. S. (1996) J. Biol.
Chem. 271, 1549115497
502. Yellen, G. (1987) Ann. Rev. Biophys. Biophys.
Chem. 16, 227246
503. Miller, C. (1991) Science 252, 10921096
504. Philipson, L. H., and Steiner, D. F. (1995)
Science 268, 372373
505. Inagaki, N., Gonoi, T., Clement, J. P., IV,
Namba, N., Inazawa, J., Gonzalez, G.,
Aguilar-Bryan, L., Seino, S., and Bryan, J.
(1995) Science 270, 11661170
506. Brnstrm, R., Efendic, S., Berggren, P.-O.,
and Larsson, O. (1998) J. Biol. Chem. 273,
1411314118
507. Ruknudin, A., Schulze, D. H., Sullivan, S. K.,
Lederer, W. J., and Welling, P. A. (1998) J. Biol.
Chem. 273, 1416514171
508. Day, M. L., Johnson, M. H., and Cook, D. I.
(1998) EMBO J. 17, 19521960
509. McCormick, K. A., Isom, L. L., Ragsdale, D.,
Smith, D., Scheuer, T., and Catterall, W. A.
(1998) J. Biol. Chem. 273, 39543962
510. Favre, I., Moczydlowski, E., and Schild, L.
(1996) Biophys. J. 71, 31103125
510a. Lipkind, G. M., and Fozzard, H. A. (2000)
Biochemistry 39, 81618170
511. Prez-Garca, M. T., Chiamvimonvat, N.,
Marban, E., and Tomaselli, G. F. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 300304
512. Kosari, F., Sheng, S., Li, J., Mak, D.-O. D.,
Foskett, J. K., and Kleyman, T. R. (1998) J. Biol.
Chem. 273, 1346913474
513. Garty, H. (1994) FASEB J. 8, 522528
514. Miller, R. J. (1992) J. Biol. Chem. 267, 14031406
515. Mitterdorfer, J., Sinnegger, M. J., Grabner, M.,
Striessnig, J., and Glossmann, H. (1995)
Biochemistry 34, 93509355
516. McPherson, P. S., and Campbell, K. P. (1993) J.
Biol. Chem. 268, 1376513768
517. Jeyakumar, L. H., Copello, J. A., OMalley, A.
M., Wu, G.-M., Grassucci, R., Wagenknecht,
T., and Fleischer, S. (1998) J. Biol. Chem. 273,
1601116020
518. Kraus, R. L., Sinnegger, M. J., Glossmann, H.,
Hering, S., and Striessnig, J. (1998) J. Biol.
Chem. 273, 55865590
519. Kim, S., Lakhani, V., Costa, D. J., Sharara, A.
I., Fitz, J. G., Huang, L.-W., Peters, K. G., and
Kindman, L. A. (1995) J. Biol. Chem. 270,
52665269
520. Mezna, M., and Michelangeli, F. (1996) J. Biol.
Chem. 271, 3181831823
521. Nicoll, D. A., Hryshko, L. V., Matsuoka, S.,
Frank, J. S., and Philipson, K. D. (1996) J. Biol.
Chem. 271, 1338513391
522. Crumpton, M. J., and Dedman, J. R. (1990)
Nature (London) 345, 212
523. Swairjo, M. A., and Seaton, B. A. (1994) Annu.
Rev. Biophys. Biomol. Struct. 23, 193213
524. Benz, J., Bergner, A., Hofmann, A., Demange,
P., Gttig, P., Liemann, S., Huber, R., and
Voges, D. (1996) J. Mol. Biol. 260, 638643
525. Burger, A., Berendes, R., Liemann, S., Benz, J.,
Hofmann, A., Gttig, P., Huber, R., Gerke, V.,
Thiel, C., Rmisch, J., and Weber, K. (1996) J.
Mol. Biol. 257, 839847
526. Demange, P., Voges, D., Benz, J., Liemann, S.,
Gttig, P., Berendes, R., Burger, A., and Huber,
R. (1994) Trends Biochem. Sci. 19, 272276
527. Campos, B., Mo, Y. D., Mealy, T. R., Li, C. W.,
Swairjo, M. A., Balch, C., Head, J. F.,
Retzinger, G., Dedman, J. R., and Seaton, B. A.
(1998) Biochemistry 37, 80048010
528. Ling, G. N. (1984) In Search of the Physical Basis
of Life, Plenum, New York
529. Baker, P. F., and Willis, J. S. (1970) Nature
(London) 226, 521523
530. Kyte, J. (1972) J. Biol. Chem. 247, 76427649
531. Lingrel, J. B., and Kuntzweiler, T. (1994) J.
Biol. Chem. 269, 1965919662
532. Lutsenko, S., Daoud, S., and Kaplan, J. H.
(1997) J. Biol. Chem. 272, 52495255
533. Scheiner-Bobis, G., and Schreiber, S. (1999)
Biochemistry 38, 91989208
534. Gatto, C., Wang, A. X., and Kaplan, J. H.
(1998) J. Biol. Chem. 273, 1057810585
535. Rossier, B. C., Geering, K., and Kraehenbuhl,
J. P. (1987) Trends Biochem. Sci. 12, 483487
536. Minor, N. T., Sha, Q., Nichols, C. G., and
Mercer, R. W. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 65216525
537. Lutsenko, S., and Kaplan, J. H. (1995)
Biochemistry 34, 1560715613
538. Nelson, N., and Taiz, L. (1989) Trends Biochem.
Sci. 14, 113116
539. Solioz, M., and Vulpe, C. (1996) Trends
Biochem. Sci. 21, 237241
539a. Ueno, S., Kaieda, N., Koyama, N. (2000) J.
Biol. Chem. 275, 1453714540
540. Carafoli, E. (1994) FASEB J. 8, 9931002
541. MacLennan, D. H., Rice, W. J., and Green, N.
M. (1997) J. Biol. Chem. 272, 2881528818
542. Myung, J., and Jencks, W. P. (1995) Biochemis-
try 34, 30773083
543. Dode, L., De Greef, C., Mountian, I., Attard,
M., Town, M. M., Casteels, R., and Wuytack,
F. (1998) J. Biol. Chem. 273, 1398213994
543a. Menguy, T., Corre, F., Bouneau, L., Des-
champs, S., Vuust Moller, J., Champeil, P., le
Maire, M., and Falson, P. (1998) J. Biol. Chem.
273, 2013420143
544. Melle-Milovanovic, D., Milovanovic, M.,
Nagpal, S., Sachs, G., and Shin, J. M. (1998) J.
Biol. Chem. 273, 1107511081
545. Rulli, S. J., Horiba, M. N., Skripnikova, E., and
Rabon, E. C. (1999) J. Biol. Chem. 274, 15245
15250
546. Asano, S., Inoie, M., and Takeguchi, N. (1987)
J. Biol. Chem. 262, 1326313268
547. Seto-Young, D., Hall, M. J., Na, S., Haber, J. E.,
and Perlin, D. S. (1996) J. Biol. Chem. 271, 581
587
548. Auer, M., Scarborough, G. A., and Khlbrandt,
W. (1998) Nature (London) 392, 840843
549. Orlowski, J., and Grinstein, S. (1997) J.
Biol. Chem. 272, 2237322376
549. Orlowski, J. and Grinstein, S. (1997) J. Biol.
Chem. 272, 2237322376
549a. Wakabayashi, S., Pang, T., Su, X., and
Shigekawa, M. (2000) J. Biol. Chem. 275, 7942
7949
549b. Williams, K. A. (2000) Nature (London) 403,
112115
550. Kasho, V. N., Stengelin, M., Smirnova, I. N.,
and Faller, L. D. (1997) Biochemistry 36, 8045
8052
551. Campos, M., and Beaug, L. (1997)
Biochemistry 36, 1422814237
552. Vilsen, B. (1997) Biochemistry 36, 1331213324
552a. Boxenbaum, N., Daly, S. E., Javaid, Z. Z.,
Lane, L. K., and Blostein, R. (1998) J. Biol.
Chem. 273, 2308623092
553. Zhang, P., Toyoshima, C., Yonekura, K.,
Green, N. M., and Stokes, D. L. (1998) Nature
(London) 392, 835839
553a. Toyoshima, C., Nakasako, M., Nomura, H.,
and Ogawa, H. (2000) Nature (London) 405,
647655
450 Chapter 8. Lipids, Membranes, and Cell Coats
603. Venisse, A., Rivire, M., Vercauteren, J., and
Puzo, G. (1995) J. Biol. Chem. 270, 1501215021
604. Brennan, P. J., and Nikaido, H. (1995) Ann.
Rev. Biochem. 64, 2963
605. Liu, J., Barry, C. E., III, Besra, G. S., and
Nikaido, H. (1996) J. Biol. Chem. 271, 29545
29551
606. Mikusov, K., Mikus, M., Besra, G. S.,
Hancock, I., and Brennan, P. J. (1996) J. Biol.
Chem. 271, 78207828
607. Lopez-Marin, L. M., Quesada, D., Lakhdar-
Ghazal, F., Tocanne, J.-F., and Lanelle, G.
(1994) Biochemistry 33, 70567061
608. Sprott, G. D., Ekiel, I., and Dicaire, C. (1990) J.
Biol. Chem. 265, 1373513740
609. Peters, J., Nitsch, M., Khlmorgen, B., Golbik,
R., Lupas, A., Kellermann, J., Engelhardt, H.,
Pfander, J.-P., Mller, S., Goldie, K., Engel, A.,
Stetter, K.-O., and Baumeister, W. (1995) J.
Mol. Biol. 245, 385401
610. Thomas, S. R., and Trust, T. J. (1995) J. Mol.
Biol. 245, 568581
611. Peters, J., Baumeister, W., and Lupas, A.
(1996) J. Mol. Biol. 257, 10311041
612. Fischetti, V. A. (1991) Sci. Am. 264(Jun), 5865
613. Goward, C. R., Scawen, M. D., Murphy, J. P.,
and Atkinson, T. (1993) Trends Biochem. Sci. 18,
136139
614. Schneewind, O., Fowler, A., and Faull, K. F.
(1995) Science 268, 103106
615. Knrle, R., and Hbner, W. (1995) Biochemistry
34, 1097010975
616. Shaper, N. L., Hollis, G. F., Douglas, J. G.,
Kirsch, I. R., and Shaper, J. H. (1988) J. Biol.
Chem. 263, 1042010428
617. Lin, C. Q., and Bissell, M. J. (1993) FASEB J. 7,
737743
618. Har-El, R., and Tanzer, M. L. (1993) FASEB J.
7, 11151123
619. Mayne, R., and Burgeson, R. E., eds. (1987)
Structure and Function of Collagen Types,
Academic Press, New York, Orlando, San
Diego
620. Nimni, M. E., ed. (1988) Collagen: Biochemistry,
Biomechanics, Biotechnology, CRC Press, Boca
Raton, Florida
621. Ramachandran, G. N. (1988) Intl. J. Peptide
Prot. Res. 31, 116
622. Martin, G. R., Timpl, R., Mller, P. K., and Khn,
K. (1985) Trends Biochem. Sci. 10, 285287
623. Prockop, D. J., and Kivirikko, K. I. (1995) Ann.
Rev. Biochem. 64, 403434
624. van der Rest, M., and Garrone, R. (1991)
FASEB J. 5, 28142823
625. Byers, P. H. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 3 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 40294077, McGraw-
Hill, New York
626. Brodsky, B., and Shah, N. K. (1995) FASEB J. 9,
15371546
627. Koivu, J., and Myllyl, R. (1987) J. Biol. Chem.
262, 61596164
628. Prockop, D. J. (1990) J. Biol. Chem. 265, 15349
15352
629. Annunen, P., Autio-Harmainen, H., and
Kivirikko, K. I. (1998) J. Biol. Chem. 273, 5989
5992
630. Reiser, K., McCormick, R. J., and Rucker, R. B.
(1992) FASEB J. 6, 24392449
631. Li, S.-W., Sieron, A. L., Fertala, A., Hojima, Y.,
Arnold, W. V., and Prockop, D. J. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 51275130
632. Meek, K. M., Chapman, J. A., and Hardcastle,
R. A. (1979) J. Biol. Chem. 254, 1071010714
633. Chan, V. C., Ramshaw, J. A. M., Kirkpatrick,
A., Beck, K., and Brodsky, B. (1997) J. Biol.
Chem. 272, 3144131446
553b. MacLennan, D. H., and Green, N. M. (2000)
Nature (London) 405, 633634
553c. Zhang, Z., Lewis, D., Strock, C., and Inesi, G.
(2000) Biochemistry 39, 87588767
554. Crider, B. P., Andersen, P., White, A. E., Zhou,
Z., Li, X., Mattsson, J. P., Lundberg, L., Keeling,
D. J., Xie, X.-S., Stone, D. K., and Peng, S.-B.
(1997) J. Biol. Chem. 272, 1072110728
555. Nanda, A., Brumell, J. H., Nordstrm, T.,
Kjeldsen, L., Sengelov, H., Borregaard, N.,
Rotstein, O. D., and Grinstein, S. (1996)
J. Biol. Chem. 271, 1596315970
555a. Kelly, R. B. (1999) Trends Biochem. Sci. 24,
M29M33
556. Desjardins, M., Celis, J. E., van Meer, G.,
Dieplinger, H., Jahraus, A., Griffiths, G., and
Huber, L. A. (1994) J. Biol. Chem. 269, 32194
32200
557. Peng, S.-B., Crider, B. P., Tsai, S. J., Xie, X.-S.,
and Stone, D. K. (1996) J. Biol. Chem. 271,
33243327
558. Pearse, B. M. F., and Crowther, R. A. (1987)
Ann. Rev. Biophys. Biophys. Chem. 16, 4968
559. Schmid, S. L., and Damke, H. (1995) FASEB J.
9, 14451453
559a. Marsh, M., and McMahon, H. T. (1999) Science
285, 215220
559b. Heuser, J. (1981) Trends Biochem. Sci. 6, 6468
560. Pearse, B. M. F., and Bretscher, M. S. (1981)
Ann. Rev. Biochem. 50, 85101
561. Pishvaee, B., Munn, A., and Payne, G. (1997)
EMBO J. 16, 22272239
562. Rothman, J. E. (1996) Protein Sci. 5, 185194
563. Brodsky, F. M., Hill, B. L., Acton, S. L.,
Nthke, I., Wong, D. H., Ponnambalam, S.,
and Parham, P. (1991) Trends Biochem. Sci. 16,
208213
564. Lemmon, S. K., and Jones, E. W. (1987) Science
238, 504509
564a. Anderson, R. G. W. (1998) Ann. Rev. Biochem.
67, 199225
564b. Shin, J.-S., Gao, Z., and Abraham, S. N. (2000)
Science 289, 785788
564c. Mulvey, M. A., and Hultgren, S. J. (2000)
Science 289, 732733
565. Lowe, M., and Kreis, T. E. (1995) J. Biol. Chem.
270, 3136431371
566. Schekman, R., and Orci, L. (1996) Science 271,
15261533
566a. Eugster, A., Frigerio, G., Dale, M., and Duden,
R. (2000) EMBO J. 19, 39053917
567. Bloomer, A. C., Champness, J. N., Bricogne,
G., Staden, R., and Klug, A. (1978) Nature
(London) 276, 362368
567a. Rapoport, I., Chen, Y. C., Cupers, P., Shoelson,
S. E., and Kirchhausen, T. (1998) EMBO J. 17,
21482155
567b. Huang, K. M., DHondt, K., Riezman, H., and
Lemmon, S. K. (1999) EMBO J. 18, 38973908
567c. Owen, D. J., Vallis, Y., Pearse, B. M. F.,
McMahon, H. T., and Evans, P. R. (2000)
EMBO J. 19, 42164227
567d. Wendland, B., Steece, K. E., and Emr, S. D.
(1999) EMBO J. 18, 43834393
567e. Scales, S. J., and Scheller, R. H. (1999) Nature
(London) 401, 123124
568. Ungewickell, E., Ungewickell, H., Holstein, S.
E. H., Lindner, R., Prasad, K., Barouch, W.,
Martin, B., Greene, L. E., and Eisenberg, E.
(1995) Nature (London) 378, 632635
569. Mellman, I., Fuchs, R., and Helenius, A.
(1986) Ann. Rev. Biochem. 55, 663700
570. Brown, M. S., and Goldstein, J. L. (1974) Sci.
Am. 251(Nov), 5866
571. Brown, M. S., and Goldstein, J. L. (1986)
Science 232, 3447
572. Holstein, T. W., Benoit, M., Herder, G.,
Wanner, G., David, C. N., and Gaub, H. E.
(1994) Science 265, 402404
References
573. Ferro-Novick, S., and Jahn, R. (1994) Nature
(London) 370, 191193
574. Mayorga, L. S., Bern, W., Sarrouf, M. N.,
Colombo, M. I., Creutz, C., and Stahl, P. D.
(1994) J. Biol. Chem. 269, 3092730934
575. Burgoyne, R. D. (1988) Nature (London) 331, 20
576. Wilson, D. W., Wilcox, C. A., Flynn, G. C.,
Chen, E., Kuang, W.-J., Henzel, W. J., Block,
M. R., Ullrich, A., and Rothman, J. E. (1989)
Nature (London) 339, 355359
577. Colombo, M. I., Taddese, M., Whiteheart, S.
W., and Stahl, P. D. (1996) J. Biol. Chem. 271,
1881018816
578. Goda, Y. (1997) Proc. Natl. Acad. Sci. U.S.A. 94,
769772
579. Lugtenberg, B. (1981) Trends Biochem. Sci. 6,
262266
580. Graham, L. L., Beveridge, T. J., and Nanninga,
N. (1991) Trends Biochem. Sci. 16, 328329
581. Oliver, D. B. (1987) in Escherichia coli and
Salmonella typhi-murium (Neidhardt, F. C.,
ed), pp. 5670, Am. Soc. for Micro-biology,
Washington, DC
582. Glauner, B., Hltje, J.-V., and Schwarz, U.
(1988) J. Biol. Chem. 263, 1008810095
583. Park, J. T. (1987) in Escherichia coli and
Salmonella typhimurium (Neidhardt, F. C.,
ed), pp. 2332, Am. Soc. for Microbiology,
Washington, DC
584. Scherrer, R. (1984) Trends Biochem. Sci. 9, 242
245
585. Nikaido, H., and Vaara, M. (1987) in
Escherichia coli and Salmonella typhimurium
(Neidhardt, F. C., ed), pp. 722, Am. Soc. for
Microbiology, Washington, DC
586. Hancock, R. E. W., Karunaratne, D. N., and
Bernegger-Egli, C. (1994) in Bacterial Cell Wall
(New Comprehensive Biochemistry), Vol. 27
(Ghuysen, J.-M., and Hakenbeck, R., eds), pp.
263279, Elsevier, Amsterdam
587. Raetz, C. R. H., and Dowhan, W. (1990) J. Biol.
Chem. 265, 12351238
588. Osborn, M. J. (1969) Ann. Rev. Biochem. 38,
501538
589. Choi, D.-S., Yamada, H., Mizuno, T., and
Mizushima, S. (1986) J. Biol. Chem. 261, 8953
8957
590. Diedrich, D. L., and Schnaitman, C. A. (1978)
Proc. Natl. Acad. Sci. U.S.A. 75, 37083712
591. Raetz, C. R. H., Ulevitch, R. J., Wright, S. D.,
Sibley, C. H., Ding, A., and Nathan, C. F.
(1991) FASEB J. 5, 26522660
592. Hollingsworth, R. I., and Carlson, R. W. (1989)
J. Biol. Chem. 264, 93009303
593. Ogawa, T. (1994) Eur. J. Biochem. 219, 737742
594. Rietschel, E. T., Kirikae, T., Schade, F. U.,
Mamat, U., Schmidt, G., Loppnow, H., Ulmer,
A. J., Zhringer, U., Seydel, U., Di Padova, F.,
Schreier, M., and Brade, H. (1994) FASEB J. 8,
217225
595. Masoud, H., Moxon, E. R., Martin, A.,
Krajcarski, D., and Richards, J. C. (1997)
Biochemistry 36, 20912103
596. Aspinall, G. O., and Monteiro, M. A. (1996)
Biochemistry 35, 24982504
597. Rietschel, E. T., and Brade, H. (1992) Sci. Am.
267(Aug), 5461
598. Costerton, J. W., Irwin, R. T., and Cheng, K. J.
(1981) Crit. Rev. Microbiol 8, 303338
599. Fischer, W. (1994) in Bacterial Cell Wall (New
Comprehensive Biochemistry), Vol. 27 (Ghuysen,
J.-M., and Hakenbeck, R., eds), pp. 199215,
Elsevier, Amsterdam
600. Ganfield, M.-C. W., and Pieringer, R. A. (1980)
J. Biol. Chem. 255, 51645169
601. Johnson, S. D., Lacher, K. P., and Anderson, J.
S. (1981) Biochemistry 20, 47814785
602. Ivatt, R. J., and Gilvarg, C. (1979) J. Biol. Chem.
254, 27592765
451
References
634. Wess, T. J., Hammersley, A. P., Wess, L., and
Miller, A. (1998) J. Mol. Biol. 275, 255267
634a. Kramer, R. Z., Venugopal, M. G., Bella, J.,
Mayville, P., Brodsky, B., and Berman, H. M.
(2000) J. Mol. Biol. 301, 11911205
635. Eyre, D. R., Paz, M. A., and Gallop, P. M.
(1984) Ann. Rev. Biochem. 53, 717748
636. Miller, A. (1982) Trends Biochem. Sci. 7, 1318
637. Eyre, D. R. (1980) Science 207, 13151322
638. Wu, J.-J., and Eyre, D. R. (1984) Biochemistry
23, 18501857
639. Hanson, D. A., and Eyre, D. R. (1996) J. Biol.
Chem. 271, 2650826516
640. Kiss, I., Dek, F., Holloway, R. G., Jr., Delius,
H., Mebust, K. A., Frinberger, E., Argraves, W.
W., Tsonis, P. A., Winterbottom, N., and
Goetink, P. F. (1989) J. Biol. Chem. 264, 8126
8134
641. Tanaka, S., Avigad, G., Eikenberry, E. F., and
Brodsky, B. (1988) J. Biol. Chem. 263, 17650
17657
642. Kleman, J.-P., Aeschlimann, D., Paulsson, M.,
and van der Rest, M. (1995) Biochemistry 34,
1376813775
643. Mazzorana, M., Gruffat, H., Sergeant, A., and
van der Rest, M. (1993) J. Biol. Chem. 268,
30293032
644. Martin, G. R., Timpl, R., Miller, P. K., and
Kuhn, K. (1985) Trends Biochem. Sci. 10, 285287
645. Gunwar, S., Ballester, F., Noelken, M. E., Sado,
Y., Ninomiya, Y., and Hudson, B. G. (1998)
J. Biol. Chem. 273, 87678775
646. van der Rest, M., and Mayne, R. (1988) J. Biol.
Chem. 263, 16151618
647. de Crombrugghe, B., and Pastan, I. (1982)
Trends Biochem. Sci. 7, 1113
648. Myers, J. C., Yang, H., DIppolito, J. A.,
Presente, A., Miller, M. K., and Dion, A. S.
(1994) J. Biol. Chem. 269, 1854918557
649. Lui, V. C. H., Ng, L. J., Sat, E. W. Y., Nicholls,
J., and Cheah, K. S. E. (1996) J. Biol. Chem. 271,
1694516951
650. de Wet, W., Bernard, M., Benson-Chanda, V.,
Chu, M.-L., Dickson, L., Weil, D., and
Ramirez, F. (1987) J. Biol. Chem. 262, 16032
16036
651. Chu, M.-L., de Wet, W., Bernhard, M., and
Ramirez, F. (1985) J. Biol. Chem. 260, 2315
2370
652. Kramer, J. M. (1994) FASEB J. 8, 329336
653. Engel, J. (1997) Science 277, 17851786
654. Rosenbloom, J., Abrams, W. R., and Mecham,
R. (1993) FASEB J. 7, 12081218
655. Hukins, D. W. L. (1984) Connective Tissue
Matrix, MacMillan, New York
656. Fransson, L.-. (1987) Trends Biochem. Sci. 12,
406411
657. Burg, M. A., Tillet, E., Timpl, R., and Stallcup,
W. B. (1996) J. Biol. Chem. 271, 2611026116
658. Caplan, A. I. (1984) Sci. Am. 251(Oct), 8494
659. Heinegrd, D., and Oldberg, . (1989) FASEB
J. 3, 20422051
660. Cheng, F., Heinegrd, D., Fransson, L.-.,
Bayliss, M., Bielicki, J., Hopwood, J., and
Yoshida, K. (1996) J. Biol. Chem. 271, 28572
28580
661. Hauser, N., and Paulsson, M. (1994) J. Biol.
Chem. 269, 2574725753
662. Beck, K., Gambee, J. E., Bohan, C. A., and
Bchinger, H. P. (1996) J. Mol. Biol. 256, 909923
663. Yurchenco, P. D., and Schittny, J. C. (1990)
FASEB J. 4, 15771590
663a. Yurchenco, P. D., Birk, D. E., and Mecham, R.
P., eds. (1994) Extracellular Matrix Assembly
and Structure, Academic Press, San Diego,
California
664. Utani, A., Nomizu, M., Timpl, R., Roller, P. P.,
and Yamada, Y. (1994) J. Biol. Chem. 269,
1916719175
665. Pschl, E., Mayer, U., Stetefeld, J.,
Baumgartner, R., Holak, T. A., Huber, R., and
Timpl, R. (1996) EMBO J. 15, 51545159
666. Durkin, M. E., Gautam, M., Loechel, F., Sanes,
J. R., Merlie, J. P., Albrechtsen, R., and Wewer,
U. M. (1996) J. Biol. Chem. 271, 1340713416
667. Beckmann, G., Hanke, J., Bork, P., and Reich,
J. G. (1998) J. Mol. Biol. 275, 725730
668. Colognato-Pyke, H., ORear, J. J., Yamada, Y.,
Carbonetto, S., Cheng, Y.-S., and Yurchenco, P.
D. (1995) J. Biol. Chem. 270, 93989406
669. Kramer, J. M., Cox, G. N., and Hirsch, D.
(1985) J. Biol. Chem. 260, 19451951
670. Beck, K., Hunter, I., and Engel, J. (1990)
FASEB J. 4, 148160
671. Sasaki, T., Kostka, G., Ghring, W.,
Wiedemann, H., Mann, K., Chu, M.-L., and
Timpl, R. (1995) J. Mol. Biol. 245, 241250
672. Aeschlimann, D., and Paulsson, M. (1991) J.
Biol. Chem. 266, 1530815317
673. Reinhardt, D. P., Ono, R. N., and Sakai, L. Y.
(1997) J. Biol. Chem. 272, 12311236
673a. Kettle, S., Yuan, X., Grundy, G., Knott, V.,
Downing, A. K., and Handford, P. A. (1999) J.
Mol. Biol. 285, 12771287
673b. Ritty, T. M., Broekelmann, T., Tisdale, C.,
Milewicz, D. M., and Mecham, R. P. (1999) J.
Biol. Chem. 274, 89338940
674. Reinhardt, D. P., Keene, D. R., Corson, G. M.,
Pschl, E., Bchinger, H. P., Gambee, J. E., and
Sakai, L. Y. (1996) J. Mol. Biol. 258, 104116
675. McKusick, V. A. (1991) Nature (London) 352,
279281
676. Francke, U., and Furthmayr, H. (1994) N.
Engl. J. Med. 330, 13841385
677. Mann, K., Mechling, D. E., Bchinger, H. P.,
Eckerskorn, C., Gaill, F., and Timpl, R. (1996)
J. Mol. Biol. 261, 255266
678. Durkin, M. E., Carlin, B. E., Vergnes, J., Bartos,
B., and Merlie, J. (1987) Proc. Natl. Acad. Sci.
U.S.A. 84, 15701574
679. Richards, A. G. (1978) in Biochemistry of Insects
(Rockstein, M., ed), Academic Press, New
York (p. 205)
680. Schaefer, J., Kramer, K. J., Garbow, J. R., Jacob,
G. S., Stejskal, E. O., Hopkins, T. L., and
Speirs, R. D. (1987) Science 235, 12001204
681. Simkiss, K., and Wilbur, K. M. (1989)
Biomineralization, Academic Press, San Diego,
California
682. DeOliveira, D. B., and Laursen, R. A. (1997) J.
Am. Chem. Soc. 119, 1062710631
683. Weiner, S., and Addadi, L. (1991) Trends
Biochem. Sci. 16, 252256
684. Albeck, S., Aizenberg, J., Addadi, L., and
Weiner, S. (1993) J. Am. Chem. Soc. 115, 11691
11697
685. Bonucci, E., ed. (1992) Calcification in Biological
Systems, CRC Press, Boca Raton, Florida
686. Mann, S., Archibald, D. D., Didymus, J. M.,
Douglas, T., Heywood, B. R., Meldrum, F. C.,
and Reeves, N. J. (1993) Science 261, 12861292
687. Aizenberg, J., Hanson, J., Ilan, M.,
Leiserowitz, L., Koetzle, T. F., Addadi, L., and
Weiner, S. (1995) FASEB J. 9, 262268
688. Otsubo, K., Katz, E. P., Mechanic, G. L., and
Yamauchi, M. (1992) Biochemistry 31, 396402
689. Fledelius, C., Johnsen, A. H., Cloos, P. A. C.,
Bonde, M., and Qvist, P. (1997) J. Biol. Chem.
272, 97559763
689a. Ducy, P., Schinke, T., and Karsenty, G. (2000)
Science 289, 15011504
689b. Teitelbaum, S. L. (2000) Science 289, 15041508
690. Weiner, S., and Traub, W. (1992) FASEB J. 6,
879885
691. McLean, F. M., Keller, P. J., Genge, B. R.,
Walters, S. A., and Wuthier, R. E. (1987) J. Biol.
Chem. 262, 1048110488
692. Komori, T., Yagi, H., Nomura, S., Yamaguchi,
A., Sasaki, K., Deguchi, K., Shimizu, Y.,
Bronson, R. T., Gao, Y.-H., Inada, M., Sato, M.,
Okamoto, R., Kitamura, Y., Yoshiki, S., and
Kishimoto, T. (1997) Cell 89, 755764
693. Ducy, P., Zhang, R., Geoffroy, V., Ridall, A. L.,
and Karsenty, G. (1997) Cell 89, 747754
694. Dickman, S. (1997) Science 276, 1502
695. Rodan, G. A., and Harada, S.-i. (1997) Cell 89,
677680
696. Mundlos, S., Otto, F., Mundlos, C., Mulliken,
J. B., Aylsworth, A. S., Albright, S., Lindhout,
D., Cole, W. G., Henn, W., Knoll, J. H. M.,
Owen, M. J., Mertelsmann, R., Zabel, B. U.,
and Olsen, B. R. (1997) Cell 89, 773779
697. Price, P. A., and Williamson, M. K. (1985) J.
Biol. Chem. 260, 1497114975
698. Nishimoto, S. K., Araki, N., Robinson, F. D.,
and Waite, J. H. (1992) J. Biol. Chem. 267,
1160011605
699. Price, P. A., Rice, J. S., and Williamson, M. K.
(1994) Protein Sci. 3, 822830
700. Hohenester, E., Maurer, P., and Timpl, R.
(1997) EMBO J. 16, 37783786
701. Kelm, R. J., Jr., Swords, N. A., Orfeo, T., and
Mann, K. G. (1994) J. Biol. Chem. 269, 30147
30153
702. Fisher, L. W., Robey, P. G., Tuross, N., Otsuka,
A. S., Tepen, D. A., Esch, F. S., Shimasaki, S.,
and Termine, J. D. (1987) J. Biol. Chem. 262,
1345713463
703. Denhardt, D. T., and Guo, X. (1993) FASEB J.
7, 14751482
704. Shanmugam, V., Chackalaparampil, I.,
Kundu, G. C., Mukherjee, A. B., and
Mukherjee, B. B. (1997) Biochemistry 36, 5729
5738
704a. Rodan, G. A., and Martin, T. J. (2000) Science
289, 15081514
704b. Service, R. F. (2000) Science 289, 14981500
705. Zipkin, I., ed. (1973) Biological Mineralization,
Wiley, New York
706. Schlesinger, P. H., Blair, H. C., Teitelbaum, S.
L., and Edwards, J. C. (1997) J. Biol. Chem. 272,
1863618643
707. Alberts, B., Bray, D., Lewis, J., Raff, M.,
Roberts, K., and Watson, J. D. (1994) Molecular
Biology of the Cell, 3rd ed., Garland, New York
(p. 11821186)
708. Riggs, B. L., and Melton, L. J. (1992) N. Engl. J.
Med. 327, 620627
709. Sly, W. S., and Hu, P. Y. (1995) in The Metabolic
and Molecular Bases of Inherited Disease, 7th ed.,
Vol. 3 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 41134124, McGraw-
Hill, New York
710. Roush, W. (1996) Science 273, 1170
711. Delmas, P. D., and Meunier, P. J. (1997) N.
Engl. J. Med. 336, 558566
711a. Ho, A. M., Johnson, M. D., and Kingsley, D.
M. (2000) Science 289, 265 270
711b. Hagmann, M. (2000) Science 289, 225 226
712. Hohling, H. J., Barckhaus, R. H., and Kreftin,
E. R. (1980) Trends Biochem. Sci. 5, 811
713. Outka, D. E., and Williams, D. C. (1971)
Journal of Protozoology 18, 285297
714. Inoue, S., and Okazaki, K. (1977) Sci. Am.
236(Apr), 8392
715. Volcani, B. E. (1981) in Silicon and Silicious
Structures in Biological Systems (Simpson, T. L.,
and Volcani, B. E., eds), Springer-Verlag, New
York (p. 157)
716. Mann, S. (1988) Nature (London) 332, 119124
717. Calvert, P. (1988) Nature (London) 334, 651652
718. Watanabe, N., and Wilbur, K. M., eds. (1976)
The Mechanisms of Mineralization in
Invertebrates and Plants, Univ. N. Carolina
Press, Columbia, South Carolina
452 Chapter 8. Lipids, Membranes, and Cell Coats
Study Questions
1. Compare the chemical makeup of the extracellar
coat or matrix materials secreted by the follow-
ing cells: bacteria, fibroblasts, osteoblasts, plant
cells, fungi.
2. The iodine number of a compound is defined as the
number of grams of I
2
absorbed (through addition
to C = C bonds to give a diiodo derivative) per 100 g
of fat. NOTE: Iodine monochloride or iodine
monobromide are the usual halogenating reagents
but the iodine number is expressed in terms of
grams of I
2
. The saponification number is the
number of milligrams of KOH needed to complete-
ly saponify (hydrolyze and neutralize the resulting
fatty acids) 1 g of fat. A pure triglyceride has a
saponification number of 198 and an iodine num-
ber of 59.7.
a) What is the relative molecular mass?
b) What is the average chain length of the fatty
acids?
c) What is the number of double bonds in the
molecule?
3. Spermaceti (a wax from the head of the sperm
whale) resembles high molecular mass hydrocar-
bons in physical properties and inertness toward
Br
2
/ CHCl
3
and KMNO
4
; on qualitative analysis, it
gives positive tests only for carbon and hydrogen.
However, its IR spectrum shows the presence of an
ester linkage, and quantitative analysis gives the
empirical formula C
16
H
32
O. A solution of the wax
in alcoholic KOH is refluxed for a long time. Titra-
tion of an aliquot shows that one equivalent of base
is consumed for every 475 g of wax. Water and
ether are added to the cooled reflux mixture, and
the aqueous and etheral layers are separated. Acid-
ification of the aqueous layer yields a solid A with a
neutralization equivalent of 260 + 5. Evaporation
of the ether layer gives solid B, which could not be
titrated. Reduction of either spermaceti or A by
lithium aluminum hydride gave B as the only
product.
What is the likely structure of spermaceti?
4. Stearic acid (1.16 g) was dissolved in 100 ml of
ethanol. A 10 l portion of the resulting solution
was pipetted onto a clean surface of a dilute HCl
solution (in a shallow tray) where it spread to form
a monolayer of stearic acid. The layer was com-
pressed (by moving a Teflon barrier across the tray)
until the surface pressure started to rise sharply
and reached ~ 20 dyn / cm. Note that =
o
-
where is the measured surface tension with the
film present and
o
is the higher surface tension of
water alone. The compressed film occupied a 20 x
24 cm area. Calculate the cross-sectional area of an
alkyl chain in stearic acid. [See J. B. Davenport, in
Biochemistry and Methodology of Lipids (A. R.
Johnson and J. B. Davenport, eds.), pp. 47-83.
Wiley-Intersience, New York, 1971; and M. C.
Phillips, in Progress in Surface and Membrane Science
(J. F. Danielli, D. M. Rosenberg, and D. A. Caden-
head, eds.) Vol. 5, pp. 139-221. Academic Press,
New York, 1972.]
5. In 1925, E. Gorter and F. Grendel (J. Exp. Med. 41,
439) reported measurements in which they extract-
ed lipid from red blood cell membranes with ace-
tone, spread the lipids as a monolayer, and
measured the area of the compressed monolayer.
They then estimated the surface area of an erythro-
cyte and calculated that the ratio of the lipids (as a
monolayer) to the surface area of the red blood cell
was 1.9 2.0. More modern experiments gave the
following: each erythrocyte membrane contains 4.5
x 10
16
mol of phospholipid and 3.1 x 10
16
mol of
cholesterol.
a) If the cross-sectional areas of phospholipid and
cholesterol molecules in a membrane are taken
as 0.70 and 0.38 nm
2
, respectively, what surface
area would be occupied in a monolayer?
b) If the measured surface area of an erythrocyte is
167 m
2
, what is the ratio of the area calculated
in (a) to the area of the cell surface?
c) How might you explain the difference between
this answer and that of Gorter and Grendel?
See E. D. Korn (1966) Science 153, 1491 1498.
References
719. Lemann, J., Jr. (1993) N. Engl. J. Med. 328,
880881
720. Nakagawa, Y., Abram, V., Kzdy, F. J., Kaiser,
E. T., and Coe, F. L. (1983) J. Biol. Chem. 258,
1259412600
721. Bertrand, J. A., Pignol, D., Bernard, J.-P.,
Verdier, J.-M., Dagorn, J.-C., and Fontecilla-
Camps, J. C. (1996) EMBO J. 15, 26782684
722. Bimmler, D., Graf, R., Scheele, G. A., and Frick,
T. W. (1997) J. Biol. Chem. 272, 30733082
723. De Reggi, M., Gharib, B., Patard, L., and
Stoven, V. (1998) J. Biol. Chem. 273, 49674971
724. Cheung, H. S., Kurup, I. V., Sallis, J. D., and Ryan,
L. M. (1996) J. Biol. Chem. 271, 2808228085
725. Rothschild, B. M., and Thillaud, P. L. (1991)
Nature (London) 349, 288
726. Cabib, E., Roberts, R., and Bowers, B. (1982)
Ann. Rev. Biochem. 51, 763793
727. Ballou, C. E., and Raschke, W. C. (1974)
Science 184, 127134
728. Frevert, J., and Ballou, C. E. (1985) Biochemis-
try 24, 753759
729. Celerin, M., Ray, J. M., Schisler, N. J., Day, A.
W., Stetler-Stevenson, W. G., and Laudenbach,
D. E. (1996) EMBO J. 15, 44454453
453
Study Questions
6. The following experimental observations are relat-
ed to biological membrane structure and function.
Discuss the implications of each observation with
respect to membrane structure.
a) Many macrocyclic antibiotics (nonactin, valino-
mycin, and others) form 1: 1 complexes with
alkali metal cations in a highly selective manner.
The complexes are readily soluble in nonpolar
organic solvents. These antibiotics increase the
electrical conductance and permeability to alkali
metal cations of synthetic phospholipid mem-
branes. Valinomycin increases the electrical
conductance of thylakoid membranes of chloro-
plasts in the presence of K
+
but not in the pres-
ence of Na
+
; it also uncouples oxidative phosho-
rylation in mitochondria. (See Chapter 18.)
b) Treatment of intact chloroplasts with a galacto-
lipase releases glactose from galactosyl digly-
cerides. Treatment of red blood cell ghosts with
phopholipase C releases about 75% of the lipid
P in water-soluble form. In neither case is the
structural integrity of the membrane destroyed.
c) Using sphingomyelin as a hapten, antibodies
specific for this lipid can be produced. When
red blood cell ghosts are exposed to these anti-
bodies, it can be shown that the antibodies react,
but only on one side of the membrane.
7. Describe the structure of biological membranes and
the characteristic functions of lipid-, protein-, and
carbohydrate-containing components. Describe the
differences between inner and outer membrane
surfaces.
8. Compare the distribution of triglycerides, phos-
phatidylcholine, phosphatidylethanolamine, sphin-
gomyelins, glycolipids, and cholesterol within cells.
Consider differences between the two sides of
membranes.
9. Consider the chemistry underlying the labeling of
cell surfaces with each of the following:
a) Lactoperoxidase,
b) galactose oxidase,
c) formylmethionylsulfone methyl phosphate,
d) the diazonium salt of diiodosulfanilic acid,
e) fluorescent antibodies,
f) antibodies conjugated with ferritin.
Write the equations for the chemical reactions
involved. State what surface groups will be labeled
by each reagent. List special advantages of each of
these reagents.
10. Which would be the more effective detergent in the
pH range 2 to 3, sodium lauryl sulfonate or sodium
laurate? Why?
11. Suppose that a cell contains 10 mM Na
+
and 100
mM K
+
and that it is bathed in extracellular fluid
containing 100 mM Na
+
and 5 mM K
+
. How much
energy will be required to transport three equiva-
lents of Na
+
out and two equivalents of K
+
in?
Compare this with G for hydrolysis of ATP at
pH 7. Assume that the membrane is permeable to
Cl

.
12. An E. coli cell is said to contain about 10
5
molecules
of an envelope protein of MW = 36,500. If the
latter is spherical and the spheres are closely packed
in a hexogonal lattice, how much of the surface area
of the bacterium would be covered? What would
the diameter of the protein be? What spacing
would be required if 10
5
molecules covered the
surface completely? Suggest a shape for the protein
molecule that is consistent with the requirement.
454 Chapter 9. Enzymes: The Catalysts of Cells
D. Mechanisms of Catalysis
1. The Transition State
Quantitative transition state theory
Transition state inhibitors
Describing the transition state
Getting to the transition state
2. Microscopic Reversibility
3. Acid and Base Catalysis
Acidic and basic groups in enzymes
Acidbase catalysis of mutarotation
General base and general acid catalysis
The Bronsted relationships
Concerted acidbase or tautomeric catalysis
4. Hydrogen Bonding and the Transfer of
Protons within Active Sites
Ultrafast proton transfer
Proton transfer rates
Marcus theory
Diffusion-controlled dissociation of protons
Coupled proton transfers
Unusually strong hydrogen bonds
5. Covalent Catalysis
6. Proximity and Orientation
7. The Microenvironment
8. Strain and Distortion
9. Why Oligomeric Enzymes?
10. Summary
E. Classification of Enzymes
References
Study Questions
A. Information from Kinetics
1. Measuring the Speed of an Enzymatic Reaction
First-order reactions
Turnover numbers and units of activity
Second-order reactions
Reversible chemical reactions
2. Formation and Reaction of EnzymeSubstrate
Complexes
Linear forms for rate equations
Nonlinear equations and integrated rate equations
Kinetics with high enzyme concentrations
3. Diffusion and the Rate of Encounter of an Enzyme
with Substrate
The cage effect and rotation of molecules
The rate of substrate binding
The displacement of bound ligands by substrate
4. Reversible Enzymatic Reactions
Reactions of two or more substrates
Ping-pong mechanisms
Isomechanisms
Dead-end complexes
Handling rate equations for complex mechanisms
The rapid equilibrium assumption
Isotope exchange at equilibrium
5. Kinetics of Rapid Reactions
Flowing substrates together
Observing relaxation
Rapid photometric methods
Some results
6. Cryoenzymology
7. The Effect of pH on Enzymatic Action
B. Inhibition and Activation of Enzymes
1. Competitive Inhibitors
2. Noncompetitive Inhibition and Activation;
Allosteric Sites
3. Inhibitors in the Study of Mechanisms
4. Allosteric Effectors in the Regulation of Enzyme
Activity
5. Irreversible Inhibition of Enzymes
C. The Specificity of Enzymatic Action
1. Complementarity of Substrate and Enzyme
Surfaces
2. Stereospecificity and Prochiral Centers
Stereochemical numbering
Trigonal prochiral centers
3. Induced Fit and Conformational Changes
4. Specificity and k
cat
5. Proofreading
455
455
457
457
458
458
458
460
460
461
461
462
463
463
464
464
465
466
466
466
467
467
468
468
468
468
468
469
469
471
471
473
475
475
477
478
478
478
480
480
481
481
482
482
482
483
484
484
486
486
486
486
487
487
490
490
491
491
492
493
493
493
493
494
494
495
496
497
497
497
498
501
456
470
473
479
485
488
Boxes
Box 9-A A Historical Note on Enzymes
Box 9-B Growth Rates of Cells
Box 9-C The Sulfonamides as Antimetabolites
Box 9-D Receptors, Agonists, and Antagonists
Box 9-E Chorismate Mutase
Box 9-F Immunophilins as Rotamases
Contents
Bimolecular reactions of two molecules, A and B, to give two products, P and
Q, are catalyzed by many enzymes. For some enzymes the substrates A and B
bind into the active site in an ordered sequence while for others, binding may
be in a random order. The scheme shown here is described as random Bi Bi in a
classification introduced by Cleland. Eighteen rate constants, some second order
and some first order, describe the reversible system. Determination of these
kinetic parameters is often accomplished using a series of double reciprocal
plots (Lineweaver-Burk plots), such as those at the right.
[B]
vf
1
[A]
1
Vf
10
Vf
6
Vf
1
E
EA
EB
EAB EPQ
EQ
EP
E
k
9
k
10
k
5
k
6
B
A k
1
k
2
k
4
k
3
A
B
k
8
k
7
Q
P
Q
P
k
13
k
11
k
12
k
14
k
15
k
16
k
17
k
18
Random Bi Bi Mechanism
455
Enzymes: The Catalysts of Cells
9
Most of the machinery of living cells is made of
enzymes. Thousands of them have been extracted from
cells and have been purified and crystallized. Many
others are recognized only by their catalytic action and
have not yet been isolated in pure form. Most enzymes
are soluble globular proteins but an increasing number
of RNA molecules are also being recognized as enzymes.
Many structural proteins of the cell also act as catalysts.
For example, the muscle proteins actin and myosin
together catalyze the hydrolysis of ATP and link the
hydrolysis to movement (Chapter 19). Catalysis is one
of the most fundamental characteristics of life.
How do we recognize that a protein or RNA mole-
cule is an enzyme? The answer is that enzymes are
recognized primarily by their ability to catalyze a chemical
reaction. For this reason, an everyday operation for
many biochemists is the measurement of the catalytic
activity of enzymes. Only by measuring the rates of
the catalyzed reactions carefully and quantitatively
has it been possible to isolate and purify these remark-
able molecules.
Since the beginning of biochemical investigation
enzymes have held a special fascination for chemists
and biologists. How can these easily destroyed sub-
stances catalyze reactions with such speed and without
formation of significant quantities of side products?
Some enzymes increase the velocity of a single chemical
reaction of a specific compound by a factor of as much
as 10
10
. How can a protein do this? In this chapter
well consider both ways of measuring enzymatic
activity and basic mechanisms of catalysis.
A. Information from Kinetics
The quantitative study of catalysis by enzymes, i.e.,
the study of enzyme kinetics, is a highly developed
branch of biochemistry. It is one of our most important
means of learning about the mechanisms of catalysis
at the active sites of enzymes.
1 13a
By determining
rate constants k under a variety of conditions we can
learn just how fast an enzyme can act, how tightly it
binds its substrates to form the enzyme-substrate (ES)
complexes essential to catalysis, how specific it is with
respect to substrate structure, and how it is affected by
compounds that inhibit or activate the catalysis.
1. Measuring the Speed of an Enzymatic
Reaction
A major goal in kinetic studies is to establish a rate
equation which describes the velocity of a reaction in
terms of kinetic constants and other experimentally
measurable parameters. To measure the velocity of any
chemical reaction precisely we must start the reaction
at a definite time by rapidly mixing together the two
or more reactants. Then, while keeping the mixture at
an accurately constant temperature and pH, we must
measure the concentration of a reactant or product after
a fixed time interval, or at various times. No end of
ingenuity has gone into devising ways of doing this for
particular enzymes. Whatever the procedure, the infor-
mation we must obtain is the rate at which some con-
centration changes with time. We can then construct a
progress curve showing the decrease in concentration
[S] of the reactant (substrate) or the increase in the
concentration of product [P] with time (Fig. 9-1).
[B]
v
f
1
[A]
1
V
f
10
V
f
6
V
f
1
E
EA
EB
EAB EPQ
EQ
EP
E
k
9
k
10
k
5
k
6
B
A k
1
k
2
k
4
k
3
A
B
k
8
k
7
Q
P
Q
P
k
13
k
11
k
12
k
14
k
15
k
16
k
17
k
18
Random Bi Bi Mechanism
456 Chapter 9. Enzymes: The Catalysts of Cells
BOX 9-A A HISTORICAL NOTE ON ENZYMES
(9-1)
The velocity v of an enzymatic reaction is defined
as the rate at which a substrate disappears or at which
a product is formed, the two being identical:
v = d[S] / dt = d[P] / dt
Under the steady-state conditions that usually apply
(see p. 449), the rate of increase of product will be the same
as the rate of decrease of substrate. The units of velocity
are moles per liter per second (M s
1
) or more tradition-
ally in enzymology moles per liter per minute. We are
interested in the instantaneous velocity, which at
any time is given by the slope of the progress curve
(Fig. 9-1). We usually want to measure the velocity
immediately after the reaction is started to avoid the
decrease in rate that comes from the depletion of sub-
strate or accumulation of products. In some cases, as
in the example given in Fig. 9-1, this is difficult to do
with accuracy.
In some chemical reactions, which involve first-
order processes, a logarithmic plot of the progress curve
(log[S] vs t) gives a straight line so that the initial slope
need not be determined. However, most enzyme assays
give progress curves that remain nearly linear for only
short periods of time. In these cases we need
very sensitive methods for detecting products.
These may involve colorimetric or fluorimetric
measurements or the use of radioactive sub-
strates. One of the most sensitive approaches
is to arrange the assay so that a product of the
reaction serves as a catalyst for another enzy-
matic process, thus amplifying the amount of
final product to be measured.
14,15
Another
approach is to measure velocities of reactions
in a very small volume, e.g., 200 nanoliters,
and to continuously separate and measure
products using capillary electrophoresis and
fluorescence detection.
16
If the progress curve is not a straight line
at the beginning of the reaction (0 time) and if
the amount of compound that reacts in a fixed
t 2t
If half as much enzyme is present,
it will take twice as long to reach
a given point on the progress curve.
The initial rate is given
by the slope of this line.
At any later time, the slope
is less than at time = zero.
T
h
e

d
e
c
r
e
a
s
e

i
n
c
o
n
c
e
n
t
r
a
t
i
o
n
o
f

s
u
b
s
t
r
a
t
e
,

S
Time

[
S
]
Figure 9-1 The progress curve for an enzymatic reaction in
which the substrate S is converted into products.
While the earliest physiologists postulated a
vital force to explain the chemical reactions of
cells, the existence of biochemical substances pro-
moting reactions outside of the body was recognized
at least by the early 1600s. However, the role of
yeast in fermentation was still unknown and it was
thought that both alcoholic fermentation and animal
digestion were caused by unknown substances
called ferments. In 1752, Reamur demonstrated the
solvent power of the gastric juice of birds and by 1783
Spallanzani had extended the studies to humans
and other species.
a
In 1836 Schwann isolated the
enzyme pepsin
b
from gastric juice.
In the same year Jacob Berzelius introduced the
concept of catalysis, which he developed as a result
of studies of the effects of acids and bases in pro-
moting the hydrolysis of starch and of the effects of
metals on the decomposition of hydrogen peroxide.
Berzelius proposed the term catalyst from the Greek
katalysis, meaning dissolution. Although he
had been concerned primarily with inorganic cata-
lysts, Berzelius recognized that a natural catalyst,
an amylase that causes the hydrolysis of starch, had
already been isolated from germinating barley in
1833 and that in living plants and animals thou-
sands of catalytic processes take place.
c
Some
chemists and physiologists accepted Berzelius
concept of biochemical catalysis immediately, but
many did not. The matter was complicated by
Pasteurs discovery that yeast cells were the caus-
ative agent of alcoholic fermentation. Only after
Edward Buchners reports in 1897 that a juice
formed by grinding yeast with sand and filtering
could still ferment sugar (see Chapters 15 and 17)
was the reality of enzymes in metabolism
d
generally
accepted. The word enzyme (from the Greek in
yeast) was introduced earlier by F. W. Kuhne, pro-
fessor of physiology in Heidelberg and a person
who had accepted Berzelius concept.
c
a
Richmond, C. (1986) Trends Biochem. Sci. 11, 528 530
b
Schwann, T. (1836) Arch. Anat. Physiol., 90 138
c
Hoffmann-Ostenhof, O. (1978) Trends Biochem. Sci. 3, 186 187
d
Buchner, E. (1897) Ber. Deut. Chem. Ges. 30, 117 124
457
1
2
(9-5)
(9-4)
(9-3)
A. Information from Kinetics
interval of time is taken as the rate, an erroneous
answer will be obtained. Sometimes an integrated
rate expression that describes the time course of
product formation can be used (see Eq. 9-22). Even
when the progress curve is nonlinear as shown in Fig.
9-1, it is possible to estimate relative rates by noting
that in most cases if one unit of enzyme yields a cer-
tain amount of product in time t
1
, the same amount of
product will be formed by n units of enzyme in time
t
1
/n.
First-order reactions. In many chemical reactions
the rate of decrease of the concentration of a given
reactant [A] is found experimentally to be directly
proportional to the concentration of that reactant at
any given time:
v = d[A] / dt = k[A]
Such a reaction is described as first order and the pro-
portionality constant k is known as the rate constant.
Such first-order kinetics is observed for unimolecular
processes in which a molecule of A is converted into
product P in a given time interval with a probability
that does not depend on interaction with another
molecule. An example is radioactive decay. Enzyme
substrate complexes often react by unimolecular pro-
cesses. In other cases, a reaction is pseudo-first order;
compound A actually reacts with a second molecule
such as water, which is present in such excess that its
concentration does not change during the experiment.
Consequently, the velocity is apparently proportional
only to [A].
A first-order rate constant k has units of s
1
. When
[A] = 1, v = k. Thus, k is a measure of the speed in mol
l
1
s
1
of the reaction of a substance at unit activity.
As a first-order reaction proceeds, [A] decreases and
at time t is given by any one of the following three
equivalent expressions (Eq. 9-3). These are obtained
by integration of Eq. 9-2 in which t
0
is the time at
which the reaction was started.
[A] = A
0
e
kt
ln ( [A
0
] / [A] ) = kt
log [A
0
] log [A] = kt / 2.303
Equation 9-3 is the equation of exponential decay. A
characteristic of exponential decay is that [A] is halved
in a time that is independent of concentration. The
half-life is t :
t = ln 2 / k = 0.693 / k
The relaxation time for A is defined by Eq. 9-5 and
represents the time required for the concentration [A]
to fall to 1/ e (or ~ 0.37) of its initial value.
Turnover numbers and units of activity. When
an enzyme is catalyzing product formation at the maxi-
mum possible rate, V
max
, we can usually assume that
the active site of every molecule of enzyme contains a
substrate in the form of an intermediate enzyme
substrate complex ES and that this complex is being
converted to products according to Eq. 9-6:
d[P] / dt = V
max
= k[ES] = k[E]
t
or V
max
= k
cat
[E]
t
Here E
t
is the total enzyme, namely, the free enzyme
E plus enzymesubstrate complex ES. The equation
holds only at substrate saturation, that is, when the
substrate concentration is high enough that essentially
all of the enzyme has been converted into the inter-
mediate ES. The process is first order in enzyme but
is zero order in substrate. The rate constant k is a
measure of the speed at which the enzyme operates.
When the concentration [E]
t
is given in moles per liter
of active sites (actual molar concentration multiplied by
the number of active sites per mole) the constant k is
known as the turnover number, the molecular activity,
or k
cat
. The symbol k
cat
is also used in place of k in Eq.
9-6 for complex rate expressions in which k
cat
cannot
represent a single rate constant but is an algebraic
expression that contains a number of different constants.
Turnover numbers can be measured only when
the concentration of the enzyme is known. Partly for
this reason the activity of an enzyme is usually given
as specific activity, the units of activity per milligram
of protein. One international unit is the amount of
enzyme that produces 1 mol of product per minute
under standard (usually optimal) conditions. The
International Union of Biochemistry
17
has recommended
a larger unit, the katal (kat), the amount of enzyme
that converts one mol s
1
of substrate to product.
1 kat = 6 x 10
7
international units
1 international unit = 16.67 nkat (nanokatals)
If the enzyme is pure and is saturated with substrate
under the standard assay conditions, the following
relationships hold.
Turnover No. = katals/mol of active sites
= [nkat/mg] x M
r
x 10
6
/n
= [international units/mg] x M
r
x 10
3
/60n
Here M
r
is the relative molecular mass of the enzyme
and n is the number of active sites per molecule. Since
the activity of an enzyme is dependent on both tem-
perature and pH, these variables must be specified.
Turnover numbers of enzymes vary from <1 to
~ 10
6
s
1
. Trypsin, chymotrypsin, and many intracellular
(9-2)
(9-6)
1
2
1
2
= 1/k = t /ln 2
458 Chapter 9. Enzymes: The Catalysts of Cells
enzymes have turnover numbers of ~ 10
2
s
1
. The
fastest enzymes, which include catalase (Chapter 16),
carbonic anhydrase (Chapter 13), and
5
-3-oxosteroid
isomerase (Chapter 13), have maximum turnover
numbers of 2 x 10
5
s
1
or more. Compare these reaction
rates with those of a typical organic synthesis in the
laboratory. A reaction mixture must often be heated
for hours (k < 10
3
s
1
). Many enzymes accelerate rates
by factors of greater than 10
6
over those observed in
the absence of an enzyme at a comparable temperature
and pH. Enzymes often bring two or more substrates
together, binding them at a specific location in their
active sites. Because of this, rapid reactions can be
catalyzed even when the reactants are present in low
concentrations.
Second-order reactions. For a chemical reaction
to occur between two molecules, A and B (Eq. 9-7),
they must meet and collide.
The velocity of such a second-order process is charac-
terized by a bimolecular rate constant k
2
and is pro-
portional to the product of the concentrations of A
and B:
v = k
2
[A] [B]
The units of k
2
are M
1
s
1
. If [B] is present at unit
activity, the rate is k
2
[A], a quantity with units of s
1
.
We can see that the bimolecular, or second-order, rate
constant for reaction of A with B may be compared
with first-order constants when the second reactant
B is present at unit activity. In many real situations,
reactant B is present in large excess and in a virtually
constant concentration. The reaction is pseudo-first
order and the experimentally observed rate constant
k
2
[B] is an apparent first-order rate constant. The bimole-
cular rate constant k
2
can be obtained by dividing the
apparent constant by [B].
Reversible chemical reactions. In any reversible
process, we must consider rate constants for both the
forward and the reverse reactions. At equilibrium a
reaction proceeds in the forward direction at exactly
the same velocity as in the reverse reaction so that no
change occurs. For this reason there is always a rela-
tionship between the equilibrium constant and the rate
constants. For Eq. 9-9, k
1
is the bimolecular rate constant
for the forward reaction and k
2
the unimolecular rate
constant for the reverse reaction. The equilibrium
constant K can easily be shown, from Eq. 9-2 and 9-8,
to equal k
1
/k
2
for the reaction of Eq. 9-9.
The student should be aware that in kinetic equa-
tions rate constants are usually numbered consecutively
via subscripts and that the subscripts do not imply any-
thing about the molecularity. The system which is used
here employs odd-numbered constants for steps in the
forward direction and even-numbered constants for
steps in the reverse direction. However, many authors
number the steps in the forward direction consecutively and
those in the reverse direction with corresponding negative
subscripts.
What relationships exist between experimentally
observable rates and k
1
and k
2
for a reversible reaction?
Consider first the simplest case (Eq. 9-10):
If pure A is placed in a solution, its concentration will
decrease until it reaches an equilibrium with the B
which has been formed. It is easy to show that in this
case [A] does not decay exponentially but [A] [A]
equil
does. If log([A] [A]
equil
) is plotted against time a
first-order rate constant k, characteristic of the rate of
approach to equilibrium, will be obtained. Its relationship
to k
1
and k
2
is given by Eq. 9-11.
k = k
1
+ k
2
The relaxation time for approach to equilibrium can
be expressed as follows:

1
= k =
1
1
+
2
1
= k
1
+ k
2
and for the more complex case of Eq. 9-9 as
k =
1
= k
1
( [A]
e
+ [B]
e
) + k
2
where [A]
e
and [B]
e
are the equilibrium concentrations
of A and B.
18
2. Formation and Reaction of Enzyme
Substrate Complexes
An abundance of evidence indicates that the first
step in enzymatic catalysis is the combining of the
enzyme and substrate reversibly to form a complex,
ES (Eq. 9-14). Formation of the complex is normally
reversible. ES can either break up to form enzyme
and substrate again or it can undergo conversion to a
product or products, often by a unimolecular process.
Three rate constants are needed to describe this system
for a reaction that is irreversible overall. A complete
A + B P
k
2
(9-7)
(9-8)
A + B P
k
1
k
2
(9-9)
A B
k
1
k
2
(9-10)
(9-11)
(9-12)
(9-13)
Equilibrium constant
P
A B
K
k
k
[ ]
[ ][ ]
1
2
459
description of the kinetic behavior is fairly involved.
For example, the kinetics may be different within cells,
where molar concentrations of enzymes often exceed
those of substrate, than in the laboratory. In most
laboratory experiments the enzyme is present at an
extremely low concentration (e.g., 10
8
M) while the
substrate is present in large excess. Under these cir-
cumstances the steady-state approximation can be
used. For this approximation the rate of formation of ES
from free enzyme and substrate is assumed to be exactly
balanced by the rate of conversion of ES on to P. That is,
for a relatively short time during the duration of the
experimental measurement of velocity, the concentra-
tion of ES remains essentially constant. To be more
precise, the steady-state criterion is met if the absolute
rate of change of a concentration of a transient inter-
mediate is very small compared to that of the reactants
and products.
19
The MichaelisMenten equation (Eq. 9-15)
describes the initial reaction rate of a single substrate
with an enzyme under steady-state conditions.
This can be rearranged as follows:
Equation 9-15 provides a relationship between the
velocity observed at a particular substrate concentration
and the maximum velocity that would be achieved at
infinite substrate concentration. The quantities V
max
and K
m
are often referred to as the kinetic parameters
of an enzyme and their determination is an important
part of the characterization of an enzyme. Equation
9-15 can be derived by setting the rate of formation of
the ES complex (k1[E][S]) in the steady state equal to
its rate of breakdown, ([k
2
+ k
3
][ES]). Rearranging and
substituting K
m
, as defined in Eq. 9-15, we obtain Eq.
9-17.
Using this equation, together with a mass balance
relationship ([E] = [E]
t
[ES]), we can solve for [ES]/[E]
t
,
the fraction of enzyme combined as enzymesubstrate
complex (Eq. 9-18).
The maximum velocity, V
max
= k
3
[E]
t
, is attained only
when all of the enzyme is converted into ES. Under
other conditions v = k
3
[ES] and Eq. 9-19 holds.
[ES] / [E]
t
= v/V
max
Substituting from Eq. 9-19 into Eq. 9-18 gives the
MichaelisMenten equation (Eq. 9-15).
In many cases, the rate at which ES is converted
back to free E and S is much greater than the rate of
conversion of ES to products (k
2
k
3
). In such cases K
m
equals k
2
/k
1
, the dissociation constant for breakdown
of ES to free enzyme and substrate (sometimes called
K
s
). Thus, K
m
sometimes has a close inverse relationship
to the strength of binding of substrate to enzyme. In such
cases, 1/K
m
is a measure of the affinity of the substrate
for its binding site on the enzyme and for a series of
different substrates acted on by the same enzyme.
The more tightly bound substrates have the lower
values of K
m
. But beware! The condition that k
3
is
negligible compared to k
2
may be met with some
(poorer) substrates but may not always be met by
others. From Eq. 9-15 we see that K
m
is always greater
than or equal to K
S
, but it may be less than K
S
for more
complex mechanisms.
Figure 9-2 shows a plot of velocity against substrate
concentration as given by Eq. 9-15. The position of
V
max
on the ordinate is marked, but it should be clear
that the experimental velocity (v) can never attain V
max
unless [S] is very high relative to K
m
. The value of v
approaches V
max
asymptotically. Since K
m
is defined
as the value of [S] at which v = V
max
/ 2, its value can
be estimated from Fig. 9-2. However, K
m
cannot be
determined reliably because of the difficulty of estab-
lishing the value of V
max
from a plot of this type. Notice
that the curve of Figure 9-2 is identical in form to the
saturation curve for reversible binding shown in Fig. 7-1.
A. Information from Kinetics
S
S
S
where
v
V
K
V
K
K k k k
m m
m

+
+
max max
[ ]
[ ]
[ ]
( )
1
2 3 1
(9-15)
(9-16)
[ ][ ]
( )
[ ] [ ] E S ES ES
+

k k
k
K
m
2 3
1
(9-17)
[ ]
[ ]
[ ]
[ ]
ES
E
S
S
t m
K

+ (9-18)
(9-19)
[S]
K
m
V
max
V
max
v
1
2
Figure 9-2 Plot of observed velocity v vs substrate concen-
tration [S] for an enzyme-catalyzed reaction.
E + S ES
k
1
k
2
k
3
E + P
(9-14)
V
max
v
= 1 + K
m
/[S]
460 Chapter 9. Enzymes: The Catalysts of Cells
Linear forms for rate equations. To obtain K
m
and V
max
from experimental rate data, Eq. 9-15 can be
transformed by algebraic rearrangement into one of
several linear forms. The popular double-reciprocal
or LineweaverBurk plot of 1/ v against 1/ [S] (Fig. 9-3)
is described by Eq. 9-20. The values of K
m
/V
max
and
1/V
max
can be evaluated from the slope and intercept,
respectively, of this straight line plot.
Another linear plot, the EadieHofstee plot, is that
of v/ [S] vs v (Fig. 9-4). It is related to the Scatchard
plot (Fig. 7-3) and is fitted by Eq. 9-21.
While the point for [S] = 0 and v = 0 cannot be plotted,
the ratio v/ [S] approaches V
max
/K
m
as v approaches
zero. Notice the distribution of the points in Fig. 9-4.
Substrate concentrations were chosen such that the
increase in velocity from point to point is more or less
constant, a desirable experimental situation. The points
on the EadieHofstee plot are also nearly evenly dis-
tributed, but those of the LineweaverBurk plot are
compressed at one end. (However, if the substrate
concentrations for successive points are selected in
the ratios 1, 1/ 3, 1/ 5, 1/ 7, and 1/ 9, the spacing will
be uniform on the LineweaverBurk plot.) A second
advantage of the EadieHofstee plot is that the entire
range of possible substrate concentration from near
zero to infinity can be fitted onto a single plot.
As pointed out at the beginning of Section A,
depletion of substrate with time lowers [S] from its
initial value to some extent. This can have an especially
adverse effect on points obtained at low [S], e.g., points
on the right side of Fig. 9-3 or on the left side of Fig.
9-4. Equations 9-20 and 9-21 will be more precise if the
average substrate concentration over the time period
of the assay, rather than that at zero time,
20
is used.
Nonlinear equations and integrated rate
equations. In discussions of the control of metabolism
through regulation of enzymatic activity, it is often
better to plot v against log [S] as in Fig. 9-5. This plot
also has the virture that the entire range of attainable
substrate concentrations can be plotted on one piece
of paper if the point for [S] = 0 (at minus infinity) is
omitted. The same scale can be used for all enzymes.
The plot is S-shaped, both for simple cases that are
represented by Eq. 9-15 and for enzymes that bind
substrate cooperatively (Section B,5). Thus, the
classification or hyperbolic vs sigmoidal is lost.
However, the degree of cooperativity can be directly
measured from the midpoint slope of the curve. A
disadvantage is that it is awkward to measure V
max
from a plot of this type, and it may be preferable to
obtain it from a linear plot (Figs. 9-3 or 9-4). Alterna-
tively, computer-assisted methods can be used to
obtain both K
m
and V
max
and to fit a curve to the exper-
imental points as in Fig. 9-5.
An attractive alternative to the use of initial veloci-
ties and linear plots is to measure the kinetic parame-
ters V
max
and K
m
using points all along the progress
curve (Fig. 9-1). Various procedures for doing this
have been devised.
21,22
For example, the integrated
form of Eq. 9-15 can be given as Eq. 9-22 or 9-23.
Figure 9-3 Double-reciprocal or LineweaverBurk plot of
1/v vs 1/ [S]. The intercept on the vertical axis gives 1/V
max
and the slope gives K
m
/V
max
. The intercept on the horizontal
axis equals 1/K
m
.
1 1 1
v V
K
V
m
+
max max
[ ] S
v
1
V
max
1
K
m
1
slope =
K
m
V
max
[S]
1
v
V
K
v
K
m m
[ ]
max
S

1
(9-21)
V
max
v
[S]
v
K
m
V
max
Figure 9-4 The EadieHofstee plot of v/ [S] against v. The
slope is 1/K
m
; the intercept on the vertical axis is V
max
/K
m
and that on the horizontal axis is V
max
.
(9-20)
461
V
max
t = [S]
0
[S] + K
m
ln ( [S]
0
/ [S] )
Here [S]
0
is the initial substrate concentration at t = 0
and [S] and [P] are the substrate and product concen-
trations at any later time. In one type of linear plot,
21
based on Eq. 9-23, ([S]
0
[S]) / t is plotted against 1/ t
ln ([S]
0
/ [S]), the slope of the plot being K
m
and the
intercepts being V
max
and V
max
/K
m
. Computer pro-
grams are available for analysis of enzymatic progress
curves, even for complex mechanisms.
23,24
Kinetics with high enzyme concentrations.
Laboratory studies of the kinetics of purified enzymes
are usually conducted with enzyme concentrations of
10
7
to 10
10
M, but within cells enzyme concentrations
are probably often in the range 10
6
to 10
5
M,
25
which
may be higher than the concentrations of the substrates
upon which the enzymes act. Be cautious in drawing
conclusions about kinetics under such circumstances!
Methods have been devised for handling kinetic data
when the concentration of enzyme is greater than K
m
,
a condition that can lead to intolerably high errors if
the usual equations are applied.
26,27
3. Diffusion and the Rate of Encounter of an
Enzyme with Substrate
What determines the value of k
1
in Eq. 9-14? This
rate constant represents the process by which the sub-
strate and enzyme find each other, become mutually
oriented, and bind to form ES. If orientation and bind-
ing are rapid enough, the rate will be determined by
the speed with which the molecules can come together
by diffusion. Large molecules in solution are free to
travel for only a tiny fraction of their diameter as a
result of their frequent collision with solvent molecules.
Presence of a competitive
inhibitor will shift entire
curve according to Eq. 9-59
so that higher substrate
concentrations are required
to reach saturation.
log K
m
2 log K
m
1 log K
m
log K
m
+1
log
10
[S]
V
max
v
V
max
1
2
Figure 9-5 Plot of v against log [S] for an enzyme-catalyzed reaction.
V t K
m max
[ ] ln
[ ]
[ [ ]
+

P
S
S] P
0
0 (9-22)
The result is visible in the Brownian
movement of microscopic particles sus-
pended in a fluid. If an individual particle
is followed, it is seen to undergo a random
walk, moving in first one direction then
another. Albert Einstein showed that if the
distances transversed by such particles in
a given time t are measured, the mean
square of these x values
2
can be related
by Eq. 9-24 to the diffusion constant D
(which is usually given in units of cm
2
s
1
).

2
= 2 D t
For molecules, the Brownian move-
ment cannot be observed directly but the
diffusion constant can be measured, for example, by
observing the rate of spreading of a boundary between
two different concentrations of the substance.
28
The
diffusion constant for
1
H
2
HO (HDO) in H
2
O at 25C is
2.27 x 10
5
cm
2
s
1
, and the values for the ions K
+
and
Cl

are about the same.


29
For many small molecules D
is approximately 10
5
cm
2
s
1
, and the value decreases as
the size of the molecule increases. Thus, for the 13.7-kDa
ribonuclease, D = 1.1 x 10
6
cm
2
s
1
, and for the 500-kDa
myosin, D is ~ 1 x 10
7
cm
2
s
1
. For the spherical particle
the StokesEinstein equation (Eq. 9-25) can be used
to relate the diffusion coefficient to the radius, the
coefficient of viscosity , the Boltzmann constant k
B
,
and the temperature (Kelvin) T.
To estimate the rate constant for a reaction that
is controlled strictly by the frequency of collisions of
particles, we must ask how many times per second
one of a number n of particles will be hit by another of
the particles as a result of Brownian movement. The
problem was analyzed in 1917 by Smoluchowski,
30,31
who considered the rate at which a particle B diffuses
toward a second particle A and disappears when the
two collide. Using Ficks law of diffusion, he concluded
that the number of encounters per milliliter per second
was given by Eq. 9-26.
Number of encounters/ml s
1
=
4 (D
A
+ D
B
) (r
A
+ r
B
) n
A
n
B
Here D
A
and D
B
are the two diffusion coefficients, r
A
and
r
B
are the radii (in ) of the two particles, and n
A
and n
B
are the numbers of particles per milliliter. The number
of encounters per liter per second is (4 /1000) (D
A
+ D
B
)
(r
A
+ r
B
) N
2
[A] [B], where N is Avogadros number.
Dividing this frequency by N gives the rate of collision
v in M s
1
, a velocity that can be equated (Eq. 9-27)
with k
D
[A] [B], where k
D
is a second-order rate constant
A. Information from Kinetics
(9-24)
D
k T
r

B
6 (9-25)
(9-23)
(9-26)
462 Chapter 9. Enzymes: The Catalysts of Cells
Division of Eq. 9-31 by Eq. 9-32 gives the volume swept
out per second by one particle:
V/ t = k
B
T/ 3 cm
3
s
1
Since the collision radius for two particles of equal size
is two times the particle radius, the effective volume
swept out will be four times that given by Eq. 9-33.
Since both particles are diffusing, the effective diffusion
constant will be twice that used in obtaining Eq. 9-28.
Thus, the effective volume swept out by the particle
in a second will be eight times that given by Eq. 9-33.
The volume swept out by one mole of particles is equal
to k
D
(recall that the second-order rate constant has
dimensions of liter mol
1
s
1
). Thus, when converted
to a moles per liter basis and multiplied by 8, Eq. 9-33
should (and does) become identical with the Smolu-
chowski equation (Eq. 9-30).
The volume given by Eq. 9-33 is about 1.4 x 10
11
cm
3
, which could be represented approximately by a
cube 2.4 m on a side. If we compare this volume with
that of a cell (Table 1-2) or of an organelle, we see that
in one second an enzyme molecule will sweep out a
large fraction of the volume of a small cell, mitochon-
drion, chloroplast, etc.
The cage effect and rotation of molecules. It
is of interest to compare the bimolecular rate constant
for encounters calculated by the Smoluchowski theory
(Eq. 9-29) with the corresponding bimolecular rate
constant for molecular collisions given by the kinetic
theory of gases (Eq. 9-34).
Here m
A
and m
B
are the masses of the two particles.
This rate constant is also relatively independent of
molecular size and for spheres varies from (4 to 11) x
10
11
M
1
s
1
, over an order of magnitude greater than
the rate constant for encounters. In a solution, mole-
cules still collide at about the same rate as in a gas so
that 100 to 200 collisions occur between two particles for
each encounter. However, during the time of the single
encounter, the particles are together in a solvent cage.
While in this solvent cage, both substrate and enzyme
molecules undergo random rotational motions. Suc-
cessive collisions bring them together in different
orientations, one of which is likely to lead to a suffi-
ciently close match of complementary surfaces (of
substrate and binding site) so that formation of a
productive ES complex takes place.
Molecular rotation in a solution is described quan-
titatively by diffusion laws (analogous to Ficks laws)
(9-31)
which determines the diffusion controlled limit,
that is, the maximum possible rate of a reaction.
This equation can be rearranged to Eq. 9-28.
While this equation is thought to overestimate the
diffusion-limited rate constant slightly, it is a good
approximation. If the diffusing particles are approxi-
mately spherical, diffusion constants D
A
and D
B
can
be calculated from Eq. 9-25, and Eq. 9-28 becomes Eq.
9-29.
The value of k
D
does not vary greatly as the ratio
of radii r
A
/r
B
is changed, and in most cases it may be
assumed that r
A
/r
B
1, in which case the equation
simplifies (Eq. 9-30).
For water at 25C, the coefficient of viscosity is ~ 0.01
poise (1 poise = 10
5
newton cm
2
) which leads, accord-
ing to Eq. 9-30, to k
D
~ 0.7 x 10
10
M
1
s
1
. It was shown
by Debye
32
that this rate constant must be multiplied
by a correction factor when charged particles rather
than uncharged spheres diffuse together. This factor
may be of the order of 510 for a substrate and enzyme
carrying two or three charges and may act to either
increase or decrease reaction rates. Since the viscosity
of cytoplasm is 0.02 0.03 poise,
33
k
D
will be reduced
correspondingly.
A simple alternative derivation of Eq. 9-30 was
developed by Dexter French,
34
who also provided the
author with most of this discussion of encounter theory.
Consider a small element of volume V swept out by a
particle as it moves through the solution for a distance
equal to its own radius. This element of volume will
equal r
3
:
V = r
2
x r = r
3
It will be swept out in a time t which can be calculated
from Eq. 9-24 as r
2
/ 2D. Substituting the value of D
given by Eq. 9-25, we obtain for t:
v D D r r N k + +
4
1000

( )( )[ ][ ] [ ][ ]
A B A B D
A B A B
(9-27)
k
N
D D r r
D A B A B
M s + +

4
1000
1 1

( )( )
(9-28)
k
RT
r
r
r
r
D
A
B
B
A
+ +
j
(
,
\
,
(
2
3000
2

(9-29)
k
RT
D
M s

8
3000
1 1
(9-30)
t
r
k T

3
3

B
(9-32)
(9-33)
k
N r r
k T
m m
( )
( )
collision
A B
B
A B

+
+
j
(
,
\
,
(
,

,
,
]
]
]
]
2
1 2
1000
8
1 1

(9-34)
463
for which a rotary diffusion constant is defined.
35,36
Consider a group of molecules all oriented the same
way initially, then undergoing rotary diffusion until
their orientations become random. If we measure the
orientation of each molecule by an angle we see that
initially the value of cos is 1 but that when the angles
become random the mean value of cos averaged over
all molecules is zero. The rotary relaxation time is
the time required for the mean value of cos to fall to
1/e (which occurs when = 68.5). For a sphere is
given by Eq. 9-35.
Ellipsoidal or rod-shaped molecules have two different
rotary diffusion constants while, if the dimensions of
the molecules are different along all three axes, three
constants must be specified.
36
From Eq. 9-35 we can calculate (if = 0.01 poise)
the following values for a small spherical molecule
(substrate) of ~ 1 nm length and for a spherical enzyme
of 5 nm diameter:
r = 0.5 nm 1.3 x 10
9
s
1
r = 2.5 nm 1.0 x 10
7
s
1
We see that smaller molecules rotate much faster than
large ones and that rotational relaxation times for small
proteins are of the same order of magnitude as k
D
for
diffusion-limited encounter. However, for very large
molecules, especially long rods, the rotary relaxation
time about a short axis may be a large fraction of a second.
The rate of substrate binding. At very low sub-
strate concentrations the MichaelisMenten equation
(Eq. 9-15) simplifies as follows:
v = ( V
max
/K
m
) [S]
Since V
max
= k
cat
[E]
t
(Eq. 9-6) and at low [S] most of
[E]
t
is free E, we obtain the following equation:
2
v = ( k
cat
/K
m
) [E] [S]
From this we see that k
cat
/K
m
is the apparent second-
order rate constant for the reaction of free enzyme with
substrate. As such it cannot exceed the diffusion con-
trolled limit k
D
of Eqs. 9-28 to 9-30 which falls in the
range of 10
9
10
11
M
1
s
1
. Experimentally observed
Later
orientation

Initial
orientation


1
2
3
8
k T
r
B
(9-35)
values of k
cat
/K
m
are always less than this limit, indi-
cating that a certain time is required for a substrate
molecule to become oriented and seated in the active
site.
37
However, for several real enzymes values of
k
cat
/K
m
of 10
7
to 3 x 10
8
M
1
s
1
have been observed.
For triose phosphate isomerase Albery and Knowles
obtained a value of 4 x 10
8
M
1
s
1
, so close to the diffu-
sion controlled limit that these authors regard this
enzyme as a nearly perfect catalyst, one that could not
have evolved further because it is already catalyzing
the reaction with substrate at almost the maximum
velocity that is possible.
37 39
The displacement of bound ligands by sub-
strate. Jenkins pointed out that in many instances a
substrate must displace another ligand from the active
site to form the ES complex.
40
For example, a binding
site for an ionic substrate often already contains an
ion, either of the product P or some other ion X which
Jenkins calls a substrate surrogate. For this situation
Eq. 9-14 must be replaced by the following set of
equations:
Here k
2
and also k
4
and k
5
, are second-order rate con-
stants. The release of product, as determined by k
4
and k
5
, may be rate-limiting. At zero time the reverse
reactions may be ignored, and steady-state analysis
shows that the MichaelisMenten equation (Eq. 9-16b)
will be replaced by Eq. 9-39. Here, D is a constant and
A is also constant if X is present at a fixed concentration.
When k
5
, the rate constant for displacement of product
by substrate, is very small, this equation simplifies to
Eq. 9-40.
At high concentrations of X the second term becomes
negligible and the equation becomes identical to the
MichaelisMenten equation (Eq. 9-16) except that K
m
,
the apparent Michaelis constant, now increases as [X]
(9-37)
(9-36)
EX S +
k
1
k
2
X ES +
k
3
EP
product release
P
X
P
k
5
k
4
S
(9-38)
A. Information from Kinetics
V k k
k
k k k
k k
K
m
max
[ ]
[ ]
( )
[ ] [ ]
[ ] ( [ ])
+
+
+

+
+ + +
1
1 1
1
2 3
1
3 5 1
4 5
X
S X S
S D A S
(9-39)
V
k k k k k
k k K
m
max
[ ] ( [ ] ) [ ]
[ ] [ ]
+ + +
+ +
1
1
3 4 2 3 1
3 4
X X S
X S
(9-40)
,
v
v
464 Chapter 9. Enzymes: The Catalysts of Cells
v
V
K K K K
f
m m m

+ + +
f
eqA B B A
A B
A B A B
[ ][ ]
[ ] [ ] [ ][ ]
(9-44a)
1 1
1
v V
K K K K
m m m
f f
A B eqA B
A B A B
+ + +
j
(
,
\
,
(
[ ] [ ] [ ][ ]
(9-44b)
(9-42)
increases (Eq. 9-41). This is exactly what is observed
for competitive inhibitors where
Thus substrate surrogates at high concentrations are
often competitive inhibitors. However, at low [X] the
second term in Eq. 9-41 may be important and X then
serves as an activator. Under many circumstances, Eq.
9-39 does not simplify further and a nonlinear relation-
ship between 1/ v and 1/ [S] is observed, with the shape
of the curves being influenced by the values of constants
A and D (see also Section B,4).
40
4. Reversible Enzymatic Reactions
For some enzyme-catalyzed reactions the equilib-
rium lies far to one side. However, many other reac-
tions are freely reversible. Since a catalyst promotes
reactions in both directions, we must consider the
action of an enzyme on the reverse reaction. Let us
designate the maximum velocity in the forward direc-
tion as V
f
and that in the reverse direction as V
r
. There
will be a Michaelis constant for reaction of enzyme
with product K
mP
, while K
mS
will refer to the reaction
with substrate.
As in any other chemical reaction, there is a rela-
tionship between the rate constants for forward and
reverse enzyme-catalyzed reactions and the equilib-
rium constant. This relationship, first derived by the
British kineticist J. B. S. Haldane and proposed in his
book Enzymes
41
in 1930, is known as the Haldane
relationship. It is obtained by setting v
f
= v
r
for the
condition that product and substrate concentrations
are those at equilibrium. For a single substratesingle
product system it is given by Eq. 9-42.
K
eq
= V
f
K
mP
/V
r
K
ms
Because the Haldane relationship imposes con-
straints on the values of the velocity constants and
Michaelis constants, it is of some value in understand-
ing regulation of metabolism. Consider the case that
the maximum forward velocity V
f
is high and that K
mS
has a moderately low value (fairly strong binding of
substrate). If the reaction is freely reversible (K
eq
~ 1)
and the velocity of the reverse reaction V
r
is about the
same as that of the forward reaction, it will necessarily
be true (see Eq. 9-42) that the product P will also be
fairly tightly bound. If V
r
<< V
f
, the value of K
mP
will
have to be much lower than that of K
mA
. In such a
situation P will remain tightly bound to the enzyme
and since V
r
is low, it will tend to clog the enzyme.
Such product inhibition may sometimes slow down
a whole pathway of metabolism. In such a case, the
only way that an enzymatic sequence can keep going
in the forward direction is for product P to be removed
rapidly by a subsequent reaction with a second enzyme.
The first enzyme may be thought of as possessing a
kind of one-way valve that turns off the flow in a
metabolic pathway when the concentration of its
product rises.
Reactions of two or more substrates. Enzymes
frequently catalyze the reaction of two, three, or even
more different molecules to give one, two, three, or
more products. Sometimes all of the substrate mole-
cules must be bound to an active site at the same time
and are presumably lined up on the enzyme molecule
in such a way that they can react in proper sequence.
In other cases, the enzyme may transform molecule A
to a product, and then cause the product to react with
molecule B. The number of variations is enormous.
1,10,12
The order in which two molecules A and B bind
to an enzyme to form a complex EAB may be com-
pletely random or it may be obligatorily ordered. Both
situations occur with real enzymes. Cleland introduced
a widely used method of depicting the possibilities.
42
For example, Eq. 9-43 shows the reaction of A and B in
an ordered sequence to form the complex EAB which is
then isomerized to EPQ, the complex formed by bind-
ing the two products P and Q to the enzyme. The rate
constant to the left of each vertical arrow or above
each horizontal arrow refers to the reaction in the
forward direction as indicated by the arrow while the
other constants (to the right or below the arrows) refer
to the reverse reactions. The velocity in the forward
direction for an enzyme with ordered binding is given
by Eq. 9-44a,
which may also be written in the reciprocal form (Eq.
9-44b):
An alternative form of this equation (Eq. 9-45),
proposed by Dalziel,
43
is sometimes used.
K
k k
k
m

+
2 3
1
[ ] X
(9-41)
EPQ EAB EQ E EA E
A
k
1
k
2
B
k
3
k
4 k
5
k
6
P
k
7
k
8
Q
k
9
k
10
(9-43)
,
,
465
The kinetic parameters of Eq. 9-44 are V
f
, the maximum
velocity in the forward direction, the two Michaelis
constants, K
mB
and K
mA
, and the equilibrium constant
K
eqA
, for reversible dissociation of the complex EA and
which is equal to k
2 /
k
1
. The relationship between the
parameters of Eq. 9-44 (K
m
s, V s, and K
eqA
s) and the
rate constants k
1
k
10
is not obvious. However, remem-
ber that the parameters are experimental quantities
determined by measurements on the enzyme. Some-
times, but not always, it is possible to deduce some of
the values of individual rate constants from the experi-
mental parameters.
An equation similar to 9-44a can be written for the
velocity v
r
of the reaction of P and Q. Also an equation
can be written in similar form for v
f
v
r
, i.e., the instan-
taneous velocity of the reaction in any mixture of all
four components A, B, P, and Q.
The kinetic parameters of Eq. 9-44b are often
obtained from experimental data through the use of
reciprocal plots (Fig. 9-6). However, Eq. 9-44b is linear
only if the concentration of one or the other of the sub-
strates A and B is kept constant. For this reason a
series of experiments is usually performed in which
[A] is varied while [B] is held constant. Then [A] is
held constant and [B] is varied. Each of these experi-
ments leads to a family of lines (Fig. 9-6A) whose
slopes and intercepts are measured. The slopes and
intercepts of this family of curves are then plotted
against the reciprocal of the second concentration, i.e.,
[ ]
[ ] [ ] [ ][ ]
E
A B A B
t
f
v
+ + +

0
1 2 12
(9-45)
the one that was held fixed.
From a set of these secondary plots, V
f
and one
of the Michaelis constants can be determined (Fig. 9-6B
and C). Using two sets of secondary plots, all of the
constants of Eq. 9-44 may be established. Alternatively,
a computer can be used to examine all of the data at
once and to obtain the best values of the parameters.
The latter approach is desirable because estimates of
the standard deviations of the parameters can be
obtained. However, the user must take care to ensure
that the experimental errors are correctly estimated and
are not simply estimates of how well the computer has
fitted the points on the assumption that they contain
no error.
23
The meaning of the kinetic parameters may be
slightly difficult to grasp. V
f
is the velocity that would
be obtained if both [A] and [B] were high enough to
saturate the enzyme. Each K
m
corresponds to that for
a simple system in which the concentration of the
second substrate is at a high, saturating value.
For the bimolecular reaction that we have consid-
ered, there are two Haldane relationships:
Of these, only the first is ordinarily used.
Ping-pong mechanisms. A common type of
mechanism that is especially prevalent for enzymes
with tightly bound cofactors has been dubbed
ping-pong because the enzyme alternates between
Figure 9-6 Reciprocal plots used to analyze kinetics of two-substrate enzymes. (A) Plot of 1/ v
f
against 1/ [A] for a series
of different concentrations of the second substrate B. (B) A secondary plot in which the intercepts from graph A are plotted
against 1/ [B]. (C) Secondary plot in which the slopes from graph A have been plotted against 1/ [B]. The figures have been
drawn for the case that K
mA
= 10
3
M, K
mB
= 2 K
mA
, and K
AB
= K
eqA
K
mB
(Eq. 9-46) = K
mA
/ 200 and [A] and [B] are in units of moles
per liter. EadieHofstee plots of v
f
/ [A] vs v
f
at constant [B] can also be used as the primary plots. The student can easily
convert Eq. 9-44 to the proper form analogous to Eq. 9-21.
[B] = 0.5 K
mA
[B] = K
mA
[B] = 2K
mA
[B] = 3K
mA
[B] = 4K
mA
i
n
t
e
r
c
e
p
t
s
s
l
o
p
e
s
v
f
1
slope =
K
mB
V
f
slope =
K
AB
V
f
[A]
1
[B]
1
[B]
1
K
a
K
ia
K
b
1
K
b
V
f
5
V
f
K
mA
V
f
10K
mA
V
f
10
V
f
6
V
f
1
V
f
1
A B C
A. Information from Kinetics
K
V K K
V K K
V
V
K K
K K
m
m
m
m
eq
f P dQ
r dA B
f
r
dP Q
A dB

j
(
,
\
,
(
2
(9-46)
466 Chapter 9. Enzymes: The Catalysts of Cells
two forms E and E (Eq. 9-47). Substrate A reacts via
complex EA to form E, a modified enzyme that often
contains an altered coenzyme. At the same time A is
changed to product P still bound to the enzyme. P
dissociates leaving E which is then able to react with
the second substrate B and to go through the second
half of the cycle during which E is converted back to
E. An example is provided by the aminotransferases
(Eq. 14-25) in which the coenzyme pyridoxal phosphate
is interconverted with pyridoxamine phosphate.
The rate equations of the ping-pong mechanism
resemble that for the ordered bimolecular reaction (Eq.
9-44), but each has one less term (Eq. 9-48):
or
A diagnostic feature of the ping-pong mechanism
is that the families of lines (in double reciprocal plots)
obtained when one substrate is held constant while
the other is varied no longer intersect as they do in Fig.
9-6A but are parallel. They must be truly parallel and
the experimentalist must be aware that nearly parallel
lines may sometimes be observed for sequential reac-
tions. Thus, if K
eqA
of Eq. 9-44b is small enough the
last term of that equation will be small compared to
the other terms and the equation will be approximated
by Eq. 9-48b. The reaction will appear to be ping-pong
even though it is sequential and the reaction proceeds
through the ternary complex EAB.
One less kinetic parameter can be obtained from
an analysis of the data for a ping-pong mechanism than
can be obtained for ordered reactions. Nevertheless,
in Eq. 9-47, twelve rate constants are indicated. At
least this many steps must be considered to describe
the behavior of the enzyme. Not all of these constants
can be determined from a study of steady-state kinetics,
but they may be obtained in other ways.
Isomechanisms. A catalyst functions over and
over again without being altered. However, during
a single turnover it is likely to undergo a temporary
change. For example, if an enzyme assists in removing
a proton from a substrate, some functional group of
4 3
2 1
E EA
EB EAB
A
k
1
[A]
k
2
A
k
8
B
k
6
B
k
3
[B]
k
9
[A]
k
7
[B] k
5
k
4
P
(9-50)
Ping-pong mechanism for reaction
of A and B to form P and Q
E EP E EA E
A
k
1
k
2
Q
k
11
k
12
B
k
7
k
8 k
3
k
4
P
k
5
k
6
EB
k
9
k
10
EQ
(9-47)
v
V
K K
m m
f
f
B A
A B
A B A B

+ +
[ ][ ]
[ ] [ ] [ ][ ]
1 1
1
v V
K K
m m
f f
A B
A B
+ +
j
(
,
\
,
(
[ ] [ ]
(9-48a)
(9-48b)
the enzyme will be protonated during part of the
catalytic cycle. This proton must dissociate before
another catalytic cycle begins, and in some instances
this dissociation may be a rate-limiting step. Northrup
and coworkers developed the use of product inhibition
to identify such isomechanisms.
44
Dead-end complexes. In the steady state of
action of an enzyme with ping-pong kinetics, part of
the enzyme is in form E and part in form E. Ideally, E
would have affinity only for A and Q, while E would
have affinity only for B and P. However, in many real
situations E also binds B and P weakly; similarly, E
binds A and Q. This is because the products and
reactants usually have structural features in common.
The propensity of enzymes with ping-pong kinetics
to form dead-end complexes (also called abortive
complexes) may sometimes have a regulatory func-
tion. In Eq. 9-49 the reactions of Eq. 9-47 have been
rearranged to depict a situation in which product P
normally undergoes a sequence of further reactions.
However, if P accumulates to a high enough concen-
tration it can react reversibly to form a dead-end com-
plex EP. This is an effective form of product inhibition
which can be relieved only by a lowering of the con-
centration of P through its further metabolism.
Handling rate equations for complex mecha-
nisms. While steady-state rate equations can be
derived easily for the simple cases discussed in the
preceding sections, enzymes are often considerably
more complex and the derivation of the correct rate
equations can be extremely tedious. The topological
theory of graphs, widely used in analysis of electrical
networks, has been applied to both steady-state and
nonsteady-state enzyme kinetics.
4550
The method em-
ploys diagrams of the type shown in Eq. 9-50. Here
EP
(nonproductive)
E
A
Q
P
E
B
further reactions
(9-49)
467
maintained. As can be seen from the graph, this ex-
change rate increases monotonically as substrate
concentrations are increased. This is also true for the
rate of ATPADP exchange. The fact that both ex-
change rates increase continuously indicates random
binding of substrates.
55
The inequality of the two
maximal exchange rates suggests that release of glucose
6-phosphate may be slower than that of ADP.
Figure 9-7B shows similar plots for lactate dehydro-
genase.
53
In this case, after an initial rise (that is not
regarded as significant), the pyruvate*lactate exchange
reaches a high constant value as the amount of pyruvate
is increased (with a constant [pyruvate] / [lactate] ratio
of 1/35). However, the NAD*NADH exchange in-
creases rapidly at first but then drops abruptly as the
pyruvate and lactate concentrations continue to in-
crease. This suggests an ordered mechanism (Eq. 9-43)
in which NAD
+
and NADH represent A and Q, respec-
tively, and pyruvate and lactate represent B and P. As
the concentrations of B and P become very high, the
(9-51)
the reaction of an enzyme with two substrates A and
B with a random order of binding is depicted. (In
contrast, Eq. 9-43 shows the case of ordered binding
of two substrates.) When complex EAB is formed, it
can decompose to free enzyme and to the single prod-
uct P. Each one of the nodes, which are numbered 1 4
in the diagram, corresponds to a single form of the
enzyme. The appropriate first-order rate constant or
apparent first-order constant is placed by each arrow.
The methods provide easy rules for deriving from
such a scheme the steady-state rate equation.
The importance of the simplified schematic methods
is apparent when one considers that the steady-state
rate equation for Eq. 9-50 would have 6 terms in the
numerator and 12 terms in the denominator.
51
In the
more complex case in which EAB breaks down to two
products P and Q with a random order of release, the
rate equation contains 672 terms in the denominator.
In such cases it is worthwhile to enlist the help of a
computer in deriving the equation.
24,52 54
The rapid equilibrium assumption. Rate equa-
tions for enzymes are often simplified if a single step,
e.g., that of reaction of complex EAB to product in Eq.
9-50, is rate limiting.
54a
If it is assumed that all reaction
steps preceding or following the rate-limiting step are
at equilibrium, the equation for random binding with
a two-substrate and two-product reaction simplifies to
one whose form is similar to that obtained for ordered
binding (Eq. 9-44). In the absence of products P and Q
Eq. 9-44 will correctly represent the steady-state rate
equation corresponding to Eq. 9-50. However, this
simplification may not be valid for a very rapidly
acting enzyme.
Isotope exchange at equilibrium. Consider the
reaction of substrates A and B to form P and Q (Eq.
9-51). If both reactants and both products are present
with the enzyme and in the ratio found at equilibrium
no net reaction will take place. However, the reactants
and products will be continually interconverted under
the action of the enzyme. Now if a small amount of
highly labeled reactant (A* or B*) is added, the rate at
which isotope is transferred from the labeled reactant
into one or the other of the products can be measured.
In general, a label in one of the substrates will appear
in only one of the products.
Figure 9-7A shows the rate of exchange of isotopi-
cally labeled glucose (glucose*) with glucose 6-phosphate
as catalyzed by the enzyme hexokinase (Chapter 12).
The exchange rate is plotted against the concentra-
tion of glucose 6-phosphate with the ratio [glucose] /
[glucose 6-phosphate] constant at 1/19, such that an
equilibrium ratio for reactants and products is always
A + B P + Q
Figure 9-7 (A) Effect of glucose and glucose 6-phosphate
concentrations on reaction rate of yeast hexokinase at equi-
librium. Reaction mixtures contain 1 2.2 mM ATP, and 25.6
mM ADP at pH 6.5. From Fromm et al.
51
(B) Effect of lactate
and pyruvate concentrations on equilibrium reaction rates
of rabbit muscle lactate dehydrogenase. Reaction mixtures
contained 1.7 mM NAD
+
, and 30 46 M NADH in Tris-
nitrate buffer, pH 7.9, 25C. From Silverstein and Boyer.
53
10
4
0 0.5
NAD NADH
pyruvate lactate
Lactate concentration, mol l
1
(pyruvate/lactate = 1/35)
E
q
u
i
l
i
b
r
i
u
m

r
e
a
c
t
i
o
n

r
a
t
e
,
m
o
l

l
-
1

m
i
n
-
1


1
0
3
8
6
2
1.0
10
5
0 2 4
ATP ADP
glucose glucose 6-P
G 6-P molarity 10
2
, G/G 6-P = 1/19
E
q
u
i
l
i
b
r
i
u
m

r
e
a
c
t
i
o
n

r
a
t
e
,
m
o
l

l
-
1

m
i
n
-
1


1
0
4
A. Information from Kinetics
A
B
468 Chapter 9. Enzymes: The Catalysts of Cells
enzyme shuttles back and forth between EA and EQ,
but these two complexes rarely dissociate to give free
enzyme and A or Q. Hence, the A*Q exchange rate
drops.
In other cases a label may be transferred from A
into P or from B into Q. Information on such exchang-
es has provided a valuable criterion of mechanism
which is considered in Chapter 12, Section B,4.
5. Kinetics of Rapid Reactions
The fastest steps in an enzymatic process cannot be
observed by conventional steady-state kinetic methods
because the latter cannot be applied to reactions with
half-times of less than about 10 s. Consequently, a
variety of methods have been developed
18,56 59a
to
measure rates in the range of 1 to 10
13
s
1
.
Flowing substrates together. One of the first
rapid kinetic methods to be devised consists of rapidly
mixing two flowing solutions together in a special
mixing device and allowing the resulting reaction
mixture to move at a rate of several meters per second
down a straight tube. At a flow velocity of 10 m s
1
a
solution will move 1 cm in 10
3
s. Observations of the
mixture are made at a suitable distance, e.g., 1 cm, and
with various flow rates. Using spectrophotometry or
other observation techniques, the formation or disap-
pearance of a product or reactant can be followed. The
special advantage of this technique is that observation
can be made slowly. However, it may require large
amounts of precious reactant solutions, e.g., those of
purified enzymes.
In the stopped flow technique two solutions are
mixed rapidly by the flow technique during a period
of only 1 2 (or a few) milliseconds. A ram drives the
solutions from syringes through a mixing chamber
into an observation chamber. After the flow stops
light absorption, fluorescence, conductivity, or other
property, is measured. A means of rapid observation
of changes during the reaction is essential. For example,
light absorption may be measured by a photomultiplier
with data being collected by a computer. Relaxation
times as short as a few milliseconds or less can be
observed in this way.
59a,b
Observing relaxation. Kinetic measurements
over periods of tens of microseconds or less can be made
by rapidly inducing a small displacement from the
equilibrium position of a reaction (or series of reactions)
and observing the rate of return (relaxation) of the
system to equilibrium. Best known is the temperature
jump method devised by Eigen and associates. Over a
period of about 10
6
s a potential difference of ~ 100 kV
is applied across the experimental solution. A rapid
electrical discharge from a bank of condensers passes
through the solution (without any sparking) raising
the temperature 2 10 degrees. All the chemical equi-
libria for which H 0 are perturbed. If some property,
such as the absorbance at a particular wavelength or
the conductivity of the solution, is measured, very
small relaxation times can be determined.
While it may not be intuitively obvious, if the
displacement from equilibrium is small, the rate of
return to equilibrium can always be expressed as a
first-order process (e.g., see Eq. 9-13). In the event that
there is more than one chemical reaction required to
reequilibrate the system, each reaction has its own
characteristic relaxation time. If these relaxation times
are close together, it is difficult to distinguish them;
however, they often differ by an order of magnitude
or more. Therefore, two or more relaxation times can
often be evaluated for a given solution. In favorable
circumstances these relaxation times can be related
directly to rate constants for particular steps. For
example, Eigen measured the conductivity of water
following a temperature jump
18
and observed the rate
of combination of H
+
and OH

for which at 23C


equals 37 x 10
6
s. From this, the rate constant for
combination of OH

and H
+
(Eq. 9-52) was calculated
as follows (Eq. 9-53):
k = 1/ { ( [ OH

] + [ H
+
] )} = 1.3 x 10
11
M
1
s
1
Pressure jump and electric field jump methods
have also been used, as have methods depending upon
periodic changes in some property. For example, absorp-
tion of ultrasonic sound causes a periodic change in
the pressure of the system.
Rapid photometric methods. Another useful
method has been to discharge a condenser through
a flash tube over a period of 10
12
to 10
4
s, causing a
rapid light absorption in a sample in an adjacent parallel
tube. Following the flash, changes in absorption spec-
trum or fluorescence of the sample can be followed.
The availability of intense lasers as light sources has
made it possible to follow the results of light flashes of
5 10 picosecond duration and to measure extremely
short relaxation times (Chapter 23).
58,59
Some results. Rapid kinetic methods have revealed
that enzymes often combine with substrates extremely
quickly,
60
with values of k
1
in Eq. 9-14 falling in the
range of 10
6
to 10
8
M
1
s
1
. Helixcoil transitions of
polypeptides have relaxation times of about 10
8
s,
but renaturation of a denatured protein may be much
slower. The first detectable structural change in the
vitamin A-based chromophore of the light-operated
proton pump bacteriorhodopsin occurs in ~ 5 x 10
8
s,
while a proton is pumped through the membrane in
H
+
+ OH H
2
O

(9-53)
(9-52)
469
~10
4
s.
61
Interconversion between chair and boat forms
of cyclohexane derivatives may have ~ 10
5
s at room
temperature, while rotation about a C N bond in an
amide linkage may be very slow with ~ 0.1 s. The
nonenzymatic hydration of the aldehyde pyridoxal
phosphate via Eq. 13-1 occurs with = 0.01 0.1 s,
depending upon the pH.
59
6. Cryoenzymology
An alternative to studying rapid reactions is to cool
enzymes to a subzero temperature (down to 100C)
where reactions proceed more slowly.
60,62
The enzyme
must be dissolved in a suitable cryosolvent, often
containing 50 80% by volume of an organic solvent or
solvents such as methanol, ethanol, dimethyl sulfoxide,
dimethyl formamide, or ethylene glycol. Those con-
taining methanol are especially desirable because of
their low viscosities. Kinetics can be studied by various
spectroscopic methods and stopped-flow, temperature-
jump, and other rapid-reaction techniques can be
applied. One goal of cryoenzymology is to stabilize
otherwise unstable intermediates. X-ray crystallo-
graphic measurements can also be made at these low
temperatures and it should be possible to observe
structures of intermediate ES complexes. A problem
is that the forms of the complexes stabilized may be
side-products rather than true intermediates. How-
ever, as discussed in Chapter 3, a combination of low-
temperature Laue X-ray diffraction and a laser-induced
temperature jump may be feasible.
7. The Effect of pH on Enzymatic Action
Because proteins contain many acidic and basic
groups it is not surprising that the activity of enzymes
often varies strongly with pH. However, it is usually
found that the state of protonation of only a few groups
has a strong effect on activity. This is understandable
because most ionized groups are on the outer surface
of protein molecules and most are not close to an active
site. Protonation or deprotonation of those groups will
hardly ever have a major influence on events in the
active site. Often only one or two ionizable groups
have highly significant effects.
For many enzymes a plot of V
max
against pH is
a bell-shaped curve (Fig. 9-8). The optimum rate is
observed at some intermediate pH, which is often,
but not always, in the range of pH 6 9. This type of
curve can be interpreted most simply by assuming the
presence in the active site of two ionizable groups and
three forms of the enzyme with different degrees of
protonation: E, EH, and EH
2
.
A. Information from Kinetics
Let us designate the acid dissociation constants (K
a
) for
the two groups in the enzyme as K
1E
and K
2E
and those
of the ES complex K
1ES
and K
2ES
. However, as discussed
in Chapter 6, Section E,2 these consecutive K
a
values
cannot necessarily be assigned to single groups. They
may belong to a system of interacting groups. Each
may be the sum of more than one microscopic K
a
and
there may be extensive tautomerism within the active
site. The rate constants k
1
, k
2
, and k
3
define the rates of
formation and breakdown of the ES complex.
If it is assumed that only form EHS reacts to give
products, the bell-shaped curves of Fig. 9-8 are obtained.
The frequent observation of such curves supports the
model of Eq. 9-54. It also suggests that the two ioniz-
able groups may be intimately involved in catalysis,
one as an acid and the other as a conjugate base (see
Section E,5).
For the simple case illustrated in Eq. 9-54, the pH
dependence of the initial maximum velocity, the appar-
ent Michaelis constant, and V
max
/K
m
are given by Eqs.
9-55 to 9-57.
EH
2
k
3
EH + products EH EHS
EH
2
S
E ES
K
1E
K
2E
K
2ES
K
1ES
Both ionizable
groups protonated
No protons on either
ionizable group
k
1
k
2
S +
(9-54)
Figure 9-8 Expected dependence of V
max
on the pH accord-
ing to Eq. 9-54 with k
3
[E
t
] = 1, pK
1ES
= 6, and pK
2ES
as given on
the graph. After Alberty.
63
Computer-drawn graph courtesy
of Carol M. Metzler.
4 5 6 7 8 9 10
pH
0.0
0.2
0.4
0.6
0.8
1.0
11
V
m
a
x
6 7 8 10
470 Chapter 9. Enzymes: The Catalysts of Cells
BOX 9-B GROWTH RATES OF CELLS
How do we correctly describe the rate at which
a cell grows? Consider bacteria in their rapidly
growing log phase.
a
Each cell divides after a fixed
length of time, the doubling time. For E. coli this
may be as short as 17 min in the early stages of
growth but becomes somewhat longer as time goes
on. A mean value of about 26 min. for E. coli at 37C
is typical. In contrast, the doubling time for mam-
malian cells in tissue culture is often about one day.
If a given volume of culture contains N
0
cells
initially, the number N
n
after n cell divisions will be:
N
n
= 2
n
N
0
From this equation we can calculate that a single
bacterium with a generation time of 20 min can
produce 2
144
cells in 48 h of exponential growth.
The exponential growth rate constant k is equal
to the number of doublings per unit time. Thus, k is the
reciprocal of the doubling time. It is easy to show
that the number of bacteria present at time t will be
given by the following equation.
N
t
= 2
kt
N
0
This can be rewritten in a form that can be used to
determine k by counting the number of bacteria at
zero time and at time t.
kt = log
2
(N
t
/N
0
) = log
10
(N
t
/N
0
) / 0.301
Another way of expressing the growth is to
equate the rate of increase of the number of bacteria
with a growth rate constant multiplied by the
number of bacteria present at that time.
dN/dt = N
This is a general equation for an autocatalytic reac-
tion and N could be replaced with a concentration,
for example, the total content of cellular matter per
liter in the medium. From the two preceding equa-
tions the following can be derived.
a
= k ln 2 = 0.69k
When bacteria are transferred to new medium
there is usually a lag before exponential growth
begins. Exponential growth eventually stops and the
culture enters stationary phase, which is usually
followed by relatively rapid death of cells in the
culture. It is often desirable to study cell growth
under conditions of continuous cultivation in
which a constant generation time is maintained but
the density of cells in the medium does not increase.
This can be done with a simple device known as the
chemostat.
a,b
A culture vessel containing bacteria
is stirred to ensure homogeneity. Fresh culture
medium continuously enters the vessel from a
reservoir and part of the content of the vessel, sus-
pended bacteria included, is continuously removed
through another tube. The bacterial population in
the vessel builds up to a constant level and can be
maintained at the same level for relatively long
periods of time.
Here are some other statistics on cell growth.
Bacteria growing exponentially expand their linear
dimensions by 1.5 nm and synthesize 1.6 x 10
7
Da of
new cell material in one second. This is equivalent
to ~1000 small proteins and includes 23 ribosomes
and 3000 base pairs of DNA at each growing point.
The much larger HeLa human tumor cell grows by
0.13 nm / s but makes 4.6 x 10
8
Da/ s of material.
c
a
Stanier, R. Y., Douderoff, M., and Adelberg, E. A. (1970) The
Microbial World, 3rd ed., Prentice-Hall, Englewood Cliffs, New
Jersey (pp. 298 324)
b
Smith, H. L., and Waltman, P., eds. (1995) The Theory of the
Chemostat; Dynamics of Microbial Competition, Cambridge
University Press, London and New York
c
Pollard, E. C. (1973) in Cell Biology in Medicine (Bittar, E. E., ed),
pp. 357 377, Wiley, New York
V
k
K K
k
t t
max
[ ]
( [ ] [ ])
[ ]

+ +

+ +
3
1 2
3
1
E
H H
E
F
ES ES 2ES
K
k k
k
K K
K K
K F F
m m E ES

+ +
+ +

+ +
+ +
( ) ( [ ] [ ])
( [ ] [ ])
2 3
1
1 2
1 2
2 2
1
1
H H
H H
E E
ES ES
V
K
k k
k k K K
k
K F
m
t
m E
max
( ) ( [ ] [ ])
[ ]

+ +

+ +
1 3
2 3 1 2
3
2
1
1 H H
E
E E
(9-55)
(9-56)
(9-57)
The denominators (which are the Michaelis pH
functions given by the first three terms of Eq.
3-6) represent the fraction of enzyme or of ES
complex in the monoprotonated state. The
pH dependence of enzymatic action is often
more complex than that shown in Fig. 9-8 and
given by the foregoing equations. However,
it is easy to write Michaelis pH functions (see
Chapter 3) for enzymes with any number of
dissociable groups in both E and ES and to
write appropriate equations of the type of
471
Eqs. 9-55 to 9-57. Bear in mind that if the free sub-
strate contains groups dissociating in the pH range of
interest, a Michaelis pH function for the free substrates
will also appear in the numerator of Eq. 9-56. If the pH
dependence of the enyzme is regulated by a conforma-
tional change in the protein, there may be a coopera-
tive gain or loss of more than one proton and the
Michaelis pH function must reflect this fact. This can
sometimes be accomplished by addition of a term
related to Eq. 7-45. For more information see Dixon
and Webb,
64
Cleland,
65,66
or Kyte.
67
Plots of log V
max
(or log-specific activity) and log
K
m
or log (V
max
/K
m
) versus pH yield graphs of the
type shown in Fig. 9-9. The curved segments of the
graphs that extend for about 1.5 pH units on either
side of each pK
a
are asymptotic to straight lines of
slope 1 when a single proton is involved or of a higher
slope for multiple cooperative proton dissociation.
The straight lines can be extrapolated and intersect at
the pK
a
values. (However, it is better to fit a complete
curve of theoretically correct shape to the points.)
Note that the curved line always passes below or
above the intersection point at the value of log 2 = 0.30
except in the case of cooperative proton dissociation
when it is closer.
Upward turns in the curve of log K
m
vs pH corre-
spond to pK
a
values in the free enzyme or substrate
and downward turns to pK
a
values in ES. This approach
to analysis of pH dependence has been adopted widely
but often incorrectly. For example, many published
curves have very sharp bends in which the curved
portion covers less than 3 pH units and the curve is
much closer than 0.30 to the extrapolated point. This
suggests cooperative proton binding and an apparent
pK
a
that is related to K of Eq. 7-21. Cooperativity is
always a possibility if a conformational change in the
protein is involved.
67
The simple treatment given above is based on the
assumption that all proton dissociations are rapid
compared to k
cat
, that enzyme in only one state of
protonation binds substrate, and that ES in only one
state of protonation yields products. These assump-
tions are not always valid. It also assumes that both
binding and dissociation of substrate are rapid, that is,
to use Clelands terminology the substrate is not sticky.
For a sticky substrate that dissociates more slowly than
it reacts to form products (k
3
> k
2
; Eq. 9-54), the values
of pK
1E
will be lowered and pK
1E
of Eq. 9-53 will be
raised by log (1 + k
3
/k
2
).
65,66
In addition to the articles
by Cleland, other detailed treatments of pH effects
have been prepared by Brocklehurst and Dixon
69
and
Tipton and Dixon.
70
Fumarate hydratase (fumarase), which is dis-
cussed in Chapter 13, catalyzes the reversible hydra-
tion of fumaric acid to malic acid (Eq. 13-11). It was
one of the first enzymes whose pH dependence was
studied intensively. A bell-shaped pH dependence
for both forward and reverse reactions was observed
by Alberty and coworkers
63
and, using Eqs. 9-54 and
9-55, the two apparent pK
a
values were measured.
B. Inhibition and Activation of Enzymes
The action of most enzymes is inhibited by many
substances. Inhibition is often specific, and studies of
the relationship between inhibitor structure and activity
have been important to the development of our concepts
of active sites and of complementarity of surfaces of
biomolecules. Inhibition of enzymes is also the basis
of the action of a very large fraction of important drugs.
Inhibition may be reversible or irreversible, the latter
leading to permanent inactivation of the enzyme. Often,
but not always, irreversible inhibition is preceded by
reversible binding of the inhibitor at a complementary
site on the enzyme surface.
1. Competitive Inhibitors
Inhibitors with close structural similarities to a
substrate tend to bind to the substrate site. In truly
competitive inhibition, substrate and inhibitor not only
1 2 3 4 5 6 7 8 9 10
1
2
3
4
l
o
g
1
0
V
m
a
x
pK
1ES
, 4.2 pK
2ES
, 7.6
pK
2ES
0
0.8
1.6
2.4
3.2
4 5 6 7 8 9 10 11
pK
1E
pK
1ES
pH

l
o
g
1
0
K
m
pK
2E
pH
Figure 9-9 (A) Plot of log V
max
vs pH for a crystalline bacterial
-amylase. From Ono, et al.
68
(B) Theoretical curve of log
K
m
vs pH for Eq. 9-56 with pK
1E
= 5, pK
2E
= 10, pK
1ES
= 6, and
pK
2ES
= 7. Courtesy of C. Metzler.
A
B
B. Inhibition and Activation of Enzymes
472 Chapter 9. Enzymes: The Catalysts of Cells
compete for the same site but also their binding is
reversible and mutually exclusive. The affinity of the
inhibitor for the enzyme is expressed quantitatively
through the inhibition constant K
i
which is the disso-
ciation constant of the enzyme inhibitor complex EI :
K
i
= [E] [I] / [EI]
Using the steady state assumption for the mechanism
shown in Eq. 9-14, and writing a mass balance equation
that includes not only free enzyme and ES but also EI
we obtain an equation relating rate to substrate con-
centration. It is entirely analogous to Eq. 9-15 but K
m
is replaced by an apparent Michaelis constant, K
m
:
The relationships between v and [S] and between 1/ v
and 1/ [S] are as follows:
A commonly used test for competitive inhibition
is to plot 1/ v vs 1/ [S] (Eq. 9-61), both in the absence
of inhibitor and in the presence of one or more fixed
concentrations of I. The result, in each case, is a family
of lines of varying slope (Fig. 9-10) that converge on
one of the axes at the value 1/V
max
. We see that the
maximum velocity is unchanged by the presence of
inhibitor. If sufficient substrate is added, the enzyme
will be saturated with substrate and the inhibitor
cannot bind. The value of K
i
can be calculated using
Eq. 9-61 from the change in slope caused by addition
of inhibitor.
The effect of a fixed concentration of a competitive
inhibitor on a plot of v against log [S] (Fig. 9-11) is to
shift the curve to the right, i.e., toward higher values
of [S], but without any change in shape (or in the value
of V
max
).
Another plot, introduced by Dixon,
71
is that of 1/ v
versus [ I ] at two or more fixed substrate concentrations.
The student should be able to demonstrate that this
plot contains a family of straight lines that intersect
at a point to the left of the origin with coordinates
[I] = K
i
and 1/v = 1/V
max
. This plot may fail to distin-
guish certain types of inhibition discussed in the next
section.
72
Competitive inhibition is extremely common and
has great significance for metabolic control and for the
effects of drugs and of poisons. Simple ions are often
competitive inhibitors. Since many biochemical sub-
stances carry negative charges, anions such as Cl

, HCO
3

,
HPO
4
2
, and acetate

frequently act as competitive


inhibitors.
Two special classes of competitive inhibitors are
characterized by slow binding and slow, tight binding
to active sites.
73 77
Among the very tight-binding
inhibitors, which may also be slow to dissociate from
active sites, are transition state inhibitors discussed in
Section D,1.
(9-58)
(9-59)
(9-60)
(9-61)
V
max
v
V
max
/K
m
v
/
[
S
]V
max
/2K
m
without inhibitor
[I] = K
i
[I] = 2K
i
[I] = 2K
i
[I] = K
i
without inhibitor
1/V
max
1/K
m
1/[S]
1
/
v
Figure 9-10 Effect of a competitive inhibitor on the Eadie
Hofstee plot (top) and on a double reciprocal plot (bottom).
The apparent K
m
(Eq. 9-59) is increased by increasing [ I ], but
V
max
is unchanged.
Figure 9-11 Plots of v vs. log [S] for competitive and non-
competitive inhibition.
V
max
v
log
10
[S]
[I]/K
2
=[I]/K
1
= 1
Noncompetitive inhibition
Competitive
inhibition
Uninhibited
[I]/K
i
= 1
K
m
K
m

V
max
1
2
K K
K
m m
+
j
(
,
\
,
(
1
[ ] I
i
,
[ ]
max
S

V
K K
m m
, ,
v v
1 1
1 + +
j
(
,
\
,
(
V
K
V K
m
max max
[ ]
[ ]
S
I
i
v
473
2. Noncompetitive Inhibition and Activation;
Allosteric Sites
If an inhibitor binds not only to free enzyme but
also to the enzyme substrate complex ES, inhibition is
noncompetitive. In this case, S and I do not mutually
exclude each other and both can be bound to the en-
zyme at the same time. Why does such an inhibitor
slow an enzymatic reaction? In most instances, the
structure of the inhibitor does not show a close simi-
larity to that of substrate, which suggests that the
binding of inhibitors is at an allosteric site, that is,
at a site other than that of the substrate. The inhibition
of the enzyme may result from a distortion of the three-
dimensional structure of the enzyme which is caused
by the binding of the inhibitor. This distortion may be
transmitted to the active site even though the inhibitor
binds far from that site. In some cases two distinctly
different conformers of the protein may exist, one
binding substrate well and the other binding inhibitor
well (Fig. 9-12). In other instances the bound inhibitor
may interfere with the catalytic action by partially
overlapping the active site. In either case the ES com-
plex reacts to give product in a normal way, but the
ESI complex reacts more slowly or not at all.
Binding of a substance to an allosteric site some-
times has the effect of increasing the activity of an
enzyme rather than inhibiting it. This may occur
because the activator stabilizes the conformation
that binds substrate best (Fig. 9-12). The quantitative
treatment of such activation is similar to that of inhibi-
tion; allosteric inhibitors and activators are often con-
sidered together and are referred to as modifiers or
BOX 9-C THE SULFONAMIDES AS ANTIMETABOLITES
The development of the sulfa drugs,
a c
deriv-
atives of sulfanilamide, originated with studies of
the staining of protozoal parasites by Paul Erhlich.
In 1932 it was shown that the red dye 2,4-diamino-
azobenzene-4'-sulfonamide (Prontosil) dramatically
cured systemic infections by gram-positive bacteria.
Subsequent studies revealed that bacteria converted
the azo dye to sulfanilamide, a compound with
strong bacteriostatic activity; that is, it inhibited
bacterial growth without killing the bacteria. Al-
though sulfanilamide had been used in large quan-
tities since 1908 as an intermediate in synthesis of
dyes, its potential as an antibacterial agent had not
been recognized.
In 1935, D. D. Woods found that the growth
inhibition of sulfanilamide was reversed by yeast
extract.
d
From this source, in 1940 he isolated
p-aminobenzoic acid and demonstrated that the
inhibitory effect of 3 x 10
4
M sulfanilamide was
overcome by 6 x 10
8
Mp-aminobenzoate. The
relationship between the two compounds was
shown to be strictly competitive. If the sulfanil-
amide concentration was doubled, twice as much
p-aminobenzoate was required to reverse the
inhibition as before. These facts led to the formula-
tion by Woods and by P. Fildes (in 1940) of the
antimetabolite theory.
d,e
It was proposed that
p-aminobenzoate was needed by bacteria and that
sulfanilamide competed for a site on an enzyme
designed to act on p-aminobenzoate. We now know
that the idea was correct and that the enzyme on
which the competition occurs catalyzes the synthesis
of dihydropteroic acid (Fig. 25-19), a precursor to
folic acid.
While sulfanilamide itself is somewhat toxic, a
variety of related drugs of outstanding value have
been developed. Over 10,000 sulfonamides and
related compounds have been tested for antibacterial
action.
a
Bardos, T. J. (1974) Top. Curr. Chem. 52, 63 98
b
Gale, E. F., Cundliffe, E., Reynolds, P. E., Richmond, M. H., and
Waring, M. J. (1972) The Molecular Basis of Antibiotic Action,
Wiley, New York
c
Shepherd, R. G. (1970) in Medicinal Chemistry, 3rd ed.
(Burger, A., ed), pp. 255 304, Wiley (Interscience), New York
d
Woods, D. D. (1940) Brit. J. Exp. Pathol. 21, 74 90
e
Fildes, P. (1940) Lancet 1, 955 957
B. Inhibition and Activation of Enzymes
NH
2
H
2
N N
N SO
2
NH
2
Prontosil
N
H H
S
N
H R
N
H H
C
O O

R=H in sulfanilamide
Sulfonamides
p-Aminobenzoate
O O
474 Chapter 9. Enzymes: The Catalysts of Cells
inhibition cannot be reversed completely.
Figure 9-12 shows a plot of 1/ v against 1/ [S] at a
series of fixed values of [ I ]. For the case that K
1
= K
2
(classical noncompetitive inhibition), a family of
reciprocal plots that intersect on the horizontal axis at
a value of 1/K
m
is obtained. On the other hand, if K
1
and K
2
differ (the general case of noncompetitive
inhibition), the family of curves intersect at some other
point to the left of the vertical axis and, depending
upon the relative values of K
1
and K
2
, either above or
below the horizontal axis. The example illustrated is
for K
2
= 0.5K
1
; that is, for the binding of M to ES being
twice as strong as that to E.
Figure 9-11 shows inhibition data for both the
noncompetitive and the competitive cases plotted vs
log [S]. The shift of the midpoint to the right in each
case reflects the tendency of the inhibitor to exclude
the substrate from binding, while the lowered value of
the maximum velocity in the case of noncompetitive
inhibition results from the failure of the substrate to
completely displace the inhibitor from the enzyme
effectors. A general scheme
78,79
is given by Eq. 9-62.
Here K
1
and K
2
are equilibrium constants for dissocia-
tion of M from EM and ESM, respectively, while K
dS
and K
dS
are the dissociation constant of ES and of ESM
(to S and EM), respectively. Notice that, K
dS
is not
independent of the others (Eq. 9-63):
K
dS
= K
dS
K
2
/K
1
We have already considered competitive inhibition
which is obtained when K
2
= 0 (and therefore K
dS
= 0).
For this case, M is always an inhibitor and no activation
is possible. Notice that the inhibition will appear
competitive even if M binds at an allosteric site as in
Fig. 9-13 or if the inhibited form does not react at all
with substrate. Noncompetitive inhibition will be
observed if ESM is formed but does not react, i.e., if
k
4
= 0. Then the rate equation in reciprocal form will
be given by Eq. 9-64.
We see that 1/ V
max
is multiplied by a term containing
[ I ] and K
2
. It is a characteristic of noncompetitive
inhibition that the maximum velocity is decreased
from that observed in the absence of the inhibitor.
Also, no matter how high the substrate concentration,
M (modifier)
K
1
EM + S
+
M
K
2
ESM
+
product
K
ds
E + S ES product
K
ds k
3
k
4
(9-62)
(9-63)
1 1
1 1
2 1
v V K
K
V K
m
+
j
(
,
\
,
(
+ +
j
(
,
\
,
(
max max
[ ]
[ ]
[ ] I
S
I
(9-64)
max
2/V
max
1/K
m
1/[S]
1
/
v
[I]/K
2
=[I]/K
1
= 1
0 1/K
m
uninhibited
1/V
max
[I]/K
2
=0.5, [I]/K
1
= 1
Figure 9-12 Double reciprocal plots for two cases of non-
competitive inhibition.
Figure 9-13 (A) An enzyme with binding sites for allosteric
inhibitor I and activator J. Conformer A binds inhibitor I
strongly but has little affinity for activator J or for substrate
S. Conformer B binds S and catalyzes its reaction. It also
binds activator J whose presence tends to lock the enzyme
in the on conformation B. Conformers A and B are desig-
nated T and R in the MWC model of Monod, Wyman, and
Changeux.
80
(B) Inhibited and activated dimeric enzymes.
Conformer A (or T)
Activator
Conformer B (or R)
J
K
t
I
Inhibited dimer
Activated dimer in proper
conformation to bind
substrate at both sides
S
S
I
J
S
A
B
475
even as [S] becomes very high. If an inhibitor binds
only to ES and not to E, i.e., K
1
= 0, a family of parallel
double-reciprocal plots of 1/v vs 1/[S] will be obtained.
This case is referred to as uncompetitive inhibition.
Multisubstrate enzymes with either ordered sequential
or ping-pong mechanisms also often give parallel line
plots with inhibitors.
Uncompetitive inhibitors of liver alcohol dehydro-
genase (Chapter 15) could be used to treat cases of
poisoning by methanol or ethylene glycol.
81 83
The
aim is to prevent rapid oxidation to the toxic acids
HCOOH and HOCH
2
COOH, which lower blood pH,
while the alcohols are excreted. Uncompetitive inhibi-
tors have an advantage over competitive inhibitors as
therapeutic agents in that the inhibition is not over-
come when the substrate concentration is saturating.
84
If rate constant k
4
of Eq. 9-62 is not zero we may
observe either inhibition or activation. If k
3
= k
4
the
effect of a modifier will be to alter the apparent K
m
by
either increasing it (inhibition) or decreasing it (activa-
tion). The maximum velocity will be unchanged.
Monod et al.
80
referred to enzymes showing such
behavior as K systems. On the other hand, if K
1
= K
2
and k
3
differs from k
4
we have a purely V system. In
the general case a modifier affects both the apparent
K
m
and V
max
. Although in the foregoing discussion it
has been assumed that activators bind at allosteric sites,
as was pointed out in Section A,11, ions and other
small molecules (substrate surrogates) that act as
competitive inhibitors at high concentrations may be
activators at low concentrations.
40
Activation of enzymes by specific metallic ions is
often observed. In many instances the metallic ion is
properly regarded as a second substrate which must
bind along with the first substrate before reaction can
occur. Alternatively, the complex of the organic sub-
strate with the metal ion can be considered the true
substrate. Thus, many enzymes act upon the magne-
sium complex of ATP (Chapter 12). The enzymes can
either be regarded as three-substrate enzymes requiring
Mg
2+
+ ATP
4
and another substrate or as two-substrate
enzymes acting upon ATPMg
2
and a second substrate.
3. Inhibitors in the Study of Mechanisms
A substrate analog will frequently inhibit only one
of the two forms of a multisubstrate enzyme with a
ping-pong mechanism.
1,72
Reciprocal plots made for
various inhibitor concentrations consist of a family of
parallel lines reminiscent of uncompetitive inhibition.
Observation of such parallel line plots can support a
ping-pong mechanism for an enzyme but cannot prove
it because in some cases parallel lines are observed for
inhibition of enzymes acting by an ordered sequential
mechanism. The following question arises naturally
for any ordered bimolecular reaction (Eq. 9-43): Of the
two substrates required by the enzymes, which one
binds to the enzyme first? If the concentration of one
substrate is kept constant while varying concentrations
of an inhibitory analog of that substrate are added and
1/ v is plotted against the reciprocal of the concentration
of the other substrate, parallel lines are obtained if,
and only if, the substrate of fixed concentration is B,
the substrate is adding second in the binding sequence,
and if I is its analog. The substrate binding first (A) is
the one whose concentration was varied in the experi-
ment.
Product inhibition (Section A,12) can also provide
information about mechanisms. For example, if 1/ v is
plotted against 1/ [A] in the presence and absence of
the product Q, the product will be found to compete
with A and to give a typical family of lines for compet-
itive inhibition. On the other hand, a plot of 1/ v vs
1/ [B] in the presence and absence of Q will indicate
noncompetitive inhibition if the binding of substrates
is ordered (Eq. 9-43). In other words, only the AQ
pair of substrates are competitive. Product inhibition
is also observed with enzymes having ping-pong
kinetics (Eq. 9-47) as a result of formation of nonpro-
ductive complexes.
4. Allosteric Effectors in the Regulation of
Enzyme Activity
The binding of a substance at an allosteric site
with the induction of a conformational change forms
the basis for many aspects of regulation. The term
allostery (allosterism) usually refers to the effects of
allosteric modifiers, which may be either inhibitors or
activators, on oligomeric enzymes. However, as we
have already seen (Eq. 9-62), monomeric enzymes may
also be subject to allosteric regulation by modifiers.
Consider a monomer that contains binding sites for
substrate, inhibitor, and activator and which exists in
conformations A and B as in Fig. 9-13. Let us assume
(see Eq. 7-31) that conformer B binds both substrate
and activator well but that it binds inhibitor poorly or
not at all. On the other hand, A binds inhibitor well
but binds substrate and activator poorly. This simple
combination of two conformers with different binding
properties provides a means by which enzymes can be
turned on or off in response to changing conditions.
If an inhibitory substance builds up to a high
concentration within a cell, it binds to conformer A; if
the inhibitor concentration is high enough, virtually
all of the enzyme will be locked in the inactive confor-
mation A. The enzyme will be turned off or at least
reduced to a low activity. On the other hand, in the
presence of a high concentration of activator the en-
zyme will be turned on because it is locked in the B
conformation. The relative concentrations of inhibitor,
activator, and substrate within a cell at any given time
B. Inhibition and Activation of Enzymes
476 Chapter 9. Enzymes: The Catalysts of Cells
According to the MWC model, in the presence of
inhibitor and activator at normalized concentrations
and an enzyme will still follow Eq. 9-65, but the
allosteric constant L will be replaced by an apparent
allosteric constant L (Eq. 9-70).
86
Figure 9-14 shows
plots of Y vs. log for two different values of L for a
tetramer with a specific value assumed for c. In both
cases, Y approaches 1 as log increases, but since we
are dealing with noncompetitive inhibition at high
values of L, much of the enzyme will be in the T(A)
conformation at saturation.
Noncompetitive inhibition cannot be completely
reversed by very high substrate concentrations. Monod
et al. defined for an allosteric enzyme a function of
state R (Eq. 9-71) which is the fraction of total enzyme
in the (B) conformation:
In a K system (Section B,2) it is the value of that
determines the velocity with which an enzyme reacts.
Figure 9-14 also shows as a function of log []. Note
that when L is low does not approach zero even
when [S] 0. In other words, the enzyme is never
completely turned off, just as when L is high the
enzyme is never completely turned on.
Figure 9-14 may be compared with Fig. 9-11 which
shows similar curves for noncompetitive inhibition
of a monomeric enzyme. The significant difference
between the two figures is that saturation of the oligo-
meric enzyme occurs over a narrower concentration
range than does that of the monomer, i.e., saturation
of the oligomeric enzyme, especially in the presence of
inhibitor, is cooperative. Note that cooperative binding
of substrate requires that the free enzyme be largely
in conformation T (A), as it is in the presence of an
inhibitor. Allosteric interactions between two iden-
tical molecules, whether of substrate or of effector, are
described by Monod et al.
80
as homotropic interactions.
Such interactions lead to cooperativity or anticooperat-
ivity in binding. Allosteric interactions between two
different molecules, e.g., a substrate and an activator
are designated heterotropic.
For many enzymes the MWC model is unrealisti-
cally simple. The more general treatment of binding
equilibria given in Chapter 7 may be applicable. How-
ever, in addition to K systems there are V systems in
which a conformational change alters the maximum
velocity (see Eq. 9-62)
87
and sometimes both substrate
will determine what fraction of the enzyme is in active
conformation B. It is this interplay of inhibitory and
activating effects that provides the basis for much of
the regulation of cell chemistry.
80,85
The effects of inhibitors or activators on the kinetics
of the monomeric enzyme of Fig. 9-13 can be described
by Eq. 9-62 to 9-64. Separate terms for both inhibition
and activation can be included. The equilibrium between
the two conformers can also be indicated explicitly
according to Eq. 7-30. However, for monomeric en-
zymes it is usually not profitable to try to separate the
two constants K
t
and K
BX
which describe the conforma-
tional change and binding of substrate or activator,
respectively, in Eq. 7-30.
Most intracellular enzymes are oligomeric, and
the binding of allosteric effectors leads to additional
interesting effects. Binding constants or dissociation
constants must be defined for both inhibitor and acti-
vator to both conformers A and B. Since all species
must be taken into account in the mass balance, the
equations are complex. However, the MonodWyman
Changeux (MWC) model (Chapter 7) gives a relatively
simple picture. The saturation curve for an oligomeric
enzyme following this model may be derived from Eq.
7-39 and is given by the following expression:
Here, L is the allosteric constant which is given (for
a dimer) by Eq. 7-36. The constant c is the ratio of
dissociation constants K
BS
and K
AS
for the two conformers:
c = K
BS
/K
AS
Notice that in this chapter dissociation constants (K
d
) of
ES complexes are being used, whereas the equations of
Chapter 7 are all written in terms of association constant
(K
f
). The parameter is defined as follows, where K
BS
equals 1/ K
BX
of Chapter 7.
= [S] / K
BS
)
To take account of effects of inhibitor and activator the
ratios of dissociation constants of I from BI and AI and
of activator J from BJ and AJ are defined as in Eq. 9-68.
Likewise, normalized concentrations of I and J are
defined (Eq. 9-69) as and , respectively.
Y
+ + +
+ + +

Lc c
L c
n n
n n


( ) ( )
( ) ( )
1 1
1 1
1 1
(9-65)
(9-67)
d
K
K
e
K
K
K K
> <

BI
AI
BJ
AJ
AI BJ
I J
1 1
[ ] [ ]
(9-68)
(9-69)

+ +
+ +
,

,
]
]
]
L L
d e
n
( )( )
( )( )
1 1
1 1

(9-70)
R
L c
n
n n

+
+ + +

( )
( ) ( )
1
1 1


function of state (9-71)
R
R
R
(9-66)
R
477
Figure 9-14 Fractional saturation Y and function of state
for hypothetical tetrameric enzymes following the MWC
model. Curves are calculated for two different values of the
apparent allosteric constant L (Eq. 9-70) and for c = 0.1 (Eq.
9-66). After Rubin and Changeux.
86
affinity and maximum velocity.
The fact that data can be fitted to an equation is not
proof that a mathematical model is correct; other models
may predict the results just as well. For example, Jenkins
has shown that the presence of ions that act as substrate
surrogates (Section A,11) can produce cooperative or
anticooperative binding curves for substrates.
40,88
Rabin
89
suggested the following explanation of coop-
erativity for a monomomeric enzyme with a single
substrate-binding site. In its active conformation E the
enzyme reacts with substrate rapidly and the ES com-
plex yields product rapidly as in the upper loop of Eq.
9-72. However, a slow conformational change inter-
converts E and E, a less active form with much lower
affinity for substrate.
At the same time, the complex ES, if formed, can
equilibriate with ES and thereby alter the conforma-
tional state of the protein. At low substrate concentra-
tions E will predominate and the enzymatic activity
will be low. At high substrate concentrations most E
will exist as ES and after release of product will tend
to remain in the active conformation long enough to
bind and act on another substrate molecule. This and
other kinetic models of cooperativity are discussed by
Newsholme and Start.
90
L = 4
c = 0.1
n = 4
R
Y
R
L = 10
4
0.0
0.4
0.8
1.2
0.0
0.4
0.8
1.2
2 1 0 1 2 3 4
log
10
= log
10
[S] log
10
K
BS
Y

o
r

R
Y

o
r

R
Y
The physiological significance of cooperative
binding of substrates to enzymes is analogous to that
of cooperative binding of oxygen by hemoglobin which
provides for more efficient release of oxygen to tissues.
However, in the presence of excess activator an enzyme
is locked in the R (B) conformation and no cooperativity
is seen in the binding of substrate. In this case, each
binding site behaves independently. On the other
hand, there will be strong cooperativity in the binding of
the activator. The result is that control of the enzyme is
sensitive to a higher power than the first of the activator
concentration. Likewise, the turning off of the enzyme
is more sensitive to inhibitor concentration as a result
of cooperative binding of the latter. It seems likely
that the evolution of oligomeric enzymes is at least
partly a result of the greater efficiency of control mech-
anisms based on cooperative binding of effectors.
5. Irreversible Inhibition of Enzymes
Some of our most effective drugs, such as penicillin
(Box 20-G) are irreversible inhibitors of specific enzymes.
Such inhibitors are also of practical importance to bio-
chemists who wish to inhibit specific enzymes such as
proteases that might otherwise destroy proteins they
are studying. Transition state analogs (Section E,1)
and slow-binding inhibitors (Section B,1) may appear
to inactivate an enzyme irreversibly because they
bind tightly and dissociate slowly. However, true
irreversible inhibitors, such as oxidizing agents or
alkylating agents, cause a more permanent chemical
modification, usually at or near the active site. Two
groups of irreversible inhibitors are of special interest:
affinity labeling agents and enzyme-activated inhibitors.
Affinity labeling agents (active-site directed
inactivating reagents) have two essential properties:
a high affinity for the active site of the specific enzyme to
be inhibited and the presence of a chemically highly reactive
group which can attack a functional group in a protein.
A good example is provided by derivatives of chloro-
methylketones, which are reactive alkylating agents.
One of these, N-tosylphenylalanine chloromethyl
ketone (TPCK), is a potent inhibitor of chymotrypsin.
In addition to the chloromethyl ketone group it contains
(9-72)
B. Inhibition and Activation of Enzymes
R
S ES E + P
S ES
E (inactive)
slow
CH
2
C C CH
2
Cl
O H
H
3
C S NH
O
O
Y Enzyme
l-N-tosylphenylalanine chloromethylketone (TPCK)
478 Chapter 9. Enzymes: The Catalysts of Cells
the phenylalanine side chain, which helps direct it
into the substrate binding site of chymotrypsin, and
a sulfonamide linkage that mimics the normal amide
linkages of the substrate. The group Y, which becomes
alkylated in an S
N
2-like displacement of a chloride ion
(see Eq. 9-76) is thought to be His 57 of the active site
(see Fig. 12-10). For additional discussion see Walsh,
91
Kyte,
67
and Plapp.
91a
Enzyme-activated inhibitors (also called suicide
substrates, k
cat
inhibitors, or mechanism-based inhibi-
tors) are chemically inert until they enter the active
site of their target enzymes. Then, by passing through
at least some of the normal stages of the catalytic action
of the enzyme, they are converted to reactive inter-
mediates that can become irreversibly bound to an
enzyme.
92,93
For example, a halogen atom (F, Cl, Br)
together with a proton may be eliminated from an
intermediate to give an unsaturated compound to
which a nucleophilic side chain of the protein adds.
Several of these inhibitors are discussed in later chap-
ters. Because of their high specificity many enzyme-
activated inhibitors are potential drugs. One of them,
-difluoromethylornithine (Box 14-C), is said to bind
covalently to only one protein in the body of a rat, namely,
ornithine decarboxylase, the target enzyme for the drug.
C. The Specificity of Enzymatic Action
Enzymes are usually impressively specific in their
action. The specificity toward substrate is sometimes
almost absolute. For many years urea was believed to
be the only substrate for the enzyme urease and succi-
nate the only substrate for succinate dehydrogenase.
Even after much searching for other substrates, only
one or two closely related compounds could be found
that were acted on at all. In other cases enzymes can
use a class of compounds as substrates. For example,
the D-amino acid oxidase of kidney oxidizes a variety
of D-amino acids but does not touch L-amino acids.
Almost as impressive as the substrate specificity of
enzymes is the specificity for a given type of reaction.
Many substrates are capable of undergoing a variety
of different chemical reactions, either unimolecular or
with water or some other compound present in the
cell. The enzyme catalyzes only one of these reactions.
Although side reactions may occur to a small extent,
the most impressive thing in comparing an enzyme-
catalyzed reaction with an uncatalyzed organic reaction
is that the latter often produces large amounts of side
reaction products, but the enzymatic reaction does not.
1. Complementarity of Substrate and Enzyme
Surfaces
Impressed by the specificity of enzymatic action,
biochemists early adopted a lock-and-key theory
which stated that for a reaction to occur the substrate
must fit into an active site precisely. Modern experi-
ments have amply verified the idea. A vast amount
of kinetic data on families of substrates and related
competitive inhibitors support the idea and numerous
X-ray structures of enzymes with bound inhibitors or
with very slow substrates have given visual evidence
of the reality of the lock-and-key concept. Directed
mutation of genes of many enzymes of known three-
dimensional structure has provided additional proof.
As anticipated, hydrophobic groups of substrates
or inhibitors usually contact hydrophobic regions of
the protein and the fit is tight. For example, the active
site of chymotrypsin contains a hydrophobic pocket
designed to hold a large hydrophobic side chain, thus
providing the specificity observed with this enzyme
(Table 3-2). Likewise, polar groups of substrates con-
tact polar groups of the enzyme. The interactions are
complementary, positive charges fitting against nega-
tive and with correctly formed hydrogen bonds. Trypsin,
whose structure is similar to that of chymotrypsin,
exerts its specificity for a positively charged side chain
next to the bond cleaved (Table 3-2) by virtue of the
presence of a negatively charged carboxylate at the
bottom of the hydrophobic pocket. Several C = O and
N H groups of the peptide linkages in the substrate
form hydrogen bonds to the edge of a sheet in the
protein, in effect making the substrate an added strand
in the sheet (Fig. 12-10). Aspartate aminotransferase
acts on the dicarboxylic amino acids glutamate and
aspartate. A pair of arginine side chains bind the two
carboxylates of the substrate while the NH
3
+
of the
substrate is attracted to a negatively charged group
in the coenzyme pyridoxal phosphate, which is also
present in the active site (Eq. 14-39, Fig. 14-10).
2. Stereospecificity and Prochiral Centers
Most enzymes possess an infallible ability to
recognize the difference between the right side and the
left side of an organic substrate even when the latter
has perfect bilateral symmetry. In fact, this ability is
limited to prochiral centers of molecules and is a
natural consequence of their reaction with the chiral
H
2
N
C
H
2
N
O CH
2
COO

Urea Succinate
One H can be replaced by Cl
with retention of substrate activity
and by CH
3
with partial
retention of activity
H
2
C
COO

One H can be replaced


by OH and some
substrate activity remains
479
enzyme.
94 98
Consider carbon atom number 1 of ethanol.
The two attached hydrogen atoms are chemically
identical and would react identically with a nonchiral
reagent. Nevertheless, these atoms are no more equi-
valent stereochemically than are your right and your
left arms. We say that the molecule has a prochiral
center at C-1. A prochiral center on a tetrahedrally
bonded carbon atom always contains two identical
atoms or groups but a total of three different kinds of
atoms or groups. If the priority of this H is elevated,
e.g., by substitution of
2
Hfor
1
H the configuration
would be R (the priority of this
2
H would become c).
The two hydrogen atoms on C-1 of ethanol are
called enantiotopic because replacement of one or
the other by a fourth different kind of atom or group
would produce a pair of enantiomers (page 41). This
fact suggests a way of naming the positions occupied
by two enantiotopic atoms or groups. We first assign
priorities to all of the atoms or groups attached to the
central carbon atom according to the RS system. Now,
we ask whether the configuration will be R or S when
the priority of one of the two identical groups is raised,
e.g., by substitution of one of the hydrogen atoms by
deuterium. If the configuration becomes R, that group
occupies a pro-R position; if the configuration becomes
S, it occupies a pro-S position. Referring to the preced-
ing diagram, it is easy to see (by viewing down the
bond to the group of lowest priority and applying the
usual rule for determining configuration) that if the
pro-R hydrogen (H
R
) is replaced by deuterium (
2
H), the
configuration will be R. Conversely, replacement of
H
S
by deuterium will lead to the S configuration.
When ethanol is oxidized by the action of alcohol
dehydrogenase (Eq. 9-73), only the pro-R hydrogen
atom is removed. If the reaction is reversed in such a
way that deuterium is introduced into ethanol from the
reduced coenzyme the optically active R-2-deuterio-
ethanol is formed. The ability of an enzyme to
recognize a single hydrogen of a pair of hydrogens
on a CH
2
group was at first a surprise to many bio-
chemists.
99
However, it is a natural result of the com-
plementarity of enzyme and substrate surfaces, just as
the fit of a shoe is determined by the complementarity
of surfaces of foot and shoe. Only a chiral catalyst can
have this ability.
Another example is provided by malic acid, a
chiral molecule which also contains a prochiral center
(see Eq. 9-74). In this case replacement of the pro-R or
pro-S hydrogen atom by another atom or group would
yield a pair of diastereoisomers rather than enantiomers.
Therefore, these hydrogen atoms are diastereotopic.
When L-malic acid is dehydrated by fumarate hydratase
(Chapter 13) the hydrogen in the pro-R position is
removed but that in the pro-S position is not touched.
This can be demonstrated by allowing the dehydration
product, fumarate, to be hydrated to malate in
2
H
2
O
(Eq. 9-74). The malate formed contains deuterium in
the pro-R position. If this malate is now isolated and
placed with another portion of enzyme in H
2
O, the
deuterium is removed cleanly. The fumarate pro-
duced contains no deuterium.
Inhibition of enzymes provides the basis for
many of the effects of antibiotics and other chemo-
therapeutic substances (e.g., see Box 9-B). How-
ever, some drugs act on cell surface receptors
which have ordinarily not been regarded as
enzymes. According to receptor theory, developed
around 1937, structurally similar drugs often elicit
similar responses because they bind to the same
receptor. A receptor may normally bind a hormone,
neurotransmitter, or other metabolite whose
geometry is partially shared by a drug. Binding
of drugs of one class, termed agonists in the
pharmacological literature, to an appropriate
receptor triggers a response in a cell, similar to
that of a hormone. On the other hand, compounds
of related structure often act as antagonists,
binding to receptor but failing to elicit a response.
Agonist and antagonist often act in a strictly
competitive fashion as in competitive inhibition
of enzyme action.
We know now that many receptors are en-
zymes, some of which may act quite slowly.
The active site may sometimes be far from the
receptor binding site and sometimes in a separate
subunit. The receptor can be viewed as an allos-
teric effector which binds at a distant site or as
a ligand for a regulatory subunit of the enzyme
complex. Alternatively, the active site may be
viewed as the site for relaying a signal received
from the hormone or other agonist.
BOX 9-D RECEPTORS, AGONISTS, AND
ANTAGONISTS
C. The Specificity of Enzymatic Action
H O C
CH
3
H
R
H
S
proS
proR
a
b
H O C
CH
3
H
R
H
S
O C
CH
3
H
+ 2[H] (removed to a coenzyme)
(9-73)
480 Chapter 9. Enzymes: The Catalysts of Cells
(9-75)
Trigonal prochiral centers. Planar trigonal atoms,
such as those of aldehydes, ketones, and alkenes, are
also prochiral if they are attached to three different
kinds of atoms or groups. The two faces are enantio-
topic or diastereotopic, if another chiral center is present.
Hanson
94,94a
proposed that the faces be named as indi-
cated in Eq. 9-75. The trigonal atom is viewed from one
side and the three groups surrounding the carbon atom
are given priorities, a, b, and c, as in the RS system
(Chapter 2). If the sequence a, b, and c of priorites is
clockwise, the face toward the reader is re (rectus); if
counterclockwise, it is si (sinister). Priorities can often be
assigned on the basis of the atomic numbers of the first
atoms in the three groups (as in the example shown)
but, if necessary, replica atoms must be added as
described in Chapter 2. Replica atoms attached to
an originally trigonal carbon are ignored completely,
but those attached to O or N of C = O or C = N may be
required to establish the priorities of the groups around
the carbon atom. For C = O and C = N the faces of the
O and N atoms are taken to be the same as that of the
attached trigonal carbon atom. In C = C the two carbons
may have their re faces either on the same side or on
opposite sides of the group.
Notice that addition of an atom of
2
H to the re face
of acetaldehyde would give R-deuteroethanol (Eq. 9-75;
reverse of Eq. 9-73). This is the reaction catalyzed by
alcohol dehydrogenase. Addition of
1
H to the re face
places the entering hydrogen in the pro-R position.
Addition to the si face would place it in the pro-S
HO C
CH
3
H
R
H
O C
H
CH
3
a
b
c
+ 2[H]
re (or Re) face is toward the reader
si (or Si) face is behind the page
Stereochemical numbering. In some prochiral
molecules, such as glycerol, the two ends of the main
carbon chain form identical groups. Since the two
ends are distinguishable to an enzyme, it is important
to decide which should be labeled C-1 and which C-3.
Hirschmann proposed a stereochemical numbering
system
100
in which the carbons are numbered beginning
with the end of the chain that occupies the pro-S position.
In this numbering system derivatives of the parent
prochiral compound are given the prefix sn-. Thus,
glycerol phosphate, used by cells to construct phos-
pholipids, usually bears a phosphate group on the
CH
2
OH in the pro-R position of glycerol and is
therefore sn-3-glycerol phosphate.
The citrate ion, a very important prochiral meta-
bolic intermediate, has three prochiral centers at C-2,
C-3, and C-4, respectively. That at C-3 distinguishes
the pro-R and pro-S arms and determines the stereo-
chemical numbering. Citrate containing
14
C in the sn-1
position is called sn-citrate[1-
14
C] and is the form of
labeled citrate that is synthesized in living cells from
oxaloacetate and [1-
14
C]acetyl coenzyme A (see Fig.
10-6). The first step in the further metabolism of citrate
is the elimination of the OH group from C-3 together
with the H
R
proton from C-4 through the action of the
enzyme aconitate hydratase (aconitase). In this case
the proton at C-4 (in the pro-R arm) is selected rather
than that at C-2.
C C
COO

OOC
HO
H
S
H
R
H
C C
COO

OOC
H
H
from H
S
of
L-Malate
Fumarate
H
2
O
L-Malate
+
(9-74)
HO C
C
C HO
H
H
OH
H
H
H
pro-S
pro-R
1
2
3
Stereochemical numbering for a
symmetric prochiral molecule.
Carbons are numbered begin-
ning with the end of the chain
that occupies the pro-S position.
C
H
R
H
S
COO

C
C COO

OOC
HO
H
S
H
R
COO

C
1
C
H
S
H
R
HO COO

C H
R
H
S
COO

2
4 5
proS arm
proR arm
1
2
3
4
5
Citrate
Citrate ion numbered
according to the
stereochemical system
Drawn using the
Fisher convention
481
position. Fumarate has two trigonal carbon atoms.
The si faces of both are toward the viewer in the fol-
lowing structure (also shown in Eq. 9-74). Referring
to Eq. 9-74, we see that an HO

ion from water adds


to the si face of one carbon atom of fumarate to give
s-malate (L-malate). At the same time, a proton com-
bines with the re face of the adjacent unsaturated
carbon atom to enter the pro-R position.
3. Induced Fit and Conformational Changes
Results of many X-ray studies indicate that the
lock-and-key picture of enzyme action must be modi-
fied. If an enzyme is a lock and the substrate the
key, the entrance of the key into the lock often induces a
conformational change in the protein. Binding of substrates
may be imperfect in the initially formed ES complex
but may be more nearly perfect a few nanoseconds or
microseconds later as the protein readjusts its structure
to accommodate the substrate. The substrate has in-
duced a fit. For example, when an amino acid substrate
binds to aspartate aminotransferase one whole domain
of the enzyme moves inward, packing hydrophobic
side chains of the protein against the substrate (Chap-
ter 14). This strengthens the electrostatic interactions
between the ion pairs that orient the substrate and
align it for reaction. Similar conformational changes
have been observed for citrate synthase, glycogen
phosphorylase, various kinases (Chapter 12), alcohol
dehydrogenase (Chapter 15), and a growing list of
other enzymes. Accompanying changes in circular
dichroism, ultraviolet spectra, and sedimentation
constants are often observed.
For many other enzymes the observed conforma-
tional changes are subtle. A single loop of polypeptide
chain or even of a side chain moves to cover the bound
substrate. With trypsin and other serine proteases a
flexible segment of the peptide chain becomes immo-
bilized and forms more hydrogen bonds after substrate
binds than before. It is probably quite unusual for a
major unfolding and refolding of parts of a protein to
take place. The term induced fit usually refers to
substrate binding, but as substrates are interconverted
within active sites successive changes in geometry and
in charge distribution occur. Small conformational
changes may be required at several stages of an enzy-
matic reaction to ensure that complementarity of
substrate and enzyme is preserved.
101
In line with this
idea are the facts that proteins are less tightly packed
C C
COO

H
H

OOC
a
b
c
b
c
a
Fumarate
The si faces of both trigonal
carbon atoms are toward
the viewer.
at active sites than in other parts of the molecules, and
that active sites often lie between domains and usually
are formed by several loops of the peptide chain
(Chapter 2).
102
Although conformational changes allow proteins
to maintain good complementarity with substrates, it
does not follow that substrates are therefore bound very
tightly. This is easiest to understand for reversible
reactions in which a substrate is the product of the
reverse reaction. Not only binding but also the rate
of release of product must be rapid. Very tight binding
would retard release and cause product inhibition.
Furthermore, as we can see from Eq. 9-37, the velocity
of catalysis is the product of [S][E] x k
cat
/K
m
, where [E]
is, free enzyme. Catalysis can be made more efficient in
two ways: by increasing k
cat
or by decreasing K
m
, that
is, by tighter binding. However, tighter binding will be
advantageous only if K
m
is not so low that the enzyme
is approaching saturation with substrate. Otherwise,
[E] will fall as K
m
is lowered and little increase in rate
will be observed.
2
In fact, it appears that enzymes have evolved to
have high values of k
cat
and high values of K
m
, that is,
weak binding of substrates.
2
Frequently, in one confor-
mational state of a protein the active site is open, with
solvent molecules or substrate surrogates present,
while in a second state of nearly equal energy it is
closed around the substrate. The functional groups
of the enzymes active site can be bound either to
external ions or to ionic groups of substrates and either
to water or to hydrogen-bonding groups of substrates.
In some cases two ionic groups in the protein may pair
with each other in the open conformation and with
ionic groups of the substrate in the closed conformation.
Thus, the energy changes accompanying the confor-
mational changes can be small but very good comple-
mentarity can exist in the ES complex, an important
factor in establishing specificity.
4. Specificity and k
cat
Enzyme specificity is often observed not only in
binding but also in the rate at which ES is converted to
products. Thus, it is the values of k
cat
/ K
m
that determine
specificity. Good examples are provided by chymo-
trypsin and related serine proteases (Chapter 12),
2
for
which substrates with the shortest chains are often
bound as well as those with longer chains but react
more slowly. For example, N-acetylphenylalanine amide
binds to chymotrypsin about as well as does the longer
N-acetylphenylalanylalanine amide but reacts only 1/47
as fast.
2
One might anticipate that increasing the length
of the substrate would make it bind more tightly
because of the greater number of contacts between
substrate and enzyme. It has often been suggested
that the reason that this does not happen is that the
C. The Specificity of Enzymatic Action
482 Chapter 9. Enzymes: The Catalysts of Cells
binding of the longer, more specific substrates distorts
the enzyme and that the binding energy is now stored in
the enzyme. It is as if the enzyme contained an internal
spring which would be compressed when the substrate binds.
This would keep the binding weak but the energy in
the spring might then be used to increase the velocity.
5. Proofreading
Although enzymes may be very specific they do
make mistakes. This is of particular concern for pro-
cesses such as protein synthesis in which the correct
amino acid is placed at each position in the sequence
with an error rate that has been estimated for E. coli as
only 1 in 10
4
. It would be impossible for an enzyme
designed to attach valine to its specific transfer RNA
to avoid attaching the smaller alanine if discrimination
between the two were based solely on the Gibbs energy
differences of binding.
103
However, it would be easier
for an enzyme to exclude larger amino acids. This prob-
lem may be resolved by use of multistep screening.
104,105
For example, isoleucyl-tRNA synthetase (Chapter 29)
does occasionally attach the smaller valine to the
specific tRNA,
Ile
but when it does the enzyme in a
proofreading and editing step hydrolyzes off the
incorrect amino acid. The active site for this hydro-
lyzing activity, whether at a different place on the
enzyme surface or created by a conformational
change, may be able to exclude sterically the larger
isoleucyl residue while acting on the valyl-tRNA.
This editing mechanism for isoleucyl-tRNA synthetase
was demonstrated directly in 1998 by X-ray crystallo-
graphy on complexes of the enzyme with L-isoleucine
and L-valine. Both substrates fit into the ATP-requir-
ing synthetic site but neither isoleucine nor isoleucyl-
tRNA will fit into the editing site which is located in
an adjacent -barrel domain.
104,105
Proofreading steps
based on differing chemical properties as well as size
can also be visualized.
103,106
D. Mechanisms of Catalysis
Kinetic studies tell how fast enzymes act but by
themselves say nothing about how enzymes catalyze
reactions. They do not give the chemical mechanism
of catalysis, the step-by-step process by which a
reaction takes place. Most of the individual steps
involve the simultaneous breaking of a chemical bond
and formation of a new bond. Consider a simple
displacement reaction, that of a hydroxyl ion reacting
with methyl iodide to give the products methanol and
iodide ions.
The reaction can be thought of as an attack by the
OH

ion on the back side of the carbon atom of the


methyl groups with a simultaneous displacement of
the I

.
1. The Transition State
A reaction such as that of Eq. 9-76 is not instantan-
eous, and at some time between that at which the reac-
tants exist and that at which products have been formed
the C I bond will be stretched and partially broken
and the new C O bond will be partially formed. The
structure at this point is not that of an ordinary com-
pound and is energetically unstable with respect to both
the reactant and the products. The intermediate struc-
ture of the least stability is known as the transition
state. Although no one has actually seen a transition
state structure, we might represent that for Eq. 9-76 as
follows:
The negative charge is distributed between the attack-
ing HO

group and the departing iodide. The bonds


to the central carbon atom are no longer tetrahedrally
arranged but the C H bonds lie in a single plane and
the partial bonds to the OH and iodine atom lie at
right angles to that plane.
It is useful in discussing a reaction mechanism to
construct a transition state diagram in which Gibbs
energy G is plotted against reaction coordinate (Fig.
9-15A). Energy E or enthalpy H may be plotted in the
same way and authors frequently do not state whether
G, E, or H is meant. The reaction coordinate is usually
not assigned an exact physical meaning but represents
the progress from reactants toward products. It is
directly related to the extent to which an existing bond
has been stretched and broken or a new one formed.
The high energy point is the transition state. A some-
what more detailed idea of a transition state is obtained
from a contour diagram such as that of Fig. 9-15B. Here,
energy is plotted as a function of two distances, e.g.,
the lengthening C I bond distance and the shortening
C O distance for Eq. 9-76. The path of minimum
energy across the saddle point representing the
transition state is indicated by the dashed line.
In a reaction coordinate diagram the difference
in value of G between reactants and products is the
H
C
H H
I HO CH
3
+ I
HO

(9-76)
HO I
H
H
H

Transition state structure


C
483
overall Gibbs energy change G for the reaction, while
the difference in G between the transition state and
reactants is G

, the Gibbs energy of activation. The


magnitude of G

represents the energy barrier to a


reaction and largely determines the rate constant.
The diagrams in Fig. 9-15 are too simple because
enzymatic reactions usually occur in several interme-
diate steps. There will be transition states for each
step with valleys in between. The valleys correspond
to intermediate species, which are sometimes very
unstable. The passing from reactants to products in
an enzymatic reaction can be likened to wandering
through a series of mountain ranges of various heights
and finally reaching the other side.
Quantitative transition state theory.
107 113
In
the 1880s Arrhenius observed that the rate of chemical
reactions varies with temperature according to Eq. 9-77
in a manner similar to the variation of an equilibrium
constant with temperature (integrate Eq. 6-37 and
compare). Here, k is a first-order rate constant, the
quantity E
a
is known as the Arrhenius activation
energy, and the constant A is referred to as the pre-
exponential factor or the frequency factor.
k = Ae
E
a
/RT
The Arrhenius equation, together with studies of
the effects of salts on reaction rates and observation of
quantitative correlations between rates and equilibrium
constants, suggested that a rate constant for a reaction
might be a product of a constant term which is nearly
independent of temperature and a constant K

which
has the properties of an equilibrium constant for for-
mation of the transition state. Eyring made this quan-
titative in 1935 with his absolute rate theory
112
according to which all transition states break down
with a rate constant k
B
T/h. Eyring reached this
conclusion by assuming that the rate of a chemical
reaction is determined by the frequency of stretching of
the bond that is being broken in the transition state. To be
more precise, it is the normal-mode oscillation of the
transition state complex along the reaction coordi-
nate.
109
This frequency was deduced by describing
the vibrational energy as h (from quantum mechanics)
and as k
B
/T (from classical mechanics) and setting
them equal.
h = k
B
T and
= k
B
T/ h
k
B
is the Boltzmann constant
h is Plancks constant.
The right side of Eq. 9-78 is usually multiplied by a
transmission coefficient , which may vary from
1 to 0.1 or even much less. However, for lack of any
better value, is usually assumed to be 1. From Eq.
9-78, at 25C = 6.2 x 10
12
s
1
. This is the maximum
rate for a chemical reaction of molecules in the transi-
tion state. This is the rate for a single molecule and
must be multiplied by the concentration of the reacting
substance X in the transition state. This concentration
[X]

is determined by the equilibrium constant


K

= [X]

/ [X]. The velocity of the reaction becomes



The first-order rate constant is
Since G

= RT lnK

, Eq. 9-80 can be rewritten as Eq.


9-81, in which G

is the Gibbs energy of activation.


Transition state
Reactants
Products
G

(
o
r

E
)
Reaction coordinate
G

G
High energy region of
dissociated molecules,
e.g., X
+
or X*
Length, X A of bond breaking
L
e
n
g
t
h
,

X






B

o
f

b
o
n
d

f
o
r
m
i
n
g
Figure 9-15 (A) Transition state diagram illustrating Gibbs
energy vs reaction coordinate for conversion of reactants to
products in a chemical reaction. (B) Contour map of Gibbs
energy vs interatomic bond distances for reaction B + X A
B X + A.
(9-77)
D. Mechanisms of Catalysis
A
B
(9-78)
(9-79)
(9-80)


d X
dt
X
B
[ ]
[ ] v
k T
h
K

k K
k T
h
K
1


d X dt
X
B
[ ]
[ ]

(9-81)
k
k T
h
e
G RT
1

B

s
1
s
1
484 Chapter 9. Enzymes: The Catalysts of Cells
At 25C, G

in kJ/ mol, the following practical form


of Eq. 9-81 can be written.
k
1
= 6.2 x 10
12
e
G

/2.48
s
1
log k
1
= 12.79 G

/5.71
and G

= 73.0 5.71 log k


1
Equation 9-81 is approximate and a more correct
statistical mechanical treatment is available.
113,114
See
also comment on p. 288 about log k (and log k
1
) being
unitless. Employing Eq. 6-14, we may expand Eq. 9-81
as follows:
From this it appears that H

E
a
, the Arrhenius
activation energy. A more correct treatment gives
H

= E
a
RT for reactions in solution. However,
since RT at 25C is only 2.5 kJ mol
1
, the approximation
that H

= E
a
is often used. The preexponential term,
in parentheses in Eq. 9-82, depends principally on S

,
the entropy change accompanying formation of the
transition state. The quantities G

, H

, and S

are
sometimes measured for enzymatic reactions but useful
interpretations are difficult.
Equations 9-81 and 9-82 also show that a small
decrease in transition state energy will give a large
increase in rate; stabilization of the transition state by
5.7 kJ / mol (1.4 kcal / mol) will increase the rate 10-fold.
If G

= 400 kJ/mol, as for a strong covalent single


bond, k
1
= 5 x 10
58
s
1
and the half-life t = .693 / k
1
=
1.3 x 10
57
s (4 x 10
49
years, greater than the ~ 10
10
years
estimated age of the universe). If G

= 100 kJ / mol,
log k
1
4.7 s
1
, k
1
= 1.9 x 10
5
, and t 10 h. This is
about the rate of a typical nonenzymatic model
reaction at 25C. If G

= 50 kJ / mol, log k
1
4, as for
a fast enzyme.
We can conclude that enzymes make use of relatively
small energy differences in catalyzing reactions. The ener-
gies of numerous van der Waals interactions of hydro-
gen bonds and electrostatic attraction or repulsion of
charges are sufficient. Nevertheless, we can see that
anything that stabilizes the transition state (decreases
G

) will increase the rate of reaction. The role of a


catalyst is to permit the formation of a transition state
of lower energy (higher stability) than that for the
uncatalyzed reaction. Stabilization of the transition
state of a reaction by an enzyme suggests that the
enzyme has a higher affinity for the transition state
than it does for substrate or products, an idea that
appears to have been expressed first by Haldane
41
and popularized by Pauling.
115
I think that enzymes are molecules that are comple-
mentary in structure to the activated complexes of the
reactions that they catalyze. The attraction of the enzyme
(9-82)
molecule for the activated complex would thus lead to a
decrease in its energy, and hence to a decrease in energy
of activation of the reaction and to an increase in the
rate of the reaction.
Transition state inhibitors. Suppose that a
chemical reaction of a compound S takes place with
rate constant k
N
through transition state T. Let the
equilibrium constant for formation of T be K
N

. Assume
that an enzyme E can combine either with S with disso-
ciation constant K
dS
or with the compound in its transi-
tion state structure T with dissociation constant K
dT
(Eq. 9-83).
If equilibrium is assumed for all four sets of double
arrows it is easy to show that Eq. 9-84 holds.
K
E

/K
N

= K
dS
/K
dT
According to transition state theory, if the transmission
coefficient = 1, T and ET will be transformed to prod-
ucts at the same rate. Thus, if the mechanisms of the
nonenzymatic and enzymatic reactions are assumed
the same, the ratio of maximum velocities for first-
order transformation of ES and S will be given by Eq.
9-85. For some enzymes the ratio
k
E
/k
N
= K
E

/K
N

= K
dS
/K
dT
k
E
/ k
N
may be 10
8
or more. Thus, if K
dS
10
3
the constant
K
dT
would be 10
11
. The enzyme would be expected
to bind the transition state structure (T) 10
8
times more
tightly than it binds S.
The foregoing reasoning suggests that if structural
analogs of T could be found for a particular reaction,
they too might be very tightly bound more so than
ordinary substrate analogs.
116
Wolfenden
117
listed a
series of compounds which may be transition state
inhibitors of this type and many others have been
described.
117a
Nevertheless, very tight, reversible
binding is no proof that an inhibitor is actually binding
to the transition state structure of the enzyme. For
example, an inhibitor may bind to a conformational
form of the enzyme that lies off to a side rather than
directly on the reaction pathway for catalysis.
Describing the transition state. How can we
describe the structure of the transition state when we
have no direct knowledge of its structure? We can try
(9-83)
1
2
1
2
k
k T
h
e e
S R H RT

j
(
,
\
,
(
B

E
K
dS
ES
+
E
K
dT
ET
+
product
K
E

S T product
K
N

(9-84)
(9-85)
s
1
485
BOX 9-E CHORISMATE MUTASE
Bacteria, fungi, and plants convert products of
glucose metabolism into the aromatic amino acids
phenylalanine, tyrosine, and tryptophan by a com-
plex sequence of reactions that is described in Chap-
ter 25. One of these reactions, the conversion of
chorismate into prephenate, is unique among
enzyme-catalyzed reactions. It is a Claisen rear-
rangement that occurs readily without catalysis in an
aqueous solution. However, the rate is increased
over one million-fold by the enzyme chorismate
mutase. The enzyme from Bacillus subtilis is a trimer
of identical, small 127-residue subunits. Many
studies, both of the nonenzymatic and enzymatic
reactions, have suggested a pericyclic mechanism
involving a polar chairlike transition state whose
presumed structure is shown in the accompanying
equation.
a f
The transition state analog, whose
a
Gray, J. V., Eren, D., and Knowles, J. R. (1990)
Biochemistry 29, 8872 8878
b
Turnbull, J., Cleland, W. W., and Morrison, J. F.
(1991) Biochemistry 30, 7777 7782
c
Rajagopalan, J. S., Taylor, K. M., and Jaffe, E. K.
(1993) Biochemistry 32, 3965 3972
d
Chook, Y. M., Gray, J. V., Ke, H., and Lipscomb,
W. N. (1994) J. Mol. Biol. 240, 476 500
e
Gray, J. V., and Knowles, J. R. (1994) Biochemistry
33, 9953 9959
f
Lyne, P. D., Mulholland, A. J., and Richards, W.
G. (1995) J. Am. Chem. Soc. 117, 11345 11350
g
Lee, A. Y., Karplus, P. A., Ganem, B., and Clardy,
J. (1995) J. Am. Chem. Soc. 117, 3627 3628
h
Xue, Y., Lipscomb, W. N., Graf, R., Schnappauf,
G., and Braus, G. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 10814 10818
i
Wiest, O., and Houk, K. N. (1995) J. Am. Chem.
Soc. 117, 11628 11639
j
Kast, P., Hartgerink, J. D., Asif-Ullah, M., and
Hilvert, D. (1996) J. Am. Chem. Soc. 118, 3069
3070
k
Lin, S. L., Xu, D., Li, A., Rosen, M., Wolfson, H.
J., and Nussinov, R. (1997) J. Mol. Biol. 271, 838
845
l
Ma, J., Zheng, X., Schnappauf, G., Braus, G.,
Karplus, M., and Lipscomb, W. N. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 14640 14645
C
O

O
OH
O
C
COO
CH
2
CH
2
O
C O
O
COO
HO
CH
2
O
C O
O
COO
HO
H
OOC
H HO
CH
2
C
COO
O
CH
2
O
OH
O
O
N
N
N
Arg 28
H
H
H
N
Transition
state
O
O
H
2
N
N
N
Prephenate

+
H
Transition
state analog
H

H
Arg 12
Bound
conformation
H
Chorismate
H
H
N
H
H
H

+
Lys 39
(Arg 90 in B Subtilis)
+
+ H

structure is also depicted, is a powerful inhibitor.


X-ray structural studies show that this inhibitor
binds in the conformation shown with a number of
interactions with polar side chain groups.
d,g
Some
of these interactions are shown for the E. coli enzyme
in the figure;
g
they are different in enzymes from
B. subtilis
d
and yeast.
h
However, in no case are there
groups in obvious positions to serve as acidbase
catalysts and the maximum velocity V
max
is inde-
pendent of pH.
b
NMR studies show that the bound
product prephenate displays large shifts in some
13
C resonances, indicating strong interaction with
the polar side chains of the protein.
c
Theoretical
calculations also suggested that these interactions,
especially those with Arg 90 and Glu 78 of the B.
subtilis enzymes,
f l
help to stabilize the partial charge
separation in the polar transition state. Studies of
mutant enzymes have confirmed that
the charged Arg 90 (Lys 39 for E. coli)
side chain is essential.
k
D. Mechanisms of Catalysis
486 Chapter 9. Enzymes: The Catalysts of Cells
to predict the atomic coordinates of all of the atoms of
the reacting substrates and of the protein. The transi-
tion state structure of the substrate must be between
that of the last ES complex and the first EP complex
formed. It should be similar to that of a transition
state inhibitor. We can hope to obtain suitable X-ray
or NMR structures and deduce an appropriate enzyme
structure by molecular modeling. Theoretical calcula-
tions using quantum chemical methods are of value in
this effort.
118,119
Lengths of the breaking and forming bonds in
a transition state are often estimated using kinetic
isotope effects (KIEs) on the velocity of the reac-
tion.
118 122
This is somewhat indirect. Nevertheless,
by measuring these effects for a large number of sub-
strates and several isotopes a picture of the transition
state structure that is good enough to use as a model for
design of an enzyme inhibitor may be obtained.
117a,121 124
Other physical measurements such as Raman difference
spectroscopy of isotopically labeled inhibitors can also
be of value.
125
Getting to the transition state. Since some
bonds must lengthen and some shorten and bond
angles must also change, it may not be simple for an
enzyme to catalyze necessary steps prior to the rate-
limiting breakdown of the transition state. Hydrogen
bonds within the protein may need to be broken and
reformed with new bonding partners. Nonpolar side
chains may need to shift to give a packing arrange-
ment that will allow the movement of nuclei that must
occur in forming the transition state structure. The
enzyme must not only stabilize the transition state but
also hold the substrate and escort it into the transition
state configuration.
The attacking hydroxyl ion in Eq. 9-76 carries a
negative charge which becomes distributed more or
less equally between the hydroxyl and the iodine atom
in the transition state. In this state the CH
3
group
carries a partial positive charge as well. To provide
good complementarity between an enzyme and this
transition state, a change in the initial charge distribu-
tion within the enzyme will also be required. If a
positively charged group initially binds the OH

ion
it may have to lose part of its charge and a group next
to the iodide atom may have to gain positive charge.
In the nonenzymatic reaction of Eq. 9-76 the ionic
atmosphere provided by positive counterions in solution
can continuously readjust to keep the negative charge
effectively balanced at every step along the reaction
coordinate and through the transition state. Within
enzymes this adjustment may occur via redistribution
of electrical charges within the polarizable network of
internal hydrogen bonds. The enzyme structure must
allow this. Because of the complexity of an enzymatic
transition state it may be hard to compare it with the
transition state of the corresponding nonenzymatic
reaction. Effects of pressure
125a
and of temperature
125b
also raise doubts about the simple picture of Eq. 9-83.
It also follows that tight binding of substrates in the
ground state does not necessarily interfere with transi-
tion state stabilization.
126 129a
2. Microscopic Reversibility
The important statistical mechanical principle of
microscopic reversibility asserts that the mechanism of
any chemical reaction considered in the reverse direction
must be exactly the inverse of the mechanism of the forward
reaction. A consequence of this principle is that if the
mechanism of a reaction is known, that of the reverse
reaction is also known. Furthermore, it follows that
the forward and reverse reactions catalyzed by an enzyme
must occur at the same active site on the enzyme and the
transition state must be the same in both directions.
The principle of microscopic reversibility is often useful
when the likelihood of a given mechanism is being
considered. If a mechanism is proposed for a reversible
reaction in one direction the principle of microscopic
reversibility will give an unambiguous mechanism for
the reverse reaction. Sometimes this reverse mechanism
will be chemically untenable and, recognizing this, the
enzymologist can search for a better one.
3. Acid and Base Catalysis
Many reactions that are promoted by enzymes
can also be catalyzed by acids or bases or by both. An
example is mutarotation, the reversible interconversion
of the - and -anomeric forms of sugars (Eqs. 4-1
and 9-86). This reaction is catalyzed by a specific
mutarotase and also by inorganic acids and bases.
The frequently observed bell-shaped curve for the
dependence of rate of catalysis on pH (e.g., Eqs. 9-55
to 9-57 and Fig. 9-8) also suggests participation of both
protonated and unprotonated acidbase groups
present in enzymes.
Acidic and basic groups in enzymes. In this
discussion the symbols HB or H
+
B will be used for acids
and B

or B for their conjugate bases. Remember that


strong acids have low pK
a
values and that the conjugate
bases formed from them are weak bases. Likewise, very weak
acids have high pK
a
values and their conjugate bases are strong.
The following are among the pK
a
values that may be
important in considering an enzyme mechanism. How-
ever, depending upon the environment of the side chain
in a protein, these pK
a
values may fall substantially out-
side of the indicated ranges.
487
1.74 H
3
O
+
in 55.5 M water
~3.6 COOH, terminal in peptide
4.34.7 COOH in a glutamic or aspartic acid side chain
6.47.0 Imidazole (histidine) and phosphate (OPO
3
H

)
7.58.0 NH
3
+
, terminal in peptide
8.510 SH of cysteine side chains
9.511 Phenolic OH of tyrosine
~10.5 NH
3
+
of lysine side chain
~13.6 CH
2
OH of serine side chain
15.7 free HO

in 55.5 M H
2
O, 25C
Acidbase catalysis of mutarotation. The
mutarotation of glucose proceeds through the free
aldehyde form as an intermediate (Eq. 9-86). A hydro-
gen atom is removed as H
+
from the oxygen at carbon
1 in step a and a proton (probably a different one) is
transferred to the oxygen of the ring with cleavage of
the O C bond to the anomeric carbon atom. A similar
process in reverse is required for step b. Transfers of
hydrogen ions between atoms of oxygen, nitrogen, and
sulfur atoms are a common feature of biochemical
reactions. The bonds between hydrogen and O, N, and
S atoms tend to be polarized strongly, leaving a partial
positive charge on the hydrogen atoms. Consequently,
the groups are weakly acidic and protons can be trans-
ferred from them relatively easily. It is reasonable to
suppose that acid and base catalysis is related to these
proton transfers.
HO
O
HO
OH
OH
H
OH
HO
C
O
HO
OH
H
O
HO
O
HO
OH
H
O
OH
H

Free aldehyde form


H
a
b
step a). The anion is isomerized to a second anion
with the ring opened (step b). Addition of a proton
(transfer of a proton from H
3
O
+
) produces the free
aldehyde form of the sugar (step c).
The catalytic base B: might be HO

or a weaker
base such as ammonia or even water. For reactions the
rate is proportional only to the concentration of OH

and the presence of other weaker bases has no effect.


129b
Such catalysis is referred to as specific hydroxyl ion
catalysis.
19
More commonly, the rate is found to depend
both on [OH

] and on the concentration of other weaker


bases. In such cases the apparent first-order rate constant
(k
obs
) for the process is represented by a sum of terms
(Eq. 9-88). The term k
H
2
O
is the rate in
k
obs
= k
H
2
O
+ K
OH
[OH

] + k
B
[B]
pure water and represents catalysis occurring by the
action of water alone as either an acid or a base. The
last two terms represent the contributions to the catal-
ysis by OH

and by the other base, respectively. The


termk
B
[B] represents general base catalysis.
Catalysis of mutarotation by acids occurs if an acid
donates a proton to the oxygen in the sugar ring as
(9-86)
D. Mechanisms of Catalysis
HO
O
OH
H
O HO
OH
H
HO
O
OH
H
O

HO
HO
C
O

OH
HO
H
O
HO
C
OH
OH
HO
H
O

+
OH
OH
OH
B
BH
+
a
b
c
H
+
(9-87)
(9-88)
HO
O
OH
O HO
OH
H
H B
HO
C
O
OH
O HO
OH
+
H
H
+
B
H
+
(9-89)
General base and general acid catalysis. Base-
catalyzed mutarotation might be formulated as follows:
A hydroxyl ion or some other base attacks the proton
on the anomeric OH group of the sugar, removing it
to form an anion and the conjugate acid BH
+
(Eq. 9-87,
488 Chapter 9. Enzymes: The Catalysts of Cells
BOX 9-F IMMUNOPHILINS AS ROTAMASES
Since its introduction into clinical use in about
1979 the immunosuppresant cyclosporin has been
responsible for a revolution in human organ trans-
plantation.
a c
The exact mechanism of action in
suppressing T-lymphocyte-mediated autoimmune
responses is still not completely clear, but cyclosporin,
a cyclic lipophilic peptide from a fungus, was
found to bind to specific proteins that were named
cyclophilins.
d
Human cyclophilin A is a 165-residue
protein which associates, in the crystal form, as a
decamer with five-fold rotational and dihedral
symmetry.
e
This protein is also found in almost all
N
C
C
N
C
H
X H
H
O
O
trans
C
C
N
X H
O
cis
H
C O
C
C
N
O

+
Contributing
resonance form
X H
other organisms and has a highly conserved sequence.
f
Another immunosuppresant, a synthetic molecule
known only as FK506, has an action similar to that
of cyclosporin but binds to distinctly different pro-
teins, the FK506-binding proteins (FKBPs), of which
the 107-residue FKBP-12 is the best known.
g j
These
proteins also bind another immunosuppresant,
rapamycin, which, however, has different biological
properties than does FK506. Both FK506 and rapa-
mycin are macrocyclic compounds but with structures
very different from that of cyclosporin.
It was a surprise to discover that all of the cyclo-
philins and FK506-binding proteins are peptidyl
prolyl cistrans isomerases or rotamases. They
all catalyze the following simple and reversible
reaction of a prolyl peptide linkage:
Cyclosporin (black) bound
to human cyclophilin. From
Pflgl et al.
e
Courtesy of J. N.
Jansonius.
The transition state energy for the reaction is
lowered by 33 kJ mol
1
by cyclophilin A
k
and by
27 kJ mol
1
by FKBP-12
g
with corresponding rate
increases of ~ 6 x 10
5
- and 5 x 10
4
-fold, respectively.
Sarcosine
(
H
3
C
N
N
N
N
O
H
H
H
3
C
O
H
3
C
H
CH
3
O
H
H
N
N
N
N
CH
3
O
H
H
O
H
O
CH
3
O
H
H
O
N
CH
2
H
CH
3
N
O
H
N
H
O
CH
3
O
D-Ala
H
3
C
Ala
H
3
C H
MeLeu
Val
H HO
MeLeu
MeLeu
Val
N-CH
3
-Gly)
4-Butenyl-4,N-dimethyl-Thr
Aminobutyrate
4
CH
3
3
Cyclosporin A
1
2
5
10
11
8
7
6
9
MeLeu
489
BOX 9-F (continued)
a
Kahan, B. D. (1989) N. Engl. J. Med. 321, 1725 1738
b
Schreiber, S. L. (1991) Science 251, 283 287
c
High, K. P., Joiner, K. A., and Handschumacher, R. E. (1994)
J. Biol. Chem. 269, 9105 9112
d
Fruman, D. A., Burakoff, S. J., and Bierer, B. E. (1994) FASEB J.
8, 391 400
e
Pflgl, G. M., Kallen, J., Jansonius, J. N., and Walkinshaw, M. D.
(1994) J. Mol. Biol. 244, 385 409
f
Lippuner, V., Chou, I. T., Scott, S. V., Ettinger, W. F., Theg, S. M.,
and Gasser, C. S. (1994) J. Biol. Chem. 269, 7863 7868
g
Fischer, S., Michnick, S., and Karplus, M. (1993) Biochemistry 32,
13830 13837
h
Orozco, M., Tirado-Rives, J., and Jorgensen, W. L. (1993)
Biochemistry 32, 12864 12874
i
Van Duyne, G. D., Standaert, R. F., Karplus, P. A., Schreiber, S.
L., and Clardy, J. (1993) J. Mol. Biol. 229, 105 124
j
Lam, E., Martin, M. M., Timerman, A. P., Sabers, C., Fleischer,
S., Lukas, T., Abraham, R. T., OKeefe, S. J., ONeill, E. A., and
Wiederrecht, G. J. (1995) J. Biol. Chem. 270, 26511 26522
k
Eberhardt, E. S., Loh, S. N., Hinck, A. P., and Raines, R. T. (1992)
J. Am. Chem. Soc. 114, 5437 5439
l
Kakalis, L. T., and Armitage, I. M. (1994) Biochemistry 33, 1495
1501
m
Zhao, Y., and Ke, H. (1996) Biochemistry 35, 7362 7368
n
Thriault, Y., Logan, T. M., Meadows, R., Yu, L., Olejniczak, E.
T., Holzman, T. F., Simmer, R. L., and Fesik, S. W. (1993) Nature
(London) 361, 88 91
o
Konno, M., Ito, M., Hayano, T., and Takahashi, N. (1996) J. Mol.
Biol. 256, 897 908
p
Kern, D., Kern, G., Scherer, G., Fischer, G., and Drakenberg, T.
(1995) Biochemistry 34, 13594 13602
q
Gthel, S. F., Herrler, M., and Marahiel, M. A. (1996) Biochemistry
35, 3636 3640
r
Clubb, R. T., Ferguson, S. B., Walsh, C. T., and Wagner, G. (1994)
Biochemistry 33, 2761 2772
s
Edwards, K. J., Ollis, D. L., and Dixon, N. E. (1997) J. Mol. Biol.
271, 258 265
t
Rudd, K. E., Sofia, H. J., Koonin, E. V., Plunkett, G., III, Lazar,
S., and Rouviere, P. E. (1995) Trends Biochem. Sci. 20, 12 14
u
Rahfeld, J.-U., Rcknagel, K. P., Stoller, G., Horne, S. M.,
Schierhorn, A., Young, K. D., and Fischer, G. (1996) J. Biol. Chem.
271, 22130 22138
v
Matouschek, A., Rospert, S., Schmid, K., Glick, B. S., and
Schatz, G. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 6319 6323
For both enzymes the maximum velocity is indepen-
dent of pH over a wide region. For cyclophilin the
rate is nearly independent of the nature of residue X
in the foregoing structure but FKBP prefers a hydro-
phobic residue.
How do these enzymes work? One possibility
would be for the protein to transfer a proton to the
nitrogen atom of the proline ring. This would destroy
the partial double-bond character of the amide linkage
and allow free rotation about a single bond. The
same thing could be accomplished if a nucleophilic
group from the enzyme formed a covalent adduct
with the C=O of the substrate. However, this seems
unlikely because there is no suitably placed nucleo-
phile. A third possibility is that the enzyme distorts
the substrate and stabilizes the transition state using
only noncovalent interactions.
g,h,l
This would account
for the lack of pH dependence and observable sol-
vent isotope effects. However, in human cyclophilin
the guanidinium group of arginine 55 hydrogen
bonds to the peptide C = O of the substrates proline
residue and could easily shift to place a guanidinium
proton against the proline ring nitrogen effectively
protonating it and permitting rotation about the
peptide linkage.
m
Do cyclosporin and FK506 act as transition state
inhibitors? If so, we might learn something about
the mechanism from the three-dimensional structures
of the inhibited rotamases.
i,l,n,o
From these structures
as well as those with simpler substrate analogs, it is
seen that the substrate is desolvated and that many
nonbonded (van der Waals) interactions stabilize
the binding. Hydrogen bonding is also important.
p,q
The presence of distinct hydrogen bonds to the pep-
tide NH and C=O groups on the N-terminal side
of the substrate X-Pro linkage and less well-defined
bonding on the terminal side suggests that the
C-terminal part may be rotated while the enzyme
holds the N-terminal part.
p
It can also be concluded
that mechanisms are probably not identical for all
rotamases.
Cyclophilins and FKBPs are large families of
proteins. In E. coli there are two cyclophilin genes,
r,s
three encoding FK506-binding proteins, and one of
D. Mechanisms of Catalysis
a third family of bacterial rotamases (parvulins) that
lack sequence similarity to the other two families.
t,u
Yeast contains at least five cyclophilins
v
and even
more eukaryotic FKBPs are known.
j
There is abun-
dant evidence that these proteins play an important
role in protein folding in vivo (Chapter 29).
shown in Eq. 9-89. Again, either specific acid catalysis
(by H
3
O
+
) or general acid catalysis is possible.
Enzymes are not able to concentrate protons or
hydroxyl ions to the point that specific base or acid
catalysis would be effective. However, either general
acid or general base catalysis can be accomplished by
groups present in an enzyme in their normal states of
protonation at the pH of the cell. Thus, if one of the
enzymes must be dissociated to its conjugate base
and a second must be protonated for reaction to take
place (as in Eq. 9-54), it is reasonable to suppose that
these two groups participate in acidbase catalysis.
Of the acidic and basic groups present in proteins, the
imidazole group of histidine would appear to be the
490 Chapter 9. Enzymes: The Catalysts of Cells
HO
O
OH
O HO
OH
H
H B
H
HO
C
OH
OH
O HO
OH
H
B
+
(a base)
(an acid)
B (a base)
H
+
B (an acid)
(9-92)
most ideal both for general base catalysis and, because
both forms may exist in nearly equal amounts at pH 7,
for general acid catalysis. However, carboxyl, thiol,
lysyl, tyrosyl, and N-terminal amino groups are all
thought to function in various enzymes.
The mutarotase of E. coli has a turnover number
of 10
4
s
1
. The plot of log K
m
vs pH indicates two pK
a
values in the free enzyme at 5.5 and 7.6, while the plot
of log V
max
yields a single pK
a
of 4.75 for the ES com-
plex.
130
The latter might represent a carboxyl group
on an imidazole group in its conjugate base form.
Why doesnt the group having pK
a
7.6 in the free
enzyme also show a pK
a
in the ES complex? Either
the group has no catalytic function so that EH
2
S of Eq.
9-54 reacts to form products just as fast as does HES
or the pK
a
is so strongly shifted by substrate binding
that it is not detected in the log V
max
plot.
The Brnsted relationships. The effectiveness of
a specific base as a general base catalyst can usually be
related to its basicity (pK
a
) via the Brnsted equation
(Eq. 9-90). Here, k
B
is defined by Eq. 9-88 and G
B
is a
log k
B
= log G
B
+ (pK
a
)
constant for a particular reaction. A similar equation
(9-91) relates the constant k
A
for general acid catalysis
log k
HB
= log G
A
(pK
a
)
to pK
a
. These equations are linear Gibbs energy relation-
ships similar to the ones discussed in Box 6-C. For the
Brnsted equations to hold, the Gibbs energy of activa-
tion for the reaction must be directly related to the
basicity or acidity of the catalyst.
The exponents and of Eqs. 9-90 and 9-91 mea-
sure the sensitivity of a reaction toward the basicity or
acidity of the catalyst. It is easy to show that as and
approach 1.0 general base or general acid catalysis is
lost and that the rate becomes exactly that of specific
hydroxyl ion or specific hydrogen ion catalysis.
131
As or approach zero basic or acidic catalysis is
indetectable. Thus, general base or general acid catal-
ysis is most significant when or is in the neighbor-
hood of 0.5. Under these circumstances it is easy to
see how a moderately weak basic group, such as the
imidazole group of histidine, can be an unusually
effective catalyst at pH 7.
To determine or experimentally a plot of log k
B
or log k
HB
vs pK
a
(a Brnsted plot) is made and the
slope is measured. Statistical corrections (Chapter 7)
should be applied for dicarboxylic acids and for am-
monium ions from which one of three protons may be
lost from the nitrogen atom. General base or general
acid catalysis implies an important feature of any mech-
anism for which it is observed, namely, that removal
of a proton or addition of a proton is involved in the
(9-90)
(9-91)
rate-determining step of a reaction.
Concerted acidbase or tautomeric catalysis.
A third possible type of catalysis requires that a base
and an acid act synchronously to effect the breaking and
formation of bonds in a single step. Thus, tetramethyl-
glucose mutarotates very slowly in benzene containing
either pyridine (a base) or phenol (an acid). However,
when both pyridine and phenol are present, mutarota-
tion is rapid. This suggested to Swain and Brown
132
a concerted mechanism (Eq. 9-92) in which both an
acid and a base participate.
During the reaction shown, the acid BH
+
is con-
verted to its conjugate base B and the base B to its
conjugate acid H
+
B. It might seem that these agents,
having been altered by the reaction, are not serving as
true catalysts. However, a simple proton exchange will
restore the original forms and complete the catalytic
cycle. In aqueous solutions, water itself might act as
the acid or the base or even both in concerted catalysis.
The original experimental evidence for concerted
acidbase catalysis of the mutarotation in benzene is
now considered unsound
133,134
and concerted acid
base catalysis has been difficult to prove for nonenzy-
matic reactions in aqueous solution. However, mea-
surements of kinetic isotope effects seem to support
Swain and Browns interpretation.
135
Concerted acid
base catalysis by acetic acid and acetate ions may have
been observed for the enolization of acetone
136
and it
may be employed by enzymes.
136a
Swain and Brown showed that a more effective
catalyst for the mutarotation of sugars than a mixture
of an acid and a base can be designed by incorporating
the acidic and basic groups into the same molecule.
132,135
Thus, with 0.1 M tetramethylglucose in benzene solution,
0.001 M -hydroxypyridine is 7000 times as effective
a catalyst as a mixture of 0.001 M pyridine + 0.001 M
phenol. Swain and Brown suggested the following
completely concerted reaction mechanism for the
polyfunctional catalyst -hydroxypyridine in which
the hydrogen-bonded complex formed (Eq. 9-93) is
analogous to an enzymesubstrate complex. The
product of the catalyst is 2-pyridone, a tautomer of
491
2-hydroxypyridine, with which it is in a rapid revers-
ible equilibrium.
Rony called catalysis of the type illustrated in Eq.
9-93 tautomeric catalysis and suggested that its effi-
ciency lies not simply in the close proximity of acidic
and basic groups in the same molecule but also in the
ability of the catalyst to repeatedly cycle between the
two tautomeric states.
133
For an enzyme the tautomer-
ization of the free catalyst could sometimes be rate
determining (see Section A,4 on isomechanisms).
4. Hydrogen Bonding and the Transfer of
Protons within Active Sites
An extensive network of hydrogen bonds runs
throughout most proteins and may be especially
complex within active sites. The network often runs
through bound substrate molecules and immobilized
water molecules in the active site cavity. This network
arises in part because of the frequent occurrence of
acidic and basic catalytic groups in active sites and
by the fact that many substrates contain polar groups.
The structure of an enzyme often seems to be more
rigid in a complex with substrates or inhibitors than in
the free state. Does this network of linked hydrogen
bonds play a role in catalysis? If so, what? Wang
H
3
CO
O
H
3
CO
O
H
O
H
3
CO
H
N HO
H
3
CO
O
H
3
CO
O
H
O
H
3
CO
N
O
H
H
H
3
CO
C
O
H
3
CO
O
H
O
H
3
CO
H
N O
H
H
3
C
2-Hydroxypyridine
H
3
C
Complex
H
3
C
2-Pyridone
suggested that rapid transfer of protons along rigidly
and accurately held hydrogen bonds in the ES complex
may be an essential feature of enzymatic catalysis.
137
This conclusion is supported by many more recent
observations. A remaining question is whether this
proton transfer can take place in such a way that the
transition state barrier is lowered.
Ultrafast proton transfer. The diffusion-
controlled limit for second-order rate constants
(Section A3) is ~ 10
10
M
1
s
1
. In 1956, Eigen, who had
developed new methods for studying very fast reactions,
discovered that protons and hydroxide ions react much
more rapidly when present in a lattice of ice than when
in solution.
138
He observed second-order rate constants
of 10
13
to 10
14
M
1
s
1
. These represent rates almost as
great as those of molecular vibration. For example,
the frequency of vibration of the OH bond in water is
about 10
14
s
1
. The latter can be deduced directly from
the frequency of infrared light absorbed in exciting
this vibration: Frequency equals wave number (3710
cm
1
for OH stretching) times c, the velocity of light
(3 x 10
10
cm s
1
).
This ultrafast transfer of protons can be explained
as follows: The OH

ion and the proton, which is


combined with a water molecule to form H
3
O
+
, are
both hydrogen-bonded to adjacent water molecules.
In ice a chain of hydrogen-bonded water molecules
links the hydroxide and the hydrogen ions (Eq. 9-94).
By synchronous movement of electron pairs from the
OH

ion and from each of the water molecules in the


(9-93)
D. Mechanisms of Catalysis
H O

H O
H
H
O
H
H O
H
H
O
H
H O
H
H
H O
H
H
O
H
H O
H
H
O
H
H O
H
H
O
H
H O
H
H
O
H
H O
H
H
O
H
H O
H
+
H
2
O
Protons and
electrons move
Water molecules
reorient
A second pair of HO

and H
+
can now use the same pathway
a
b
(9-94)
492 Chapter 9. Enzymes: The Catalysts of Cells
a b c
A H
k
1
A H
k
2
B A H
k
3
B H B
B A
(9-97)
chain (as indicated by the little arrows) the neutraliza-
tion can take place during the time of one molecular
vibration (step a, Eq. 9-94). The positions of the oxygen
atoms remain unchanged at the end of the reaction but
the protons that were engaged in hydrogen bond for-
mation have moved toward the left and are now at-
tached to different oxygen atoms. The central chain of
water molecules can be restored to its original state if
each molecule rotates around one of its single bonds,
swinging a hydrogen from right back to left (Eq. 9-94,
step b). This must be a slower process which may
occur one molecule at a time. The rotation of the
leftmost water molecule would leave an empty space
between two O-atoms, a fault in the H-bonded chain
of the ice structure. This fault can be corrected by
rotation of the second molecule (Eq. 9-95); however,
this would create a fault at the right of the second water
molecule. This would induce rotation of the third
molecule, etc., causing the fault to migrate from left
to right across the entire chain of water molecules (Eq.
9-94, step b) and leaving the chain ready to function
again in transferring another proton.
Because of the disorder present in ice, an array
of water molecules such as that in Eq. 9-94 wouldnt
revert to its exact original form in step b. However,
active sites of enzymes are highly structured and proton
transfers may occur with precision. For example, a
synchronous shift of protons in an array of carboxylic
acid, imidazolium, and phosphate groups can be
envisioned readily (Eq. 9-96). The net effect of the
process is to transfer a proton from one end of the
chain to the other (as in Eq. 9-94) with facile tauto-
merization reactions providing the pathway. Such a
pathway might be constructed by protein side chains
to join the two sides of an active center promoting a
concerted acidbase catalyzed reaction such as that of
Eq. 9-92. Other tautomerization processes are possible
within proteins if the existence of less stable minor
tautomers of selected amide groups in the peptide back-
bone is allowed.
139
Nagle and Morowitz suggested a
process similar to that of Eq. 9-94 but involving side
chains of serine residues.
140
Proton transfer rates. Consider the reversible
reaction of a proton acceptor B with acid H-A (Eq. 9-97).
Eigen pointed out that the reaction will be fastest if the
two reactants form a hydrogen-bonded complex (Eq.
9-97, step a).
138
The hydrogen bonding shortens the
distance from the proton to B and allows for very
rapid transfer of the proton from A to B within the
hydrogen-bonded complex (step b). The activation
energy is close to zero. The complex dissociates in step
c to form the products. The reactions are reversible, even
though they have been indicated by unidirectional
arrows.
The hydrogen-bonded complexes A H - B and A
- H B can be formed readily if A and B are oxygen or
nitrogen bases such as COO

, OH, NH
2
, or imida-
zole. In such cases, as in ice, the interconversion of
the two complexes (step b in Eq. 9-97) is very fast. The
overall rate of the proton transfer will then be limited
by diffusion of B and H A together or by diffusion of
A and H B apart. It might seem that these two pro-
cesses should also both be very fast. However, the
rates will be determined by the concentration ratio
[AH- B]/[A- HB]. If [AH- B] equals
[A- H B] the rate of dissociation of A H B will be
half what it is if nearly all of the complex is A- H B.
If A H- B predominates by 1000 to 1, the rate will be
slowed much more.
The equilibrium constant for the reaction of Eq.
9-97 will be:
K
eq
AB
= K
a
HA
/K
a
HB
= k
f
/k
r
or log K
eq
= pK
a
HA
pK
a
HB
= log k
f
log k
r
Here k
f
and k
r
refer to the rates in the forward and
reverse directions. If the pK
a
s of HA and HB are equal
k
f
and k
r
will be the same, but if they are very far from
equal the reaction will be slowed in one direction. If
proton transfer is a step in an enzymatic reaction it
may be slowed enough to become rate limiting.
The difference in pK
a
values between the proton
donor and the proton acceptor in Eq. 9-97 can be
expressed as the Gibbs energy change which at 25C
is equal to 5.71 x pK
a
. This is often referred to as
the thermodynamic barrier G to a reaction and G

can be expressed as the sum of the thermodynamic


barrier G plus an intrinsic barrier G

int
. For the
proton transfer of Eq. 9-97 the intrinsic barrier (for step
b) is thought to be near zero so that G

5.71 pK
a
.
H
O
H
O
H
H O
H
H
O
H H
H
O
H
O
H
H
fault fault
(9-95)
H O
C
O H N N H O
P
O
O O
H
+
(9-96)
(9-98)
493
From this we can conclude that two pK
a
values can be
as much as eight units apart and G

will still be less


than 50 kJ / mol, low enough to permit rapid enzymatic
reactions. However, for transfer of a proton from a
C H bond to a catalytic group, for example, to form
an enolate ion for an aldol condensation (Chapter 13),
the intrinsic barrier is known to be about 50 kJ / mol.
141
In this case, to allow rapid enzymatic reaction either
the thermodynamic barrier must be very low, as a
result of closely matching pK
a
values, or the enzyme
must lower the intrinsic barrier. It may do both.
Marcus theory. Discussion of intrinsic barriers is
often approached using a quantitative theory proposed
by Marcus.
142 145
It was first applied to electron transfer
(Chapter 16) but has been used for a great variety of
nonenzymatic and enzymatic reactions. As used by
Gerlt and Gassman,
141
the Marcus formalism describes
the reaction coordinate (Fig. 9-15A) as an inverted
parabola whose shape is determined by the overall
Gibbs energy change G and the intrinsic barrier
G

int
. The value of the Gibbs energy at any point on
the curve is designated G and the reaction coordinate
x is taken as 0 for the reactants and 1 for the products.
G = 4 G

int
(x 0.5)
2
+ G (x 0.5)
Limits: G/ 4 G

int
4 G
Differentiation of Eq. 9-99 yields the position of the
transition state coordinate x

as follows:
x

= 0.5 + G/ 8 G

int
and it follows that
G

= G

int
(1 + G/ 4 G

int
)
2
Diffusion-controlled dissociation of protons.
The direct proton transfer between C-1 and C-2 during
the action of sugar isomerases may seem puzzling.
How can a highly mobile proton remain attached to a
group in the enzyme for a millisecond or more instead
of being transferred out to a solvent molecule? This
can mean that the enzyme promotes the transfer of a
hydride ion or of a hydrogen atom rather than a proton
(see Chapter 13). If so, the observed proton exchange
with solvent would be an unimportant side reaction.
On the other hand, could the group in the enzyme that
removes the proton be out of contact with the aqueous
medium and thus able to hold onto the proton more
tightly? In recent years, it has been recognized that
neither of these explanations may be necessary. An
imidazole group is a likely proton-carrying group at
many active sites and it is thought that a proton can-
not be expected to transfer out from an imidazolium
group with a rate constant greater than ~ 10
3
s
1
.
(9-99)
(9-100)
(9-101)
The argument is as follows.
146
The rate of donation
of a proton from H
3
O
+
to imidazole (reverse of Eq. 9-102)
is known to be diffusion controlled with a rate constant
of 1.5 x 10
10
M
1
s
1
.
The equilibrium constant for Eq. 9-102, calculated
from the pK
a
of 7.0 for imidazole, is 10
7
M. Since K
eq
is also the ratio of the overall rate constants for the forward
and reverse reactions, we see that for the forward reac-
tion k
f
= 10
7
x 1.5 x 10
10
= 1.5 x 10
3
s
1
. This slow rate
results from the fact that in the intermediate complex
(in brackets in Eq. 9-102) the proton is on the imidazole
group most of the time. For a small fraction of the
time it is on the coordinated molecule H
2
O but reverts
to being on the imidazole many times before the imida-
zole and OH
3
+
separate (see also Eqs. 9-97 and 9-98).
Because of this unfavorable equilibrium within the
complex, the diffusion-controlled rate of proton trans-
fer from a protonated imidazole to water is far less
than for proton transfer in the reverse direction.
Coupled proton transfers. Enzymatic reactions
often require the transfer of two or more protons.
They may move individually, one proton at a time,
or as in Eq. 9-93 they may move synchronously in a
coupled or concerted process. Such coupled move-
ment is generally not possible for heavier nuclei.
147
However, studies of solvent isotope effects using a
proton inventory technique
111,148,149
have provided
evidence favoring coupled proton transfers for a variety
of enzymes. Movement of protons along hydrogen-
bonded paths, as well as electron transfer, may take
place with some participation of quantum mechanical
tunneling.
150152
Coupling to vibrational modes of the
hydrogen-bonded protons may provide vibration-
assisted tunneling.
153154d
These reactions are associ-
ated with unusually large kinetic isotope effects.
Unusually strong hydrogen bonds. The strength
of a hydrogen bond is thought to be directly related
to the length, which is ordinarily taken to be the distance
between the two surrounding heavier atoms (see Chap-
ter 2, Section B,3). Hydrogen bonds are sometimes
classified on the basis of NH---O distances
155,156
as:
<0.25 nm very strong
0.250.265 nm strong
>0.28 nm weak
>0.37 nm no van der Waals contact but
electrostatic interaction still occurs
NH---O distances may be a little longer than these.
ImH
+
+ H
2
O [ImH
+
OH
2
] Im + OH
3
+
Very fast diffusion controlled
k ~ 1.5 10
10
M
1
s
1
(9-102)
D. Mechanisms of Catalysis
494 Chapter 9. Enzymes: The Catalysts of Cells
The 0.276-nm hydrogen bonds in ice are regarded
as moderately strong. However, if one of the oxygen
atoms in an OH---O hydrogen bond carries a nega-
tive charge, as in the maleate monoanion, it will be
shorter.
156 158
Although the proton will be closer to one
oxygen atom than to the other, it will be able to move
between them by passing a very low transition state
barrier. For this to occur the microscopic pK
a
values
for the two groups (when they are protonated) must
be similar; for the maleate anion they are identical.
The strength of a hydrogen bond can be measured
for hydrogen-bonded complexes in the gas phase and
range from 10 100 kJ/ mol and even higher
155
for such
complexes as (FHF)
.
It is more difficult to establish
the strength in the liquid state or within the active site
of an enzyme, but shorter hydrogen bonds are usually
stronger than longer ones. Hydrogen bond distances
in crystals of small molecules can be measured precise-
ly by X-ray or neutron diffraction but greater uncer-
tainty is present in distances within proteins. Lengths
and probably strengths of hydrogen bonds in proteins
can be measured by NMR methods.
158a,b
For example,
solid-state NMR measurements on crystals of amino
acids and other carboxylic acids have provided a plot
of
1
H chemical shift vs hydrogen bond length.
159
For
very short (0.24- to 0.25-nm) hydrogen bonds the
1
H
chemical shift may be as great as 21 ppm. See also
Fig. 3-30 and associated discussion. When dissolved
in an
1
H
2
O
2
H
2
O mixture strongly hydrogen-bonded
protons within a protein become enriched in
1
H. The
1
H /
2
H ratio of the hydrogen-bonded protons provides
another measure of the hydrogen bond strength.
158c
A short hydrogen bond is also present in such
cations as the following:
The pK
a
for dissociation of its proton is 12.3 and the
hydrogen-bonded proton is probably located in the
center of the bond with both amino groups sharing
the charge.
160
Enols can also form unusually strong
resonance-assisted hydrogen bonds:
The structure can be thought of as a resonance-stabi-
lized enolate anion with a proton bound between the
two oxygen atoms and equidistant from them.
156,157
Do these short low-barrier hydrogen bonds
have a special significance in enzymology? Proposals
that they contribute to stabilization of transition
states
161 166
have received some support
154,167
and
aroused controversy.
158b,168 173
In later chapters we
will examine specific enzymes in which low-barrier
hydrogen bonds have been observed.
5. Covalent Catalysis
In addition to participating in acidbase catalysis,
some amino acid side chains may enter into covalent
bond formation with substrate molecules, a phenome-
non that is often referred to as covalent catalysis.
174
When basic groups participate this may be called
nucleophilic catalysis. Covalent catalysis occurs
frequently with enzymes catalyzing nucleophilic
displacement reactions and examples will be consid-
ered in Chapter 12. They include the formation of an
acyl-enzyme intermediate by chymotrypsin (Fig. 12-11).
Several of the coenzymes discussed in Chapters 14 and
15 also participate in covalent catalysis. These coen-
zymes combine with substrates to form reactive inter-
mediate compounds whose structures allow them to
be converted rapidly to the final products.
6. Proximity and Orientation
One of the earliest ideas about enzymes was that
they simply brought reactants together and bound
them side by side for a long enough time that the
reactive groups might bump together and finally react.
How important is this proximity factor? Page and
Jencks estimated that rate enhancements by factors of
10
3
or more may be expected solely from the loss in the
entropy of two reactants when they are bound in close
proximity on an enzyme surface.
107,175,176
In view of
the large entropy decrease involved, the enthalpy of
O H O
H
H
H
0.276 nm
in ice
O
C C
O O
O
H
Monohydrogen maleate

O
C C
O O
O
H

N
H
N
CH
3
H
3
C
H
3
C CH
3
+
O
C
C
C
O
H
H
3
C CH
3
O
C
C
C
O
H
CH
3
H
3
C
H H
495
binding must be high, and if this explanation is correct
the binding of the substrates to the enzyme provides
much of the driving force for catalysis. Westheimer
described this by stating that enzymes use the substrate-
binding force as an entropy trap.
108
The losses of
translational and rotational entropy, which Page and
Jencks estimated as up to 160 to 210 J /deg /mol,
overcome the unfavorable entropy of activation that
is usual in bimolecular reactions.
How precise must the orientation of substrates be
for rapid reaction?
107,177,177a
Compounds such as the
acid shown in Eq. 9-103 form an internal ester (lac-
tone) spontaneously with elimination of water.
However, the following compound reacts at a rate
over 10
11
times that of the acid shown in Eq. 9-102.
This is presumably because its conformation is highly
restricted and the COOH is constrained to frequently
collide with the OH group.
178,179
The three methyl
groups interdigitate and form a trialkyl lock. Orientation
must also play a large role in enzymatic catalysis. As
previously emphasized, enzymes often orient substrates
precisely by formation of multiple hydrogen bonds.
Small distortions by mutation or substitution of an
essential metal by a different one can have very great
effects. For example, because of an altered coordination
pattern isocitrate dehydrogenases with Ca
2+
in the active
site has a maximum velocity of catalysis only 2.5 x 10
3
that with the normal Mg
2+
-containing enzyme.
180
Bruice and associates concluded that enzymes must
bring reacting groups into close proximity with orbitals
of the reactants properly aligned in the ground state
prior to moving to the transition state. They suggested
that enzymes preorganize the enzyme-substrate com-
plex into a near attack conformation in which the
positions of reacting groups, the arrangements of
hydrogen bonds, and the local dielectric constant in
the active site, resemble those in the transition state.
In this conformation the energy barrier to the transition
state may be very low.
128-128b
Preorganization of the
complex also acts to eliminate the slow components of
solvent reorganization required for reaction in aqueous
solutions.
128c,128d
The necessity for reacting groups of substrates
to collide with an orientation that allows productive
interaction of electronic orbitals is often called a
stereoelectronic effect. An example is the addition
reaction of Eq. 9-74. The orbital of an unshared pair
of electrons on the HO

ion must be perpendicular


to the plane of the double bond. Furthermore, if the
proton becomes attached to the adjacent carbon in a
synchronous or concerted manner it must enter from
the opposite side, as it does in Eq. 9-74.
In many biochemical reactions an alcohol or amine
is eliminated from a tetrahedrally bonded intermediate
as in Eq. 9-104. Deslongchamps proposed a stereoelec-
tronic theory
181,182
according to which elimination of
either NH R
2
or O R
1
from this intermediate will
depend upon the values of the torsion angles
1
and
2
(Eq. 9-104). The theory asserts that for rapid elimination
of OR
1
(Eq. 9-104a), unshared electron pairs on both the
O

and N atoms must be antiperiplanar to the bond


being broken (see Figure 2-2). If rotation around the
C O bond (
1
) occurs an orientation can be found in
which an electron pair on each of the two oxygens
will be antiperiplanar to the bond to NHR
2
. From this
orientation the latter will be eliminated (Eq. 9-104b).
The theory has been supported by much experimental
evidence involving reaction rates and product distri-
bution among competing reactions of small conforma-
tionally restricted organic molecules.
181 184
Although
more recent experiments
185
suggest that stereoelec-
tronic factors may be of less significance than had
been assumed, even a small decrease in transition
state energy can be significant in an enzyme-catalyzed
reaction. Enzymes may not only orient substrates in
accord with stereoelectronic principles but also be able
to promote conformational alterations in intermediates
that allow them to take advantage of stereoelectronic
factors. Examples are considered in Chapter 12.
7. The Microenvironment
A substrate bound at an active site may be in an
environment quite different than that in an ordinary
OH
COOH
O O
H
2
O
(9-103)
COOH
OH
CH
3
CH
3
CH
3
Trialkyl lock
D. Mechanisms of Catalysis
O
C
R
N
R
3
O
R
2
H
R
3
C NR
2
O

1
R
3
C OR
1
O

2
H
HOR
1
+
NHR
2
+
a
b
(9-104)
496 Chapter 9. Enzymes: The Catalysts of Cells
aqueous solution. In fact, the protein often surrounds
the substrate to the extent that the protein is the solvent.
What kind of microenvironment does the protein
provide and can it assist in the catalytic process?
Charged and dipolar groups of the protein provide
an electrostatic field that provides part of the binding
energy and which may also assist in catalysis, a con-
cept expressed by Quastel as early as 1926.
183a
The
ends of protein helices often seem to point at active
centers.
186,187
In many cases more than one N terminus
with its positive electrical potential (see Chapter 2) or
more than one C terminus (negative potential) of a
helix point to an active site. These helix dipoles may
be important in stabilizing transition state structures
188
or in altering pK
a
values of functional groups.
186
Fluctua-
tions in charge distribution within a protein and in the
hydrogen-bonding pattern of the protein with solvent
and substrate may also be important.
189
For some enzymatic reactions a transition state
will be favored by a medium of very low dielectric
constant and a correctly constructed active site can
provide just such a surrounding.
189a
Hydrophobic
groups may be packed around a site where an ion pair
or other ionic interaction between enzyme and substrate
occurs increasing the strength of that interaction.
Conformational changes may enhance such effects.
The dehydration of polar groups that must often occur
upon binding of substrate may make these groups
more reactive.
187,190,191
It has been suggested that the
substantial volume changes (V

) that sometimes occur


during formation of transition states for enzyme-
catalyzed reactions may result largely from changes
in hydration of groups on the enzyme surface and that
these changes may play an important role in catalysis.
In the past most enzymologists tacitly assumed
that the external medium in which enzymes act must
be aqueous. However, many enzymes function well
in media containing largely hydrocarbons. Enzymes
in a dry, powdered form have been suspended directly
in organic solvents.
192,193
Under these conditions,
enzymes may contain only tightly bound structural
water, together with less than one equivalent of a mono-
layer of water outside the protein. The enzymes often
remain active and are able to catalyze reactions with
an altered substrate specificity as well as different
reactions overall.
8. Strain and Distortion
The fact that enzymes appear to bind their sub-
strates in such a way as to surround and immobilize
them means that something other than the kinetic
energy of the substrate is needed to provide energy for
the ES complex to pass over the transition state barrier.
What is the source of this activation energy? As with
nonenzymatic reactions, it must come ultimately from
the translational energy of solvent or solute molecules
bombarding the complex. Can enzymes act as energy
funnels that effectively channel kinetic energy from
spots on the enzyme surface to the active site?
194
This
could either be through strictly mechanical movement
or through induction by fluctuations of the electrical
field.
195
It is often suggested that enzymes assist in catalysis
by distorting bond lengths or angles away from their
normal values. If the distorted structure were closer
to the transition state geometry than the undistorted
structure, catalysis would be assisted (see lysozyme,
Chapter 12). However, Levitt
196
concluded (see also
Fersht
2
) that forces provided this way by a protein are
small and that the protein would become distorted
rather than a substrate. On the other hand, if binding
of a substrate distorts the protein, could the resultant
stored energy be used in some way to assist in
catalysis? If binding of a substrate bends a spring
(Section C,4), can the tension in the spring then be used
to distort the enzyme to be more exactly complemen-
tary to the transition state? This concept has become
popular.
2,113,197
The amino acid side chains of proteins are usually
well packed. However, neither the side chains nor the
main chain are rigid and immobile. Some regions of
the protein will contain empty spaces packing defects.
Lumry called these mobile defects because, as a result
of fluctuations in side chain packing, they can move
within a protein from one site to another for a consid-
erable distance.
198
Much evidence, including X-ray
studies at low temperatures,
199
supports the existence
of many conformational substates in proteins. Some
substates may bind substrates better than others and
some may allow conversion to the transition state more
readily than do others. Rotation of histidine rings,
peptide linkages, OH groups, or amide side chains
199a
may be required and has, in fact, been observed for
some enzymes. Perhaps in one of the substates of an
enzymesubstrate complex an especially favorable
vibrational mode
200
leads the complex to the transition
state. In the transition state the packing of side chains
may be especially tight. A mobile defect present in the
active site may have moved elsewhere. The binding of
substrates to the protein in the transition state will also
be tighter because of the conformational alterations
that have occurred. The binding energy of the sub-
strates is now being utilized to lower the transition
state barrier. The substrate is literally squeezed into
the transition state configuration.
Is it possible that the protein domains forming an
active site act like ferroelectric crystals, which change
their dipole moment in response to a change in electric
field? The highly polarizable hydrogen-bonded net-
work, the amide linkages, imidazole rings, guanidinium
group, etc. of active sites may permit a flip-flop of the
dipole moment as in domains of ferroelectric crystals.
497
In such crystals, e.g., those of the hydrogen-bonded
KH
2
PO
4
, a 180 change of dipole-moment direction
results from very small movements of heavy atoms
together with larger movements of the hydrogen-
bonded protons.
200a,b,c
Could a similar flip-flop in a
protein domain be coupled to the passage of a substrate
over the transition state barrier? I have not seen any
discussion of this possibility, but the structure of pro-
tein domains would seem to allow it. One could also
imagine that with a small change in the hydrogen-
binding arrangement of protein groups an active site
could become preorganized to favor a flip-flop along
a hydrogen-bonded chain in a different direction for a
subsequent step in a reaction sequence.
9. Why Oligomeric Enzymes?
As we have seen (Chapter 7), a large fraction of all
proteins exist as dimers, trimers, and higher oligomers.
Oligomeric proteins raise the osmotic pressure much
less than would the same number of monomeric sub-
units and this may be crucial to a cell. Another advan-
tage of oligomers may be reflected in the fact that active
sites of enzymes are often at interfaces between two or
more subunits. This may enhance the ability of enzymes
to undergo conformational rearrangements that are
required during their action, just as hemoglobin changes
its oxygen affinity in concert with a change in inter-
subunit contacts (Fig. 7-25).
A curious observation is that crystals of the dimeric
pig heart malate dehydrogenase bind only one mole-
cule of the substrate NAD
+
per dimer tightly; the
second NAD
+
is bound weakly.
201,202
Similar anti-
cooperativity has been reported for other crystalline
dehydrogenases
203
and various other enzymes. An
intriguing idea is that anticooperativity in binding
might reflect a cooperative action between subunits
during catalysis. Suppose that only conformation A
of a dehydrogenase bound reduced substrate and
NAD
+
, while conformation B bound NADH and
oxidized substrate. If reduced substrate and NAD
+
were present in excess and if oxidized substrate were
efficiently removed from the scene by further oxidation,
the following cooperative events could occur in the
mixed AB dimer. The subunit of conformation A
would bind substrates, react, and be converted to
conformation B. At the same time, because of the
strong AB interaction, the subunit that was originally
in conformation B would be converted back to A and
would be ready to initiate a new round of catalysis.
Since conformation A has a low affinity for NADH,
dissociation of the reduced coenzyme, which is often
the slow step in dehydrogenase action, would be
facilitated.
204207
Such a reciprocating or flip-flop
mechanism, suggested first by Harada and Wolfe,
204
is attractive because it provides a natural basis for the
existence of the many known dimeric enzymes that do
not exhibit evident allosteric properties. Attempts
to verify this idea have largely failed. However,
recent crystallographic studies of both glyceraldehyde
phosphate dehydrogenase
206,207
and thymidylate
synthase
208
are consistent with the proposal. There is
still a possibility that coordinated reciprocal changes
in distribution of electrical charges in the two subunits
may also be important.
10. Summary
It appears that enzymes exert their catalytic
powers by first bringing together substrates and
binding them in proper orientations at the active site.
Second, they often provide acidic and basic groups
in the proper orientations to promote proton transfers
within the substrate. Third, groups within the enzyme
(especially nucleophilic groups) may enter into covalent
interaction with the substrates to form structures that
are more reactive than those originally present in the
substrate. Fourth, the protein often closes around the
substrate to immobilize it and to hold it in an environ-
ment, often lacking water, which could impede cataly-
sis. The enzyme is also probably able to make small
readjustments of its structure to provide good comple-
mentarity to the substrate at every stage and especially
to the transition state structure. Fifth, the enzyme may
be able to induce strain or distortion in the substrate
perhaps accompanying a conformational change within
the protein. The following question is often asked:
Why are enzymes such large molecules?
209
At least
part of the answer is that an enzyme usually interacts,
sometimes via special domains, with other proteins.
206
Another part of the answer is evident when we con-
sider that the formation of a surface complementary to
that of the substrate and possessing reasonable rigidity
requires a complex geometry in the peptide backbone.
In addition, the enzyme must provide functional groups
at the proper places to enter into catalysis. It may require
a certain bulk to provide a low dielectric medium.
Finally, if essential conformational changes occur
during the course of the catalysis, we can only be
surprised that nature has succeeded in packing so
much machinery into such a small volume.
E. Classification of Enzymes
An official commission of the International Union
of Biochemistry (IUB) has classified enzymes in the
following six categories:
210
1. Oxidoreductases. Enzymes catalyzing dehydro-
genation or other oxidation and reduction
reactions.
E. Classification of Enzymes
498 Chapter 9. Enzymes: The Catalysts of Cells
1. Fromm, H. (1975) Initial Rate Enzyme Kinetics,
Springer-Verlag, New York
2. Fersht, A. (1999) Structure and Mechanism in
Protein Science: A Guide to Enzyme Catalysis and
Protein Folding, Freeman, New York
3. Dixon, M., and Webb, E. C., eds. (1979)
Enzymes, 3rd ed., Academic Press, New York
4. Wharton, C. W., and Eisenthal, R. (1981)
Molecular Enzymology, Wiley, New York
5. Engel, P. C. (1982) Enzyme Kinetics, the Steady-
State Approach, Chapman and Hall, London
6. Hammes, G. G. (1982) Enzyme Catalysis and
Regulation, Academic Press, New York
7. Kull, F. J. (1994) Principles of Biomolecular
Kinetics and Binding, CRC Press, Boca Raton,
Florida
8. Suckling, C. J., ed. (1990) Enzyme Chemistry,
2nd ed., Chapman and Hall, New York
9. Price, N. C., and Stevens, L., eds. (1989)
Fundamentals of Enzymology, 2nd ed., Oxford
Univ. Press, Oxford,England
10. Kuby, S. A. (1990) Enzymes: a Comprehensive
Study, CRC Press, Boca Raton, Florida
11. Schulz, A. R. (1994) Enzyme Kinetics From
Diastase to Multi-enzyme Systems, Cambridge
Univ. Press, New York
12. Segel, I. H. (1975) Enzyme Kinetics:Behavior and
Analysis of Rapid Equilibrium and Steady-State
Enzyme Systems, Wiley, New York
13. Cornish-Bowden, A. (1995) Fundamentals of
Enzyme Kinetics, 2nd ed., Portland Press,
Brookfield, Vermont
13a. Purich, D. L., and Allison, R. D. (2000) Trends
Biochem. Sci. 25, 455
14. Lowry, O. H., and Passonneau, J. V. (1972) A
Flexible System of Enzymatic Analysis,
Academic Press, New York
15. Passonneau, J., and Lowry, O., eds. (1993)
Enzymatic Analysis, 1st ed., Humana Press,
Totowa, New Jersey
16. Liu, Y.-M., and Sweedler, J. V. (1995) J. Am.
Chem. Soc. 117, 88718872
17. Libecq, C., ed. (1992) Biochemical Nomencla-
ture, Portland Press, London and Chapel Hill,
North Carolina (for the International Union of
Biochemistry and Molecular Biology)
18. Caldin, E. F. (1964) Fast Reactions in Solution,
Wiley, New York
19. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York (p. 393)
20. Glick, N., Landman, A. D., and Roufogalis, B.
D. (1979) Trends Biochem. Sci. 4, N82 N83
20a. Wilkinson. (1980) Biochem. J. 80, 324
21. Orsi, B. A., and Tipton, K. E. (1979) Methods
Enzymol. 63, 159183
22. Wharton, C. W., and Szawelski, R. J. (1982)
Biochem. J. 203, 351360
23. Cleland, W. W. (1979) Methods Enzymol. 63,
103138
24. Frieden, C. (1994) Methods Enzymol. 240, 311
322
25. Schechter, A. N. (1970) Science 170, 273280
26. Cha, S. (1970) J. Biol. Chem. 245, 48144818
27. Martinez, M. B., Flickinger, M. C., and
Nelsestuen, G. L. (1996) Biochemistry 35,
11791186
28. Bull, H. B. (1971) An Introduction to Physical
Biochemistry, 2nd ed., Davis Co., Philadelphia,
Pennsylvania
29. Eisenstein, B. I. (1987) Escherichia coli and
Salmonella typhimurium, Am. Soc. Microbiology,
Washington, D.C., FC Niedhardt, ed. (pp.84
90)
30. Smoluchowski, M. (1917) Z. Phys. Chem 92,
129168
31. Caldin, E. F. (1964) Fast Reactions in Solution,
Wiley, New York (pp. 10 and 279)
32. Debye, P. (1942) Trans. Electrochem. Soc. 82,
265272
32a. Elcock, A. H., Huber, G. A., and McCammon,
J. A. (1997) Biochemistry 36, 1604916058
32b. Selzer, T. and Schreiber, G. (1999) J. Mol. Biol.
287, 409419
33. Mastro, A. M., Babich, M. A., Taylor, W. D.,
and Keith, A. D. (1984) Proc. Natl. Acad. Sci.
U.S.A. 81, 34143418
34. French, D. (1957) Brewers Digest 32, 5056
35. Cohn, E. J., and Edsall, J. T. (1943) Proteins,
Amino Acids and Peptides as Ions and Dipolar
Ions, Van Nostrand-Reinhold, Princeton, New
Jersey (pp. 9093)
36. Koenig, S. H. (1975) Biopolymers 14, 24212423
37. Blacklow, S. C., Raines, R. T., Lim, W. A.,
Zamore, P. D., and Knowles, J. R. (1988)
Biochemistry 27, 11581167
38. Albery, W. J., and Knowles, J. R. (1976)
Biochemistry 15, 56275631
39. Albery, W. J., and Knowles, J. R. (1976)
Biochemistry 15, 56315640
40. Jenkins, W. T. (1982) Adv. Enzymol. 53, 307344
41. Haldane, J. B. S. (1930) Enzymes, Longmans,
Green, New York
42. Cleland, W. W. (1970) in The Enzymes, 3rd ed.,
Vol. 2 (Boyer, P. D., ed), Academic Press, New
York
43. Dalziel, K. (1957) Acta Chem. Scand. 11, 1706
1723
44. Rebholz, K. L., and Northrop, D. B. (1994)
Arch. Biochem. Biophys. 312, 227233
45. Volkenstein, M. V., and Goldstein, B. N. (1966)
Biokhim. 31, 541547
46. Volkenstein, M. V., and Goldstein, B. N. (1966)
Biochim. Biophys. Acta. 115, 471477
47. Fromm, H. J. (1970) Biochem. Biophys. Res.
Commun. 40, 692697
References
48. Seshagiri, N. (1972) J. Theor. Biol. 34, 469486
49. Huang, C. Y. (1979) Methods Enzymol. 63, 5484
50. Chou, K., and Forsn, S. (1980) Biochem. J. 187,
829835
51. Fromm, H. J., Silverstein, E., and Boyer, P. D.
(1964) J. Biol. Chem. 239, 36453652
52. Fromm, S. J., and Fromm, H. J. (1999) Biochem.
Biophys. Res. Commun. 265, 448452
53. Silverstein, E., and Boyer, P. D. (1964) J. Biol.
Chem. 239, 39013907
54. Hurst, R. O. (1969) Can. J. Biochem. Physiol. 47,
941944
54a. Cha, S. (1968) J. Biol. Chem. 243, 820825
55. Srere, P. A. (1967) Science 158, 936937
56. Hiromi, K., ed. (1979) Kinetics of Fast Enzyme
Reaction-Theory and Practice, John Wiley, New
York
57. Rentzepis, P. M. (1978) Science 202, 174182
58. Hammes, G. G., and Schimmel, P. R. (1970) in
The Enzymes, 3rd ed., Vol. 2 (Boyer, P. D., ed),
pp. 67114, Academic Press, New York
59. Ahrens, M.-L., Maass, G., Schuster, P., and
Winkler, H. (1970) J. Am. Chem. Soc. 92, 6134
6139
59a. Gutfreund, H. (1999) Trends Biochem. Sci. 24,
457 460
59b. Beechem, J. M. (1998) Biophys. J. 74, 2141
60. Fink, A. L. (1979) Trends Biochem. Sci. 4, 810
61. Balashov, S. P., Imasheva, E. S., Ebrey, T. G.,
Chen, N., Menick, D. R., and Crouch, R. K.
(1997) Biochemistry 36, 86718676
62. Fink, A. L., and Geeves, M. A. (1979) Methods
Enzymol. 63, 336370
63. Alberty, R. A. (1956) Adv. Enzymol. 17, 164
64. Dixon, M., and Webb, E. C., eds. (1979)
Enzymes, 3rd ed., Academic Press, New York
(pp.138163)
65. Cleland, W. W. (1977) Adv. Enzymol. 45, 273
387
66. Cleland, W. W. (1982) Methods Enzymol. 87,
390405
67. Kyte, J. (1995) Mechanism in Protein Chemistry,
Garland Publ., New York
68. Ono, S., Hiromi, K., and Yashikawa, Y. (1958)
Bull. Chem. Soc. Jap. 31, 957962
69. Brocklehurst, K., and Dixon, H. B. F. (1976)
Biochem. J. 155, 6170
70. Tipton, K. F., and Dixon, H. B. F. (1979)
Methods Enzymol. 63, 183234
71. Dixon, M. (1953) Biochem. J. 55, 170171
72. Purich, L. D., and Fromm, H. J. (1972) Biochim.
Biophys. Acta. 268, 13
73. Williams, J. W., and Morrison, J. F. (1979)
Methods Enzymol. 63, 437467
74. Morrison, J. F. (1982) Trends Biochem. Sci. 7,
102105
75. Frieden, C. (1964) J. Biol. Chem. 239, 35223531
2. Transferases. These catalyze group transfer
reactions.
3. Hydrolases. Enzymes catalyzing transfer of
groups to the HO

ion of H
2
O.
4. Lyases. Enzymes promoting addition to double
bonds or the reverse.
5. Isomerases. Enzymes catalyzing rearrangement
reactions.
6. Ligases (synthetases). Enzymes that catalyze
condensation with simultaneous cleavage of ATP
and related reactions.
As an example, chymotrypsin is classified EC
3.4.4.5 according to the IUB system.
210
In this book
a more mechanistically based classification is used.
Because some official names are quite long, traditional
trivial names for enzymes often have been retained.
Remember that to be precise it is always necessary to
mention the species from which an enzyme was iso-
lated and, if possible, the strain. Also remember that
almost every significant genetic difference is reflected
in some change in some protein. It is possible that the
enzyme you are working with is slightly different from
the same enzyme prepared in a different laboratory.
499
References
76. Morrison, J. F., and Walsh, C. T. (1988) Adv.
Enzymol. 61, 201301
77. Gutheil, W. G., and Bachovin, W. W. (1993)
Biochemistry 32, 87238731
78. Di Cera, E., Hopfner, K.-P., and Dang, Q. D.
(1996) Biophys. J. 70, 174181
79. Botts, J., and Morales, M. (1953) Trans. Faraday
Soc. 49, 696707
80. Monod, J., Wyman, J., and Changeux, J. D.
(1965) J. Mol. Biol. 12, 88118
81. Plapp, B. V., Leidal, K. G., Smith, R. K., and
Murch, B. P. (1984) Arch. Biochem. Biophys. 230,
3038
82. Chadha, V. K., Leidal, K. G., and Plapp, B. V.
(1983) Journal of Medicinal Chemistry 26, 916
922
83. Cho, H., Ramaswamy, S., and Plapp, B. V.
(1997) Biochemistry 36, 382389
84. Westley, A. M., and Westley, J. (1996) J. Biol.
Chem. 271, 53475352
85. Herv, G., ed. (1989) Allosteric Enzymes, CRC
Press, Boca Raton, Florida
86. Rubin, M. M., and Changeux, J. D. (1966) J.
Mol. Biol. 21, 265274
87. Grant, G. A., Schuller, D. J., and Banaszak, L.
J. (1996) Protein Sci. 5, 3441
88. Williams, R. O., Young, J. R., and Majiwa, P.
A. O. (1979) Nature (London) 282, 847849
89. Rabin, B. R. (1967) Biochem. J. 102, 22c
90. Newsholme, E. A., and Start, C. (1973)
Regulation in Metabolism, Wiley, New York
91. Walsh, C. (1979) Enzymatic Reaction
Mechanism, Freeman, San Francisco,
California
91a. Plapp, B. V. (1982) Methods Enzymol. 87, 469
499
92. Sandler, M., ed. (1980) Enzyme Inhibitors as
Drugs, Univ. Park Press, Baltimore, Maryland
93. Bey, P. (1981) Chem. Ind. (London), 139144
94. Hanson, K. R. (1966) J. Am. Chem. Soc. 88,
27312742
94a. Prelog, V., and Helmchen, G. (1982) Angew.
Chem. Int. Ed. Engl. 21, 567583
95. Bentley, R. (1969) Molecular Asymmetry in
Biology, Vol. 1, Academic Press, New York
(pp. 4956)
96. Bentley, R. (1970) Molecular Asymmetry in
Biochemistry, Vol. 2, Academic Press, New
York
97. Alworth, W. L. (1972) Stereochemistry and its
Application in Biochemistry, Wiley-Interscience,
New York
98. Bentley, R. (1978) Nature (London) 276, 673676
99. Barry, J. M. (1997) Trends Biochem. Sci. 22, 228
230
100. Hirschmann, H. (1960) J. Biol. Chem. 235,
27622767
101. Post, C. B., and Ray, W. J. J. (1995) Biochemistry
34, 1588115885
102. Tsou, C.-L. (1986) Trends Biochem. Sci. 11, 427
429
103. Ferscht, A. R. (1980) Trends Biochem. Sci. 5,
262265
104. Fersht, A. R. (1998) Science 280, 541
105. Nureki, O., Vassylyev, D. G., Tateno, M.,
Shimada, A., Nakama, T., Fukai, S., Konno,
M., Hendrickson, T. L., Schimmel, P., and
Yokoyama, S. (1998) Science 280, 578582
106. Hopfield, J. J. (1980) Proc. Natl. Acad. Sci.
U.S.A. 77, 52485252
107. Jencks, W. P. (1975) Adv. Enzymol. 43, 219410
108. Westheimer, F. H. (1962) Adv. Enzymol. 24,
441482
109. Kraut, J. (1988) Science 242, 533539
110. Fersht, A. (1985) Enzyme Structure and
Mechanism, 2nd ed., Freeman, New York
111. Gandour, R. D., and Schowen, R. L., eds.
(1978) Transition States of Biochemical Processes,
Plenum, New York
112. Laidler, K. J. (1969) Theories of Chemical
Reaction Rates, McGraw-Hill, New York
113. Kyte, J. (1995) Mechanism in Protein Chemistry,
Garland Publ., New York (pp. 199213)
114. Marcus, R. A. (1992) Science 256, 15231524
115. Pauling, L. (1948) Nature (London) 161, 707713
116. Linehard, G. E. (1973) Science 180, 149154
117. Wolfenden, R. (1976) Annu Rev Biophys Bioeng.
5, 271306
117a. Schramm, V. L. (1998) Ann. Rev. Biochem. 67,
693720
118. Nielsen, P. A., Glad, S. S., and Jensen, F. (1996)
J. Am. Chem. Soc. 118, 1057710583
119. Zheng, Y.-J., and Bruice, T. C. (1997) J. Am.
Chem. Soc. 119, 81378145
120. Merkler, D. J., Kline, P. C., Weiss, P., and
Schramm, V. L. (1993) Biochemistry 32, 12993
13001
121. Degano, M., Almo, S. C., Sacchettini, J. C., and
Schramm, V. L. (1998) Biochemistry 37, 62776285
122. Glad, S. S., and Jensen, F. (1997) J. Am. Chem.
Soc. 119, 227232
123. Schramm, V. L., Horenstein, B. A., and Kline,
P. C. (1994) J. Biol. Chem. 269, 1825918262
124. Kline, P. C., and Schramm, V. L. (1994) J. Biol.
Chem. 269, 2238522390
125. Deng, H., Kurz, L. C., Rudolph, F. B., and
Callender, R. (1998) Biochemistry 37, 49684976
125a. Cho, Y.-K., and Northrop, D. B. (1999)
Biochemistry 38, 7470 7475
125b. Wolfenden, R., Snider, M., Ridgway, C., and
Miller, B. (1999) J. Am. Chem. Soc. 121, 7419
7420
126. Menger, F. M. (1992) Biochemistry 31, 53685373
127. Goldsmith, J. O., and Kuo, L. C. (1993) J. Biol.
Chem. 268, 1848118484
128. Lightstone, F. C., and Bruice, T. C. (1996) J.
Am. Chem. Soc. 118, 25952605
128a. Bruice, T. C., and Lightstone, F. C. (1999) Acc.
Chem. Res. 32, 127136
128b. Bruice, T. C., and Benkovic, S. J. (2000)
Biochemistry 39, 62676274
128c. Cannon, W. R., Singleton, S. F., and Benkovic,
S. J. (1996) Nature Struct. Biol. 3, 821833
128d. Cannon, W. R., and Benkovic, S. J. (1998) J.
Biol. Chem. 273, 2625726260
129. Murphy, D. J. (1995) Biochemistry 34, 45074510
129a. Warshel, A. (1998) J. Biol. Chem. 273, 27035
27038
129b. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York (pp.
210213)
130. Hucho, F., and Wallenfels, K. (1971) Eur. J.
Biochem. 23, 489496
131. Jencks, W. P. (1969) Catalysis in Chemistry and
Enzymology, McGraw-Hill, New York (pp.
170199)
132. Swain, C. G., and Brown, J. F., Jr. (1952) J. Am.
Chem. Soc. 74, 25342537 and 25382543
133. Rony, P. R. (1969) J. Am. Chem. Soc. 91, 6090
6096
134. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York (p. 199)
135. Engdahl, K.-., Bivehed, H., Ahlberg, P., and
Saunders, W. H., Jr. (1983) J. Am. Chem. Soc.
105, 47674774
136. Hegarty, A. F., and Jencks, W. P. (1975) J. Am.
Chem. Soc. 97, 71887189
136a. Williams, A. (1999) Concerted Organic and Bio-
Organic Mechanisms, CRC Press, Boca Raton,
Florida
137. Wang, J. H. (1968) Science 161, 328334
138. Eigen, M. (1964) Angew. Chem. Int. Ed. Engl. 3,
119
139. Metzler, D. E. (1979) Adv. Enzymol. 50, 140
140. Nagle, J. F., and Morowitz, H. J. (1978) Proc.
Natl. Acad. Sci. U.S.A. 75, 298302
141. Gerlt, J. A., and Gassman, P. G. (1993) J. Am.
Chem. Soc. 115, 1155211568
142. Cohen, A. O., and Marcus, R. A. (1968) J. Phys.
Chem. 72, 42494256
143. Marcus, R. A. (1969) J. Am. Chem. Soc. 91,
72247225
144. Guthrie, J. P. (1996) J. Am. Chem. Soc. 118,
1287812885
145. Guthrie, J. P. (1996) J. Am. Chem. Soc. 118,
1288612890
146. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York
(pp. 207213)
147. Dewar, M. J. S. (1984) J. Am. Chem. Soc. 106,
209219
148. Gandour, R. D., Maggiora, G. M., and
Schowen, R. L. (1974) J. Am. Chem. Soc. 96,
69676979
149. Quinn, D. M. (1987) Chem. Rev. 87, 955979
150. Klinman, J. P. (1989) Trends Biochem. Sci. 14,
368373
151. Rucker, J., Cha, Y., Jonsson, T., Grant, K. L.,
and Klinman, J. P. (1992) Biochemistry 31,
1148911499
152. Hwang, J.-K., and Warshel, A. (1996) J. Am.
Chem. Soc. 118, 1174511751
153. Barbara, P. F., Walker, G. C., and Smith, T. P.
(1992) Science 256, 975981
154. Kearley, G. J., Fillaux, F., Baron, M.-H.,
Bennington, S., and Tomkinson, J. (1994)
Science 264, 12851289
154a. Antoniou, D., and Schwartz, S. D. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 1236012365
154b. Basran, J., Sutcliffe, M. J., and Scrutton, N. S.
(1999) Biochemistry 38, 32183222
154c. Sutcliffe, M. J., and Scrutton, N. S. (2000)
Trends Biochem. Sci. 25, 405408
154d. Huang, Y., Rettner, C. T., Auerbach, D. J., and
Wodtke, A. M. (2000) Science 290, 111114
155. Hibbert, F., and Emsley, J. (1990) Adv. Phys.
Org. Chem. 26, 255379
156. Gilli, P., Bertolasi, V., Ferretti, V., and Gilli, G.
(1994) J. Am. Chem. Soc. 116, 909915
157. Perrin, C. L. (1994) Science 266, 16651668
158. Garcia-Viloca, M., Gonzlez-Lafont, A., and
Lluch, J. M. (1997) J. Am. Chem. Soc. 119,
10811086
158a. Bao, D., Huskey, W. P., Kettner, C. A., and
Jordan, F. (1999) J. Am. Chem. Soc. 121, 4684
4689
158b. Garcia-Viloca, M., Gelabert, R., Gonzlez-
Lafont, A., Moreno, M., and Lluch, J. M.
(1998) J. Am. Chem. Soc. 120, 1020310209
158c. Bowers, P. M., and Klevit, R. E. (2000) J. Am.
Chem. Soc. 122, 10301033
159. McDermott, A., and Ridenour, C. F. (1996) in
Encyclopedia of NMR, (Emsley, J. W., ed.), pp.
38203824, Wiley, Sussex, England
160. Frey, P. A. (1995) Science 269, 104106
161. Cleland, W. W., Frey, P. A., and Gerlt, J. A.
(1998) J. Biol. Chem. 273, 2552925532
162. Cleland, W. W., and Kreevoy, M. M. (1994)
Science 264, 18871890
163. Frey, P. A., Whitt, S. A., and Tobin, J. B. (1994)
Science 264, 19271930
164. Cassidy, C. S., Lin, J., and Frey, P. A. (1997)
Biochemistry 36, 45764584
165. Halkides, C. J., Wu, Y. Q., and Murray, C. J.
(1996) Biochemistry 35, 1594115948
166. Zheng, Y.-J., and Bruice, T. C. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 42854288
167. Richard, J. P. (1998) Biochemistry 37, 43054309
168. Scheiner, S., and Kar, T. (1995) J. Am. Chem.
Soc. 117, 69706975
169. Warshel, A., Papazyan, A., Kollman, P. A.,
Cleland, W. W., Kreevoy, M. M., and Frey, P.
A. (1995) Science 269, 102106
170. Shan, S.-o, Loh, S., and Herschlag, D. (1996)
Science 272, 97101
171. Ash, E. L., Sudmeier, J. L., De Fabo, E. C., and
Bachovchin, W. W. (1997) Science 278, 11281132
500 Chapter 9. Enzymes: The Catalysts of Cells
172. Shan, S.-o, and Herschlag, D. (1996) J. Am.
Chem. Soc. 118, 55155518
173. Warshel, A., and Papazyan, A. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1366513670
174. Spector, L. B. (1982) Covalent Catalysis by
Enzymes, Springer-Verlag, New York
175. Page, M. I., and Jencks, W. P. (1971) Proc. Natl.
Acad. Sci. U.S.A. 68, 16781683
176. Kirsch, J. F. (1973) Ann. Rev. Biochem. 42, 205
234
177. Koshland, D. E., Jr., Carraway, K. W., Dafforn,
G. A., Goss, J. D., and Storm, D. R. (1971) Cold
Spring Harbor Symp. Quant. Biol. 36, 1320
177a. Vill, J., Strajbl, M., Glennon, T. M., Sham, Y.
Y., Chu, Z. T., and Warshel, A. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 1189911904
178. Milstien, S., and Cohen, L. A. (1972) J. Am.
Chem. Soc. 94, 91589165
179. Karle, J. M., and Karle, I. L. (1972) J. Am.
Chem. Soc. 94, 91829189
180. Mesecar, A. D., Stoddard, B. L., and Koshland,
D. E., Jr. (1997) Science 277, 202206
181. Deslongchamps, P., Lebreux, C., and Taillefer,
R. (1973) Can. J. Chem. 51, 16651669
182. Deslongchamps, P. (1975) Tetrahedron 31,
24632490
183. Perrin, C. L., and Arrhenius, M. L. (1982) J.
Am. Chem. Soc. 104, 28392842
184. Evans, C. M., Glenn, R., and Kirby, A. J. (1982)
J. Am. Chem. Soc. 104, 47064707
185. Kuo, L. C., Fukuyama, J. M., and Makinen, M.
W. (1983) J. Mol. Biol. 163, 63105
186. Plogman, J. H., Drenth, G., Kalk, K. H., and
Hol, W. G. J. (1979) J. Mol. Biol. 127, 149162
187. Warshel, A. (1981) Biochemistry 20, 31673177
188. Hol, W. G. J., van Duijen, P. T., and Berendsen,
H. J. C. (1978) Nature (London) 273, 443446
189. Welch, G. R., ed. (1986) The Fluctuating
Enzyme, Wiley, New York
189a. Mertz, E. L., and Krishtalik, L. I. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 20812086
190. Dewar, M. J. S., and Storch, D. M. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 22252229
191. Dewar, M. J. S., and Dieter, K. M. (1988)
Biochemistry 27, 33023308
192. Yennawar, H. P., Yennawar, N. H., and Farber,
G. K. (1995) J. Am. Chem. Soc. 117, 583585
193. Schmitke, J. L., Wescott, C. R., and Klibanov,
A. M. (1996) J. Am. Chem. Soc. 118, 33603365
194. Kell, D. B. (1982) Trends Biochem. Sci. 7, 1
195. Robertson, B., and Astumian, R. D. (1992)
Biochemistry 31, 138141
196. Levitt, M. (1974) in Peptides, Polypeptides and
Proteins (Blout, E. R., Bouey, F. A., Goodman,
M., and Lotan, N., eds), p. 99, Wiley, New York
197. Avis, J. M., and Fersht, A. R. (1993)
Biochemistry 32, 53215326
198. Lumry, R., and Rosenberg, A. (1974) Colloques
Internationaux du C.N.R.S. 246, 5362
199. Rader, S. D., and Agard, D. A. (1997) Protein
Sci. 6, 13751386
199a. Word, J. M., Lovell, S. C., Richardson, J. S.,
and Richardson, D. C. (1999) J. Mol. Biol. 285,
17351747
200. Bialek, W., and Onuchic, J. N. (1988) Proc.
Natl. Acad. Sci. U.S.A. 85, 59085912
200a. Bystrov, D. S., and Popova, E. A. (1987)
Ferroelectrics 72, 147155
200b. Emsley, J., Reza, N. M., Dowes, H. M.,
Hursthouse, M. B., and Kurodo, R. (1988)
Phosphorus and Sulfur 35, 141149
200c. Rakvin, and Dalal. (1992) Ferroelectrics 135,
227236
201. Banaszak, L. A., and Bradshaw, R. A. (1975) in
The Enzymes, 3rd ed., Vol. 11 (Boyer, P. D., ed),
Academic Press, New York (pp. 369396)
202. Zimmerle, C. T., and Alter, G. M. (1993)
Biochemistry 32, 1274312748
References
203. Niefind, K., Hecht, H.-J., and Schomburg, D.
(1995) J. Mol. Biol. 251, 256281
204. Harada, K., and Wolfe, R. G. (1968) J. Biol.
Chem. 243, 41314137
205. Lazdunski, M., Petitclerc, C., Chappelet, D.,
and Lazdunski, C. (1971) Eur. J. Biochem. 20,
124139
206. Skarzynski, T., Moody, P. C. E., and Wonacott,
A. J. (1987) J. Mol. Biol. 193, 171187
207. Yun, M., Park, C.-G., Kim, J.-Y., and Park, H.-
W. (2000) Biochemistry 39, 1070210710
208. Anderson, A. C., ONeil, R. H., DeLano, W. L.,
and Stroud, R. M. (1999) Biochemistry 38,
1382913836
209. Srere, P. A. (1984) Trends Biochem. Sci. 9, 387
390
210. Nomenclature Committee of the International
Union of Biochemistry and Molecular
Biology. (1992) Enzyme Nomenclature,
Academic Press, San Diego, California
501
Study Questions
1. Give two reasons why enzymes are important to
living organisms.
2. If an enzyme catalyzes the reaction A B + C, will
it also catalyze the reaction B + C A?
3. Can an enzyme use a site to catalyze a reverse reac-
tion different from what it uses to catalyze the reaction
in the forward direction? Explain your answer.
4. Define the steady state of living cells and con-
trast with a state of chemical equilibrium.
5. How is the instantaneous initial velocity of an
enzymatic reaction measured? What precautions
must be taken to ensure that a true instantaneous
velocity is being obtained?
6. How is the rate of an enzyme action influenced by
changes in:
a) temperature
b) enzyme concentration
c) substrate concentration
d) pH
7. In what ways are enzymatic reactions typical of
ordinary catalytic reactions in organic or inorganic
chemistry, and in what ways are they distinct?
8. In general, for a reaction that can take place with
or without catalysis by an enzyme, what is the
effect, if any, of the enzyme on
a) standard Gibbs energy change of the reaction
b) energy of activation of the reaction
c) initial velocity of the reaction
d) temperature coefficient of the rate constant
9. What are the dimensions of the rate constant for
zero-, first-, and second-order reactions? If a first-
order reaction is half completed in 2 min, what is
its rate constant?
10. A sample of hemin containing radioactive iron,
59
Fe, was assayed with a Geiger-Mller counter at
intervals of time with the following results:
0 3981
2 3864
6 3648
10 3437
14 3238
20 2965
Determine the half-life of
59
Fe and the value for
the decay constant.
11. The kinetics of the aerobic oxidation of enzymati-
cally reduced nicotinamide adenine dinucleotide
(NADH) have been investigated at pH 7.38 at
30C. The reaction rate was followed spectropho-
tometrically by measuring the decrease in absor-
bance at 340 nm over a period of 30 min. The
reaction may be represented as
NADH + H
+
+ riboflavin
NAD
+
+ dihydroriboflavin
1 0.347
2 0.339
5 0.327
9 0.302
16.5 0.275
23 0.254
27 0.239
30 0.229
Determine the rate constant and the order of the
reaction.
12. You have isolated and purified a new enzyme (E)
which converts a single substrate (S) into a single
product (P). You have determined M
r
by gel
filtration as ~ 46,400. However, in SDS gel electro-
phoresis, a molecular mass of ~ 23 kDa was indi-
cated for the single protein band observed. A
solution of the enzyme was analyzed in the fol-
lowing way. The absorbance at 280 nm was found
to be 0.512. A 1.00 ml portion of the same solution
was subjected to amino acid analysis and was
found to contain 71.3 nmol of tryptophan. N-
terminal analysis on the same volume of enzyme
revealed 23.8 nmol of N-terminal alanine.
a) What is the approximate molecular mass of the
enzyme? Discuss this answer. Be sure to use
an appropriate number of significant figures in
this and other calculations.
b) What is the concentration of your enzyme in
moles per liter of active sites?
c) What is the molar extinction coefficient at 280 nm
where A = cl; A = absorbance, c = mol / liter,
and l = cell width in cm. Assume that all
spectrophotometric measurements are made
in 1.00 cm cuvettes.
A second preparation of the enzyme had an absor-
bance at 280 nm of 0.485. This enzyme was dilut-
ed very carefully: 1.00 ml into 250 ml and this
diluted enzyme was used for the following experi-
ments (I to III).
Radioactive counts per min Time (days)
A (at 340 nm) Time (days)
502 Chapter 9. Enzymes: The Catalysts of Cells
Study Questions
Experiment I. A 1.00 ml portion of the diluted
enzyme was added to 250 ml of buffered substrate
at pH 7.0 and was mixed rapidly. The resulting
initial substrate concentration [ S ]
o
was 1.000 mM.
This reaction mixture was held at 25.0C and
portions were removed periodically at time t for
analysis of the product P formed. The results
follow. Plot [ P ] vs. time.
200 0.104 2800 0.070
400 0.208 3200 0.040
800 0.392 3600 0.022
1200 0.554 4400 0.0060
1600 0.695 6000 0.00048
2000 0.800
2400 0.881
The integrated rate equation corresponding to the
Michaelis Menten equation
v = V
max
[ S ] / (K
m
+ [ S ]) is as follows:
V
max
t = [ S ]
o
- [ S ] + K
m
ln ([ S ]
o
/ [ S ])
d) Plot ([ S ]
o
- [ S ])/ t vs. l / t x ln ([ S ]
o
/ [ S ]). From
this plot evaluate K
m
and V
max
. Make this and
other plots to appropriate scale on good quality
graph paper.
e) What is k
cat
?
Experiment II. In a second experiment, a series of
test tubes were set up, each containing a different
amount of buffered substrate at pH 7 but each in a
volume of exactly 4.00 ml. The same enzyme
solution used in part d (absorbance at 280 nm =
0.485) was diluted 2.00 ml in 250 ml as in I, then
again 2.00 ml in 200 ml. Portions of 1.00 ml of this
twice diluted enzyme were added at t = 0 to each
of the test tubes of buffered substrate. The reac-
tion was stopped in just 10.0 minutes by adding
perchloric acid; a suitable reagent was added to
provide for a colorimetric determination of the
product. The results were as follows:
10.0 2.29 0.600 1.31
5.00 2.18 0.400 1.07
2.50 2.00 0.200 0.686
1.200 1.69 0.100 0.400
0.800 1.48
f) Plot l / v vs. l / [ S ] where v is in units of mol
per tube and [ S ] in millimoles / liter. Evaluate
K
m
, V
max
, and k
cat
from this plot. Fit your data
by eye. It is generally agreed that a linear least
squares fit is inappropriate unless suitable
weighting factors are used.
g) Plot the same data as v / [ S ] vs. v. Again
evaluate K
m
and V
max
.
Experiment III. The preceding experiment was
repeated but an inhibitor was present in each tube
in a concentration equal to 5.00 mM, 10.0 mM, or
25.0 mM. Two different inhibitors were used, A
and B. The following results were obtained.
5.00 2.00 1.09 1.50 0.727
2.50 0.71 1.00 1.09 0.667
1.20 1.31 0.848 0.686 0.565
0.800 1.07 0.739 0.505 0.492
0.600 0.900 0.654 0.400 0.436
0.400 0.686 0.533 0.282 0.356
0.200 0.400 0.343 0.150 0.229
0.100 0.218 0.200 0.077 0.133
h) Plot l/v vs. l / [ S ] for each of these sets of data.
For each case evaluate V
max
, apparent K
m
, and
inhibitor constants K
I
= [ I ] [ E ]/ [ EI ]. There may
be two K
I
values.
i) For uninhibited enzymes, when [ S ] = 0.4 mM
what fraction of the enzyme is ES? Free E?
j) For enzyme in the presence of 10.00 mM inhibitor
and 0.8 mM substrate, what fraction of the total
enzyme is ES? EI? free E?
k) Recall that v = [ ES ] k
cat
= [ E ][ S ] k
cat
/K
m
in many
cases. What will be the relative rates of product
formation from your substrate S and another
competing substrate S present at the same concen-
tration which also reacts with your enzyme with
K
m
= 0.01 mM and k
cat
= 200 / s if [ S ] = [ S ]?
13. Using the method of graphs, write the initial rate
equation for the following system with A, B, P, and
Q present.
Odd-numbered rate constants are for forward
reactions; even-numbered constants are for
reverse.
Time t (s) [P] (mM) Time t (s) Remaining [S] (mM)
Amount of product
(mol/tube)
[S]
(mM)
Amount of product
(mol/tube)
[S]
(mM)
EXY EQ E EA E
A
k
1
k
2
B
k
3
k
4
P
k
5
k
6
Q
k
7
k
8
[I] =
[S]
(mM)
mol of product formed in 10 min/tube
5.00 mM 5.00 mM 25.0 mM 10.0 mM
Inhibitor: B A Inhibitor: B A
503
Study Questions
a) Satisfy yourself that
The determinants (given by the method of graphs)
which provide the steady-state concentrations of
[E] and of the various exzymesubstrate and
enzymeproduct complexes are
[E] = k
2
k
7
[k
4
+ k
5
] + k
3
k
5
k
7
[B] + k
2
k
4
k
6
[P]
[EA] = k
1
k
7
[k
4
+ k
5
][A] + k
1
k
4
k
6
[A][P] + k
4
k
6
k
8
[P][Q]
[EXY] = k
1
k
3
k
7
[A][B] + k
2
k
6
k
8
[P][Q] + k
1
k
3
k
6
[A][B][P]
+ k
3
k
6
k
8
[B][P][Q]
[EQ] = k
2
k
8
[ k
4
+ k
5
][Q] + k
1
k
3
k
5
[A][B] + k
3
k
5
k
8
[B][Q]
b) To obtain an expression for v
f
the expression
for d[ A ]/ dt may be multiplied by [ E ]
t
and
divided by [ E ] + [ EA ] + [ EXY ] + [ EQ ] = [ E ]
t
.
Then the above determinants may be substituted.
c) What is V
max,forward
and V
max,reverse
? HINT:
V
max,forward
is obtained when [P] = [Q] = 0 and
[A] = [B] = . What are K
mA
and K
mB
? K
mA
is
obtained when [P] = [Q] = 0, [B] = , and v =
1/2 V
max,forward
.
d) With a knowledge of the kinetic parameters,
indicate how the eight rate constants may be
obtained if the total concentration of enzyme,
[E]
t
, is known.
14. Anticooperativity was observed in the plot of
velocity vs. substrate concentration for an enzyme.
Can this observation be explained by the Monod
Wyman Changeux model for oligomeric enzymes?
By the model of Koshland? Explain.
15. Interpret, for each of the following cases, the curve
showing measured initial velocity at constant
substrate concentration (not maximum velocity)
against pH for an enzyme-catalyzed reaction.
a) The substrate is neutral and contains no acidic
or basic groups. The Michaelis constant is
found to be independent of pH over the range
studied.
b) The substrate is neutral and contains no acidic
or basic groups, and the maximum velocity is
found to be independent of pH.
c) The substrate is an -amino acid, and the
maximum velocity is found to be independent
of pH.
6 7 8 9 10
v
pH
d[A]
dt
= k
1
[A][E] k
2
[EA]
d[B]
dt
= k
3
[B][EA] k
4
[EXY] =
d[P]
dt
= k
5
[EXY] k
6
[P][EQ] =
d[Q]
dt
= k
7
[EQ] k
8
[E][Q] =
504 Chapter 10. An Introduction to Metabolism
Contents
A. The Starting Materials
1. Digestion
2. Sources of Energy
B. Catabolism and the Synthesis of ATP
1. Priming or Activation of Metabolites
2. Interconversions of Sugar Phosphates
3. Glycolysis and Fermentation
4. Pyruvate Dehydrogenase
5. Beta Oxidation
6. The Electron Transport Chain, Oxidative
Phosphorylation
7. The Citric Acid Cycle
C. Biosynthesis
1. Reversing Catabolic Pathways
2. Photosynthesis
D. Synthesis and Turnover of Macromolecules
1. Folding and Maturation of Proteins
2. Transport of Proteins to Their
Destinations within a Cell
Signal sequences and translocation
Ticketing destinations
Vesicular transport and the Golgi system
3. Posttranslational Alterations
Proteolytic processing
Altered ends
4. Intracellular Degradation of Proteins
5. Turnover of Nucleic Acids
E. Classifying Enzymatic Reactions
References
Study Questions
505
505
507
507
507
508
508
511
511
512
515
515
515
517
517
518
519
519
520
521
521
522
522
523
527
530
531
533
Boxes
Box 10-A An Early Labeling Experiment
Box 10-B Discovery of the Citric Acid Cycle
Box 10-C Ubiquitin
Box 10-D Drawing Those Little Arrows
Tables
Table 10-1 Types of Biochemical Reactions with
Ionic Mechanisms
513
517
524
528
526
This electron micrograph of a thin section of pancreatic epithelial cells shows parts of two
secretory cells that are synthesizing proenzymes (zymogens). Nuclei (N) are seen at the top
center and lower right. Numerous ribosomes (barely seen here) line the many membranes of
the endoplasmic reticulum (ER) and are most abundant near lateral and basal (left) surfaces of
the cells. A few mitochondria (M) are present. The synthesized proteins move toward the apical
surfaces (a small piece is seen at upper right), passing through the Golgi region (G) and accumu-
lating in zymogen granules (Z) before secretion. From Porter, K. R. and Bonneville, M. A., Fine
Structure of Cells and Tissues, Lea and Febiger, Philadelphia 1973. Courtesy of Mary Bonneville.
N
505
The first section of this book has dealt with the
basic structures and with many of the complexities of
the materials formed by living cells. In the next major
section we will look at the chemical reactions that build
and maintain a cell and that permit it to grow and to
be responsive to external stimuli. These reactions are
organized into metabolic sequences or pathways that
form a complex, branched, and interconnected network.
It would be pointless to try to memorize all of them.
However, at this point it will be worthwhile to consider
the significance of a few of the major sequences, which
describe central pathways of metabolism.
Beginning in the nineteenth century, the investigation
of these pathways and of the associated enzymes has
provided much of our knowledge about the chemistry
of living things. The protein products that are encoded
in the structural genes that direct these pathways make
up a large fraction of the material in a cell. It will be
well worth the readers effort to examine the chemistry
of these metabolic sequences in detail. Regulatory
mechanisms that are applicable to them, and to other
pathways, are described in Chapter 11 and chemical
details of enzyme action are considered in Chapters
1216. Then, in Chapter 17, the chemical logic of the
reaction sequences is considered in more detail.
A. The Starting Materials
All cells of all organisms take in chemical starting
materials and give off chemical products. They all
have a source of energy and generate heat during their
metabolism. They all synthesize complex organic
substances and maintain a high degree of organization,
that is, a state of relatively low entropy. The materials
taken up by cells are often organic compounds which
not only supply material for the synthesis of cellular
constituents but also may be degraded to provide
energy. A characteristic of all cells is the ability simul-
taneously to synthesize thousands of complex proteins
and other materials and, at the same time, to break
down (catabolize) the same types of compounds. Since
cells both synthesize and catabolize most cellular com-
ponents there is a continuous turnover of the very
structural components of which they are composed.
Metabolism encompasses all of these processes.
The most rapid catabolic reactions are usually
those that provide the cells energy. Organisms vary
greatly in the materials used for food. Human beings,
as well as many other organisms, break down carbo-
hydrates, lipids, and proteins to obtain energy and
starting materials for biosynthesis. In contrast, some
organisms use only one or a few simple organic com-
pounds while autotrophs satisfy their needs entirely
with inorganic materials. Looking at all of the species
of living things we find an extraordinary range of
specialization in metabolism as well as in structure.
Nevertheless, there are many common features of
metabolism. For example, most cells utilize glucose
or a close relative as a source of biosynthetic interme-
diates. Pathways for synthesis of nucleic acids and
proteins are similar in all species. Even the control
of growth and development depends upon proteins
whose structures are often conserved throughout the
living world.
1. Digestion
We humans must digest most foods before we can
utilize them. The same is true for most bacteria, which
need amino acids generated by the breakdown of
An Introduction to Metabolism
10
N
Z
506 Chapter 10. An Introduction to Metabolism
3-Phosphoglycerate
CO
2
ATP, CO
2
H
C
COO

HO CH
3
L-lactate
Pyruvate
C
COO

CH
3
O
NADH
L-Alanine
Pyrimidines
Aspartic acid
Other sugars,
polysaccharides
H

a
t
o
m
s
h
CO
2
Photosynthesis.
Energy from light
Lipids
Coenzyme A derivatives of fatty acids
O
CH
2
CH
2
CH
2
CH
2
C
S CoA R
Acetyl-CoA
C
H
3
C
O
S CoA


o
x
i
d
a
t
i
o
n
Glycogen
Glucose
6-phosphate
O
2
H
2
O
Oxaloacetate
C COO

H
2
C
COO

O
Citric acid cycle
C
COO

OH
H
2
C
COO

H
2
C
COO

Citrate
ATP
and
NADPH
O
2
H
2
O
HO
C
C
CH
2
H
O P
O
H
Glyceraldehyde
3-phosphate
Glucose
ATP H
2
O
many
steps
G
l
y
c
o
l
y
s
i
s
CO
2
CO
2
Electron
transport
chain
ATP
generated
from
ADP + P
i
H atoms
removed from
substrates in
oxidation steps;
attached to
carriers such as
NADH, NADPH,
FADH
2
HO
C
COO

CH
2
H
O P
HO
HO
OH
OH
O
O
P
P represents the phospho group
P O
O
OH

Figure 10-1 An overall view of some metabolic sequences. Several major pathways of catabolism are indicated by heavy lines. The
glycolysis pathway, leading to pyruvate and lactate, starts at the top left, while the oxidation pathway of fatty acids is on the right.
Biosynthetic routes are shown in green lines. A few of the points of synthesis and utilization of ATP are indicated by the lighter green
lines. Some of the oxidationreduction reactions that produce or utilize the reduced hydrogen carriers NADH, NADPH, and FADH
2
are
also indicated. The citric acid cycle, a major supplier of these molecules, is shown at the bottom right, while photosynthesis, the source of
reduced hydrogen carriers in green plants, and the source of nearly all energy for life, is shown at the bottom left.
507 B. Catabolism and the Synthesis of ATP
N
N
N
N
NH
2
OH HO
O
CH
2
O
O
O
O
P P
O O

CH
2
OH OH
N
O
C
H H
NH
2
The hydrogen
is carried here
O
This OH is converted
to OPO
3
2
in NADPH
The reduced coenzyme NADH
proteins. Our digestive enzymes, which act on carbo-
hydrates, proteins, lipids, and nucleic acids, are syn-
thesized by cells in the salivary glands, stomach,
pancreas, and intestinal lining. The properties of these
enzymes are described in Chapter 12. Bacteria secrete
digestive enzymes into their surroundings. Although
most green plants have no need to digest foods, they
too have enzymes closely related to our digestive
enzymes. These enzymes break bonds in proteins or
carbohydrate polymers to allow expansion of leaves
and stems, ripening of fruits, and other physical changes.
All cells carry out processing or maturation of their
newly synthesized polymers. This often involves the
cutting off of one or more pieces of a protein, poly-
saccharide, or nucleic acid. The processing enzymes
are also relatives of digestive enzymes.
2. Sources of Energy
All cells require a source of chemical energy. This
is provided to a large extent by ATP (Chapter 6), whose
hydrolysis can be coupled to reactions of synthesis, to
transport across membranes, and to other endergonic
processes. For this reason, all cells have active path-
ways for the synthesis of ATP. Biosynthesis often also
involves chemical reduction of intermediates; therefore,
cells must have a means of generating suitable reduc-
tants. The reagent of choice is often the hydrogen-
carrying coenzyme NADPH. It is a phosphorylated form
of NADH, a derivative of the vitamin nicotinamide
(Box 15-A), whose structure follows. Reduction of
metabolic intermediates by NADPH provides a second
mechanism by which chemical energy is harnessed for
biosynthesis. A third source of energy, which is con-
sidered in Chapter 18, is the gradient of hydrogen ions
that is set up across cell membranes by oxidative or
photochemical processes.
B. Catabolism and the Synthesis of ATP
The catabolic sequences by which cells obtain
energy often appear dominant. For animals, fungi,
and nonphotosynthetic bacteria these pathways are
used to metabolize large amounts of food and to produce
large amounts of ATP. Because they are also related to
biosynthetic pathways they have a central importance
in virtually all organisms.
1. Priming or Activation of Metabolites
After a polymeric nutrient is digested and the
monomeric products are absorbed into a cell, an
energy-requiring priming reaction is usually required.
For example, the hydrolysis of fats (whether in the
gut or within cells) produces free fatty acids. Before
undergoing further metabolism, these fatty acids are
combined with the thiol compound coenzyme A to
formfatty acyl-CoA derivatives. This requires a
thermodynamically unfavorable reaction that necessi-
tates the expenditure of ATP, that is, its hydrolysis
to AMP and inorganic pyrophosphate (PP
i
). Likewise,
glucose, when taken into cells, is converted into the
phosphate ester glucose 6-phosphate (glucose-6-P),
again with an associated cleavage of ATP. The major
metabolic pathways often start with one of these two sub-
stances: a fatty acyl-CoA derivative or glucose 6-phosphate.
The structures of both are given at the top of Fig. 10-1,
which is designed to provide an overall view of meta-
bolism.
The phospho group of glucose-6-P is not very
reactive but it provides a handle which helps enzymes
to recognize and to hold onto this glucose derivative.
Likewise, the coenzyme A molecule, whose complete
structure is given in Chapter 14, provides a large and
complex handle for enzymes. However, from a chemical
viewpoint, the formation of the thioester linkage of a
fatty acid with the SH group of coenzyme A in the
acyl-CoA is more important. This alters the electronic
structure of the fatty acid molecule, activating it
toward the reactions that it must undergo subsequently.
Thus, priming reactions often provide
chemical changes essential to the mecha-
nisms of subsequent reactions.
Activation of a carboxylic acid by
formation of an acyl-CoA derivative (Eq.
10-1) is of special importance because of
its widespread use by all organisms. The
basic reaction occurs by two steps. The
first (Eq. 10-1, step a) is a displacement
reaction on the phosphorus atom of ATP
to form an acyl adenylate, a mixed anhy-
dride of the carboxylic acid and a substi-
tuted phosphoric acid. Such mixed anhy-
dride intermediates, or acyl phosphates,
are central to much of cellular energy metabolism
because they preserve the high group-transfer potential
of a phospho group of ATP (Chapter 6) while imparting
a high group-transfer potential to the acyl group. This
allows the acyl group to be transferred in step b
508 Chapter 10. An Introduction to Metabolism
O
O
O
O
P Mg
2
O

P
O
O
O
P
O
O
CH
2
O
ATP
N
N
N
N
HO OH
NH
2
O
P
O CH
2
O
Adenine
HO OH
Coenzyme A anion
Acyl adenylate
O R C
O O
CoA S

S R C
O
PP
i
2P
i
H
2
O
AMP
CoA
Acyl-coenzyme A
Fatty acid (as anion)
R C
O
O
ATP
ADP
ADP
d b
c
a

(10-1)
of Eq. 10-1 onto the sulfur atom of coenzyme A to form
an acyl-coenzyme A in which the high group transfer
potential of the acyl group is conserved (see Table 6-6).
Two additional steps are linked to this sequence. In
step c the inorganic pyrophosphate that is displaced in
step a is hydrolyzed to two molecules of HPO
4
2
(P
i
),
and in step d the AMP formed is phosphorylated by
ATP to form ADP. The overall sequence of Eq. 10-1
leads to hydrolysis of two molecules of ATP to ADP +
P
i
. In other words, two molecules of ATP are spent in
activating a carboxylic acid by conversion to an acyl-
coenzyme A.
2. Interconversions of Sugar Phosphates
The strategy employed by most cells in the catabo-
lism of several 6-carbon sugars is to convert them to
glucose 6-phosphate and, in the several steps outlined
in Fig. 10-2, to cleave this hexose phosphate to two
equivalent molecules of glyceraldehyde 3-phosphate.
This triose phosphate can then be metabolized further.
Notice the chemical nature of the reactions involved in
the formation of glyceraldehyde 3-phosphate. The
first step (Fig. 10-2, step a) is the transfer of the phospho
group from ATP to the 6 position of glucose. Step b
is the reversible opening of the sugar ring and step c
the isomerization of an aldose to a ketose. Step d is a
second transfer of a phospho group from ATP, another
priming reaction that provides the second of the phos-
phate handles for the two molecules of triose phosphate
that are formed. Step e is an aldol cleavage which breaks
the CC bond in the center of the ketose chain, while
step f is another sugar isomerization that is chemically
similar to the one in step c.
We see that in the six steps shown in Fig. 10-2 for
conversion of glucose to two molecules of glyceralde-
hyde 3-phosphate, there are only four kinds of reac-
tion: (1) phospho transfer from ATP; (2) opening of a
sugar ring; (3) aldose-ketose isomerization; and (4)
aldol cleavage. These types of reactions, which are
described in more detail in Chapters 12 and 13, occur
in many places in metabolism. As is indicated in Fig.
10-2, glucose 6-phosphate can also be generated from
glycogen or starch. The first step (Fig. 10-2, step g) is
phosphorolysis, cleavage by an inorganic phosphate
ion. This leads directly to glucose 1-phosphate. The
latter is isomerized by a phospho transfer process
(step h).
3. Glycolysis and Fermentation
A major route of breakdown of carbohydrates
is the EmbdenMeyerhof Parnas pathway, often
referred to simply as glycolysis. It is indicated on
the left side of Fig. 10-1 and in more detail in Figs.
10-2 and 10-3. The pathway begins with the reactions
of Fig. 10-2, with either free glucose or glycogen as
starting materials. Its end products may be reduced
materials such as lactic acid or ethanol, which are
formed under anaerobic conditions (Fig. 10-3). How-
ever, under aerobic conditions the product is acetyl-
coenzyme A, whose acetyl group can then be oxidized
to carbon dioxide and water in the citric acid cycle.
After the sugar chain is cleaved in the glycolysis
sequence, the two resultant molecules of glyceraldehyde
3-phosphate are oxidized to 3-phosphoglycerate (Fig.
10-3, steps a,b). The oxidant is the hydrogen carrier
NAD
+
, the oxidized form of NADH. Cells frequently
use NAD
+
to dehydrogenate alcohols to aldehydes or
ketones, with one atom of hydrogen and an electron
from the alcohol becoming attached to the NAD
+
to
give NADH and the other hydrogen being released as
H
+
(Eq. 10-2).
The oxidation of glyceraldehyde 3-phosphate is
considerably more complex. The oxidation of an
aldehyde to a carboxylic acid is a strongly exergonic
process and the oxidation of glyceraldehyde 3-phos-
phate by cells is almost always coupled to the synthesis
509
Glucose
6-phosphate
(open chain)
O
OH
OH
HOCH
2
HO
HO
Glucose
O
OH
O
CH
2
HO
HO
HO
nonreducing end unit
of glycogen
O
OH
OH
CH
2
HO
HO
O
Glucose 6-phosphate
-pyranose ring form
P
ATP
ADP
P
i
H
2
O
C H OH
C H OH
C H OH
C H HO
C
O H
Glyceraldehyde
3-phosphate
Glycolysis Gluconeogenesis
Further
metabolism
(see Fig. 10-3)
Photosynthesis
C
O H
C H OH
CH
2
OH
C O
Dihydroxyacetone
phosphate
Lipids
CH
2
OP
Fructose
1,6-biphosphate
C H HO
C O
C H OH
C H OH
CH
2
OH
Fructose
6-phosphate
C H OH
C H OH
C H HO
C O ATP ADP
P
i
H
2
O
P
i
O
OH
OP
HOCH
2
HO
HO
Glucose
1-phosphate
glycogen
synthase
UDP-glucose
UDP
UTP
PP
i
H
2
O
2P
i
d
e
b
c
f
g
h
CH
2
O P
CH
2
O P
CH
2
O P CH
2
O P CH
2
O P
a
Figure 10-2 The interconversion of glucose, glycogen, and glyceraldehyde-3-phosphate in the pathways of glycolysis,
gluconeogenesis, and glycogen synthesis. Pathways of catabolism are indicated with black lines and those of biosynthesis
with green lines.
B. Catabolism and the Synthesis of ATP
of ATP. The importance of this coupling can be under-
stood by the fact that for many organisms living by anae-
robic fermentation reactions, this one oxidation step provides
the sole source of ATP. It is the mechanism for coupling
this oxidation to synthesis of ATP that accounts for the
complexity of the glyceraldehyde 3-phosphate dehy-
drogenase reaction, whose chemistry is presented in
detail in Chapter 15. As is indicated in the simplified
version of Fig. 10-3 (step a) inorganic phosphate (P
i
) is
a reactant and the product released by the enzyme is
1,3-bisphosphoglycerate. This compound, which has
been formed by the oxidative process, is an anhydride
of phosphoric acid with 3-phosphoglyceric acid. The
phospho group of such acyl phosphates, like that of
ATP, has a high group transfer potential and can be
transferred to ADP to generate ATP (Fig. 10-3, step b).
Since each glucose molecule is cleaved to two mole-
cules of triose phosphate, two molecules of ATP are
510 Chapter 10. An Introduction to Metabolism
(10-2)
Figure 10-3 Coupling of the reactions of glycolysis with formation of lactic
acid and ethanol in fermentations. Steps a to g describe the Embden
Meyerhof Parnas pathway. Generation of 2 ATP in step b can provide all of
the cells energy.
R C
H
OH
R'
N
+
R C O
R'
N
H H
NADH
+ + H
+
+
NAD
+
(partial structure)
C
O
NH
2
C
O
NH
2
d
e
b
c
a
f
g
h
i
j
Acetyl-CoA
O
2 CH
3
SCoA C
2 ADP
2 ATP
Glyceraldehyde
3-phosphate
2P
i
1,3-Bisphosphoglycerate
C
C H
O
OH
CH
2
OP
O
3-Phosphoglycerate
C
C H
O
O
CH
2
OH
O
2-Phosphoglycerate
P 2
CH
2
Phosphoenolpyruvate (PEP)
COO
C OP 2
CH
3
COO
C O 2
Pyruvate
CH
2
COO
C OH 2
2 ADP
2 NADH + 2 H
+
2 NAD
+
L-Lactate
CH
3
COO
C H 2 HO
CH
3
CH
2
OH
2
2
2 CO
2
2 CO
2
Thiamine
diphosphate
Further metabolism
with generation of
about 36 ATP per
glucose. Three are
from reoxidation of
NADH formed in
step a.
2 H
2
O
2
2
Ethanol
C
C H
O
HO
CH
2
OP
OP
C
O
2
C
C OH H
CH
2
OP
H O
ADP
ADP
P
i
Glucose Glycogen
Glucose6P
ATP
ATP
2 ATP
Acetaldehyde
C
H O
CH
3

generated in this process by the fermentation of one


molecule of glucose. This is enough for a bacterium to
live on if it ferments enough sugar.
Further metabolism of 3-phosphoglycerate involves
an isomerization by means of a phosphotransferase
(mutase reaction, step c) to form 2-phosphoglycerate
which then loses water in an elimination reaction (step d).
Elimination of a hydroxyl group in a position relative
to a carboxyl group together with an
-proton is another very frequently
used type of metabolic reaction. In
this case the product, phosphoenol-
pyruvate (PEP), is a unique and
important metabolite. It is a key
intermediate in the biosynthesis of
aromatic amino acids and in numer-
ous other biosynthetic sequences.
In the EmbdenMeyerhof
Parnas pathway PEP transfers its
phospho group to ADP (step e) to
generate ATP and pyruvate. The
latter is shown enclosed in brackets
in Fig. 10-3 as the enolic form, which,
tautomerizes to the oxo form (step f).
The fact that the oxo form of pyru-
vate is much more stable than the
enol gives the phospho group of
phosphoenolpyruvate its high group
transfer potential. The metabolic
significance of this step is that the
phosphate handles of the glycolytic
intermediates, which were initially
transferred on from ATP, are now
returned to ADP with the regenera-
tion of an equivalent amount of ATP.
The product of this metabolic
sequence, pyruvate, is a metabolite
of central importance. Its fate depends
upon the conditions within a cell
and upon the type of cell. When
oxygen is plentiful pyruvate is
usually converted to acetyl-coenzyme
A, but under anaerobic conditions
it may be reduced by NADH + H
+
to the alcohol lactic acid (Fig. 10-3,
step h). This reduction exactly
balances the previous oxidation
step, that is, the oxidation of glycer-
aldehyde 3-phosphate to 3-phospho-
glycerate (steps a and b). With a
balanced sequence of an oxidation
reaction, followed by a reduction
reaction, glucose can be converted
to lactate in the absence of oxygen,
a fermentation process. The lactic
acid fermentation occurs not only in
certain bacteria but also in our own
muscles under conditions of extremely
vigorous exercise. It also occurs
continuously in some tissues, e.g., the
transparent lens and cornea of the eye.
511 B. Catabolism and the Synthesis of ATP
(10-6)
H
3
C C
O
COO
H
3
C C
NAD
O
S CoA
CoASH
CO
2
NADH + H
Acetyl-CoA
Pyruvate
+
+

In the well-known fermentation of sugar to etha-


nol by yeast, the pyruvate generated by glycolysis is
first decarboxylated to acetaldehyde (Fig. 10-3, step i).
This decarboxylation of a 2-oxo acid is chemically
difficult and the enzyme catalyzing it makes use of a
special reagent known as a coenzyme. For this type
of reaction, the coenzyme is the diphosphate (pyro-
phosphate) ester of thiamin (vitamin B
1
). Its mode of
action is discussed in Chapter 14. It is usually needed
when a 2-oxo acid is decarboxylated. The alcoholic
fermentation by yeast is completed by reduction of
acetaldehyde to ethanol (Fig. 10-3, step j), again using
the NADH produced in the oxidation of glyceralde-
hyde 3-phosphate. The conversion of glucose to lactic
acid or to ethanol and CO
2
are just two of many fer-
mentation reactions, most of which are carried out by
bacteria and which are dealt with further in Chapter
17. An important requirement is that the Gibbs energy
change for the overall fermentation reaction be suffi-
ciently negative that ATP synthesis can be coupled to
it. Thus, using data from Table 6-4:
C
6
H
12
O
6
(glucose) 2 C
3
H
5
O
3
(lactate) + 2H
+
G (pH 7) = 196 kJ/mol (46.8 kcal/mol)
2ADP
3
+ 2P
i
2
+H
+
H
2
O + ATP
4
G (pH 7) = +69 kJ/mol
Sum: C
6
H
12
O
6
+ 2ADP
3
+ 2P
i
2

H
2
O + 2 C
3
H
5
O
3
+2H
+
+ 2 ATP
4
G (pH 7) = 127 kJ/mol
A requirement for all fermentations is the existence
of a mechanism for coupling ATP synthesis to the
fermentation reactions. In the lactic acid and ethanol
fermentations this coupling mechanism consists of the
formation of the intermediate 1,3-bisphosphoglycerate
by the glyceraldehyde 3-phosphate dehydrogenase
(Fig. 10-3, step a). This intermediate contains parts
of both the products ATP and lactate or ethanol.
4. Pyruvate Dehydrogenase
In most organisms undergoing aerobic metabolism,
pyruvate is oxidized to acetyl-CoA in a complex process
involving its decarboxylation (Eq. 10-6). This oxidative
decarboxylation, like the decarboxylation of pyruvate
to acetaldehyde, requires thiamin diphosphate. In
addition, an array of other catalysts participate in the
process (see Fig. 15-15). Among these are the electron
carrier flavin adenine diphosphate (FAD), which is
derived from the vitamin riboflavin. Like NAD
+
, this
(10-3)
(10-4)
(10-5)
compound can accept two electrons (plus two protons)
to form FADH
2
. However, it sometimes serves as a
one-electron carrier.
Acetyl-CoA is another major metabolic intermediate.
It is an acyl-CoA of the type mentioned in Section 1
and can also be formed from acetate, ATP, and coen-
zyme A. Although the human diet contains some acetic
acid, the two major sources of acetyl-CoA in our bodies
are the oxidative decarboxylation of pyruvate (Eq. 10-6)
and the breakdown of fatty acid chains. Let us consider
the latter process before examining the further metab-
olism of acetyl-CoA.
5. Beta Oxidation
Whether fatty acids are oxidized to obtain energy
or are utilized for biosynthesis, they are first converted
to their acyl-CoA forms and are then cleaved to the
two-carbon units represented by the acetyl groups of
acetyl-CoA. The beta oxidation sequence, by which
this occurs, is represented by the solid vertical arrow
on the right side of Fig. 10-1 and is shown in greater
detail in Fig. 10-4. We see from the latter figure that
there are two dehydrogenation steps. The first (step b)
removes hydrogen atoms from the - and -carbon
atoms to produce a trans , unsaturated fatty acyl-CoA
(Enoyl-CoA). The hydrogen acceptor is FAD. It is
needed because this reaction requires a more powerful
oxidant than does the dehydrogenation of an alcohol,
for which NAD
+
is adequate. Addition of water to the
double bond of the unsaturated acyl-CoA (step c)
N
N
NH
2
N
N
O
OH HO
CH
2
O
P
O
O O
P O
O
O
H
2
C
C
CH
2
N
N
N
N O
H
O
H
3
C
H
3
C
H)
3
(HO

The coenzyme flavin adenine diphosphate (FAD)


512 Chapter 10. An Introduction to Metabolism
Figure 10-4 Reactions of fatty acid activation and of
breakdown by oxidation.
(10-7)
Enoyl hydratase
FAD
FADH
2
Acyl dehydrogenase
d
e
b
c
a
NAD
H + NADH
3-Hydroxyacyl-CoA
dehydrogenase
Fatty acid
R C
C C
O
S CoA
H
H
R C
C C
O
S CoA
H
H
H
H
RCH
2
CH
2
COO
trans
AMP + PP
i
ATP
Fatty acyl-CoA synthetases
CoASH
H
2
O
O
S CoA
R C
CH
2
C
H
OH
S CoA
H
R C
CH
2
C
O
O
S CoA
R'
CH
2
C
O
SCoA
CH
2
CH
3
C
O
S CoA
Recycle
shortened
Acyl-CoA

Activation
of a fatty acid
B
e
t
a

o
x
i
d
a
t
i
o
n
Cleavage
by thiolysis

+
+
Acyl-CoA
Enoyl-CoA
L-3-Hydroxyacyl-CoA
3-Oxo-acyl-CoA
Acetyl-CoA
generates an alcohol which is then dehydrogenated
(step d) by NAD
+
to form a -oxoacyl-CoA. This is
cleaved (step e) by a reaction (thiolysis) with another
molecule of coenzyme A to form acetyl-CoA and a
new acyl-CoA with a shortened chain. The latter is
recycled by passage through the oxidation sequence
repeatedly until the chain is shortened to a 2- or 3-carbon
fragment, acetyl-CoA, or propionyl-CoA. Since most
dietary fatty acids contain an even number of carbon
atoms, acetyl-CoA is the predominant product.
The beta oxidation of fatty acids, like the dehydro-
genation of pyruvate to acetyl-CoA, takes place within
the inner matrix of the mitochondria in eukaryotic
organisms. The reduced hydrogen carriers FADH
2
and NADH transfer their electrons to other carriers
located within the inner membrane of the mitochon-
dria. In bacteria the corresponding reactions occur
with electron carriers present in the plasma membrane.
Both FAD and NAD
+
are regenerated in this way and
are able to again accept hydrogen from the beta oxida-
tion reactions. This transfer of hydrogen atoms from
substrates to hydrogen carriers is typical of biological
oxidation processes.
6. The Electron Transport Chain, Oxidative
Phosphorylation
Reoxidation of the reduced carriers NADH and
FADH
2
actually involves a sequence of electron carriers,
the electron transport chain, whose function is
indicated below the circle near the center of Fig. 10-1.
The oxidation of reduced NADH by O
2
(Eq. 10-7) is a
highly exergonic process and is accompanied by the
NADH + H
+
+ 1/2 O
2
NAD
+
+ H
2
O
G (pH 7) = 219 kJ/mol
generation of about three molecules of ATP (from
ADP and inorganic phosphate). This process, termed
oxidative phosphorylation, is the principal source of
usable energy (in the form of ATP) provided by break-
down of both carbohydrates and fats in the human
body.
The mechanism of oxidation of NADH in the elec-
tron transport chain appears to occur by transfer of a
hydrogen atom together with two electrons (a hydride
ion H

). Oxidation of FADH
2
to FAD might occur by
transfer of two hydrogen atoms or by transfer of H

+
H
+
. However, it is useful to talk about all of these
compounds as electron carriers with the under-
standing that movement of one or both of the electrons
may be accompanied by transfer of H
+
. The electron
transport complex is pictured in a very simplified
form in Fig. 10-5.
The electrons donated from NADH or other reduc-
tants, upon entering this complex, travel from one
carrier to the next, with each carrier being a somewhat
more powerful oxidant than the previous one. The
513
NADH+H
+
Flavin 1
(FeS)
Flavin 2
Flavin 3
(FeS) (FeS)
QCyt b (FeS)
Succinate
FADH
2
bound to
soluble
enzymes
Cyt c
2
CuCyt aa
3
Cyt c
O
2
O
2
Electron-
transferring
flavoprotein
Mitochondrial
inner membrane
Mitochondrial
matrix
2H
2
O
Cytosol
Figure 10-5 An abbreviated version of the electron transport
chain of mitochondria. Four electrons reduce O
2
to 2 H
2
O. For
details see Figs. 18-5 and 18-6.
BOX 10-A AN EARLY LABELING EXPERIMENT
In 1904, long before the advent of radioactive
tracers, Knoop synthesized fatty acids labeled by
chemical attachment of a benzene ring at the end
opposite the carboxyl group. He prepared these
compounds with both odd and even numbers of
carbon atoms in straight chains and fed them to
dogs. From the dogs urine he isolated hippuric
acid and phenylaceturic acid, which are the amides
of glycine with benzoic acid and phenylacetic acid,
respectively. Knoop showed that the phenylacetic
acid was produced from those fatty acids with an
a
Weinhouse, S. (1995) FASEB J. 9, 820821
b
Quastel, J. H. (1984) Trends Biochem. Sci. 9, 117118
even number of carbon atoms, while the benzoic
acid was formed from those with an odd number.
From these experimental results, Knoop deduced
that fatty acid degradation occurs two carbon atoms at a
time and proposed his famous oxidation theory.
Later experiments using isotopic labeling with
13
C verified Knoops proposals,
a
but study of isolated
enzymes was not possible until after the discovery
of coenzyme A in 1950. Then, studies of fatty acid
oxidation by extracts from isolated mitochondria
established the details of the pathway.
b
n
C
O
OH
CH
2
C
O
OH
CH
2
C
O
N
CH
2
n is
even
Phenylacetic acid
H
COOH CH
2
glycine
Phenylaceturic acid
C
O
OH
Benzoic acid
Hippuric acid
C
O
N
H
COOH CH
2
n is
odd
B. Catabolism and the Synthesis of ATP
final carrier, known as cytochrome aa
3
or
cytochrome oxidase, reacts with molecular
oxygen. Each molecule of O
2
, together with
4 H
+
, is converted to two molecules of water.
This stoichiometry requires that two mole-
cules of NADH pass four electrons down the
chain for each O
2
reduced.
The chemical structures of the components
of the mitochondrial electron transport chain
are varied but fall into several distinct classes.
Most are proteins but these proteins always
contain special coenzymes or prosthetic
groups able to engage in oxidationreduction
reactions. At the substrate end of the chain
NADH passes a hydrogen atom together
with its bonding electron pair to a riboflavin-
containing coenzyme riboflavin 5'-phosphate
(FMN), which is tightly bound into a protein
(designated Flavin 1 in Fig. 10-5). Similar
flavoproteins, Flavin 2, and Flavin 3, act
as oxidants for FADH
2
arising during beta
514 Chapter 10. An Introduction to Metabolism
S
Fe
S from cysteine
Fe
2
S
2
Fe
4
S
4
oxidation and oxidation of succinate, whose signifi-
cance to metabolism is discussed in the next section.
A significant difference between NADH and
FADH
2
is that the former diffuses freely between the
dehydrogenases that transfer hydrogen to it and the
flavoprotein NADH dehydrogenase (Flavin 1) that
reoxidizes it. However, FAD, whether in its oxidized
state or as FADH
2
, stays tightly bound to proteins at
all times. Only hydrogen atoms or electrons are trans-
ferred into or out of these proteins. During beta oxida-
tion the FADH
2
generated remains tied to the fatty acyl-
CoA dehydrogenase protein. However, the two hydrogen
atoms of this FADH
2
are transferred to another molecule
of FAD, which is bound to an electron-transferring
flavoprotein. This protein carries electrons one at a
time, as FADH, by diffusion to the inner surface of the
inner mitochondrial membrane. There it transfers the
hydrogen atoms that it carries to the flavoprotein desig-
nated Flavin 2 in Fig. 10-5. The electron-transferring
flavoprotein can also be viewed as a carrier of single
electrons, each accompanied by H
+
.
A second group of electron carriers in mitochon-
drial membranes are the ironsulfur [FeS] clusters
which are also bound to proteins. Ironsulfur proteins
release Fe
3+
or Fe
2+
plus H
2
S when acidified. The
inorganic clusters bound into the proteins have
characteristic compositions such as Fe
2
S
2
and Fe
4
S
4
.
The sulfur atoms of the clusters can be regarded as
sulfide ions bound to the iron ions. The iron atoms
are also attached to other sulfur atoms from cysteine
side chains from the proteins. The FeS proteins are
often tightly associated with other components of the
electron transport chain. For example, the flavoproteins
Flavin 1, Flavin 2, and Flavin 3 shown in Fig. 10-5 all
contain FeS clusters as does the Q-cytochrome b
complex. All of these FeS clusters seem to be one-
electron carriers.
A third hydrogen carrier of mitochondrial mem-
branes, and the only one that is not unequivocally
associated with a specific protein, is the isoprenoid
quinone ubiquinone or coenzyme Q (Q in Fig. 10-5).
Ubiquinone apparently serves as a common carrier,
collecting electrons from three or more separate input
ends of the chain and directing them along a single
pathway to O
2
.
The final group of mitochondrial redox components
are one-electron carriers, small proteins (cytochromes)
that contain iron in the form of the porphyrin complex
known as heme. These carriers, which are discussed
in Chapter 16, exist as several chemically distinct
types: a, b, and c. Two or more components of each
type are present in mitochondria. The complex cyto-
chrome aa
3
deserves special comment. Although cyto-
chromes are single-electron carriers, the cytochrome aa
3
complex must deliver four electrons to a single O
2
molecule. This may explain why the monomeric complex
contains two hemes and two copper atoms which are
also able to undergo redox reactions.
1,2
Although the components of the electron transport
chain have been studied intensively, there is still some
mystery associated with the process by which ATP
synthesis is coupled to electron transport (oxidative
phosphorylation). A theory originally proposed by
Mitchell
3
and now generally accepted
4,5
is that passage
of electrons through the chain pumps protons from
the inside to the outside of the tight inner mitochon-
drial membrane. As a result, protons accumulate
along the outside of this membrane, as do negative
counterions along the inside. The membrane becomes
charged like a miniature electrical condenser. The
synthesis of ATP takes place in the small knoblike ATP
synthase which is partially embedded in the same
membranes. The Gibbs energy for the formation of
ATP from ADP and inorganic phosphate is apparently
supplied by the flow of protons back to the inside of
the membrane through the ATP synthase. Possible
N
N
N
N
N
N
N
N
Fe
O
O
O
O
Iron in a heme in a b type cytochrome
515 C. Biosynthesis
ways in which this may occur as well as many other
aspects of electron transport are dealt with in Chapter 18.
7. The Citric Acid Cycle
The 2-carbon acetyl units removed from fatty acid
chains or generated from sugars by glycolysis and the
action of pyruvate dehydrogenase must be completely
oxidized to carbon dioxide to provide cells with the
maximum amount of energy. The oxidation of an acetyl
group is a difficult chemical process, and probably for
this reason nature has devised an elegant catalytic
cycle, the citric acid cycle, which is indicated at the
lower right in Fig. 10-1 and is shown in detail in Fig.
10-6. The cycle begins when the 4-carbon oxaloacetate
(also spelled oxalacetate) is condensed with an acetyl
group of acetyl-CoA to form the 6-carbon citrate.
Then, in the remaining reactions of the cycle, two
carbon atoms are removed as CO
2
and oxaloacetate
is regenerated. Several oxidation steps are involved,
each of which feeds additional reducing equivalents
(i.e., hydrogen atoms removed from substrates) into
the pool of hydrogen carriers and allows for more
synthesis of ATP via the electron transport chain. The
importance of this pathway to an organism can be
understood from the fact that when glucose is oxidized
completely to CO
2
and water via the citric acid cycle,
about 38 molecules of ATP are formed, 19 times as
much as by fermentation.
Although oxaloacetate is regenerated and there-
fore not consumed during the operation of the citric
acid cycle, it also enters other metabolic pathways.
To replace losses, oxaloacetate can be synthesized from
pyruvate and CO
2
in a reaction that uses ATP as an
energy source. This is indicated by the heavy gray line
leading downward to the right from pyruvate in Fig.
10-1 and at the top center of Fig. 10-6. This reaction
depends upon yet another coenzyme, a bound form
of the vitamin biotin. Pyruvate is formed from break-
down of carbohydrates such as glucose, and the need
for oxaloacetate in the citric acid cycle makes the oxida-
tion of fats in the human body dependent on the con-
current metabolism of carbohydrates.
C. Biosynthesis
At the same time that cells break down foodstuffs
to obtain energy, they are continuously creating new
materials. The green lines in Fig. 10-1 indicate some
pathways by which such biosynthesis takes place.
Examining the left side of the figure, we see that either
pyruvate or 3-phosphoglycerate can be converted back
to glucose 6-phosphate and that the latter can be used
to synthesize glycogen or other sugars or polysaccha-
rides.
1. Reversing Catabolic Pathways
Breakdown of carbohydrates is thermodynamically
spontaneous. Therefore, cells cannot simply use catabolic
pathways operating in reverse without finding ways of
coupling the cleavage of ATP to synthesis. In the formation
of glucose from pyruvate in the liver, a process known
as gluconeogenesis, there are three distinct points
at which the enzymes used differ from those used in
catabolism: (1) Pyruvate is converted to phosphoenol-
pyruvate by a mechanism utilizing more than one
molecule of ATP, a pathway that is discussed in detail
in Chapter 17; (2) as is shown in Fig. 10-2, fructose
1,6-bisphosphate is hydrolyzed to fructose 6-phosphate
and inorganic phosphate by a phosphatase rather
than through reversal of step d, which would form
ATP; and (3) glucose-6-phosphate is hydrolyzed by a
phosphatase rather than following the reverse of step
a in Fig. 10-2. Furthermore, glycogen is synthesized
from glucose 6-phosphate, not by reversal of the phos-
phorylase reaction (Fig. 10-2, step g), but via a new
intermediate, uridine diphosphate glucose (UDPG),
whose formation involves cleavage of UTP. The latter
is generated by phospho transfer from ATP. Inorganic
pyrophosphate (PP
i
) is a product of UDPG formation
and is removed by hydrolytic cleavage to two molecules
of inorganic phosphate by the enzyme pyrophosphatase.
This reaction helps to make the biosynthesis thermo-
dynamically spontaneous by removing one of the
reaction products.
A similar situation exists in the case of fatty acid
synthesis, which proceeds from acetyl-CoA and reverses
fatty acid breakdown. However, both carbon dioxide
and ATP, a source of energy, are needed in the synthetic
pathway. Furthermore, while oxidation of fatty acids
requires NAD
+
as one of the oxidants, and generates
NADH, the biosynthetic process often requires the
related NADPH. These patterns seen in biosynthesis
of sugars and fatty acids are typical. Synthetic reactions
resemble the catabolic sequences in reverse, but distinct
differences are evident. These can usually be related to
the requirement for energy and often also to control
mechanisms.
N
OH HO
O
CH
2
O
O
O
O
P P
O O O
Uridine diphosphate glucose (UDPG)
H
N
O
O
OH
OH
O
HO
CH
2
OH

516 Chapter 10. An Introduction to Metabolism
d
e
b
c
a
d
f
O
C
C
H

H
Oxaloacetate
OH
C
C
H

H
H
2
C
Citrate
C
C
OH
H

H
H
Malate
CH
2
H
2
C
Succinate
C
C
H
H
Fumarate
C
CoA
CH
2
O
H
2
C
S
Succinyl-CoA
C
CH
2
O
H
2
C
H
C
C
HO
H
H
2
C
threo-D
s
-Isocitrate
(2R-3S-isocitrate)
O
C
S
CH
3
CoA
Acetyl-CoA
From
carbohydrate
metabolism
Oxidative
decarboxylation
Synthesis of regenerating
substrate oxaloacetate
ATP, Biotin
Pyruvate
carboxylase
CO
2
NAD
+
Oxidation
of fatty acids
*
H
C
C
O
H
2
C
Oxalosuccinate
C
C
H
H
2
C
cis-Aconitate
CoASH
Pyruvate
H

OH
*
*
*
*
*
*
*
*
H
2
O
The oxidants
used in the four
dehydrogenation
steps are indicated.
Their reduced forms are
reoxidized by the electron
transport system to
provide energy
(ATP) for
cell use.
CoASH
GTP
GDP
P
i
CoASH
Oxidative
decarboxylation
H
2
O

NAD
+
NAD
+
NAD
+
(NADP
+
)
FAD

O C
CH
3
g
h
i
CO
2
CO
2
CO
2
COO

COO

COO

COO

COO

COO

COO

COO

COO

OOC

COO

COO

OOC

COO

OOC

COO

OOC

COO

COO

COO

COO

COO

COO

COO

Thiamin diphosphate
2-oxoglutarate
Figure 10-6 Reactions of the citric acid cycle (Krebs tricarboxylic acid cycle). Asterisks designate positions of isotopic label
from entrance of carboxyl-labeled acetate into the cycle. Note that it is not the two carbon atoms from acetyl-CoA that are
immediately removed as CO
2
but two atoms from oxaloacetate. Only after several turns of the cycle are the carbon atoms of
the acetyl-CoA completely converted to CO
2
. Nevertheless, the cycle can properly be regarded as a mechanism of oxidation of
acetyl groups to CO
2
. Green daggers () designate the position of
2
H introduced into malate as
2
H
+
from the medium. FAD

designates covalently bound 8-histidyl-FAD (see Chapter 15).


517
2. Photosynthesis
The principal means of formation of glucose in
nature is through photosynthesis in green plants (Fig.
10-1, lower left). Light energy is captured by chloro-
phyll and is used to transfer electrons from chlorophyll
to other electron carriers, the most important of which
is NADP
+
. It is reduced to NADPH which is used to
reduce carbon dioxide to sugar phosphates in a complex
series of reactions known as the reductive pentose
phosphate pathway, which is described in Chapter
17. ATP is also required for photosynthetic reduction
of CO
2
. It is generated by allowing some of the electrons
to flow back through an electron transport chain in the
membranes of the chloroplasts. This chain closely
resembles that from Q to cytochrome c in mitochondria
(Fig. 10-5), and the generation of ATP in this photo-
synthetic phosphorylation occurs in a manner anal-
ogous to that in the electron transport chain of mito-
chondria. In green plants the electrons removed from
chlorophyll in one light-requiring reaction are replaced
by electrons formed during the cleavage of water in a
second light-dependent reaction, a reaction that also
releases oxygen, O
2
, and generates hydrogen ions
(H
+
). The first stable product from reduction of CO
2
in photosynthesis is 3-phosphoglycerate. It can be
converted to sugars by pathways analogous to those
employed by animals in gluconeogenesis. One inter-
esting difference is the use in chloroplasts of NADPH
+ H
+
in reduction of 3-phosphoglycerate (reverse of
step a in Fig. 10-3).
A small number of other biosynthetic pathways,
which are used by both photosynthetic and nonphoto-
synthetic organisms, are indicated in Fig. 10-1. For
example, pyruvate is converted readily to the amino
acid L-alanine and oxaloacetate to L-aspartic acid;
the latter, in turn, may be utilized in the biosynthesis
of pyrimidines. Other amino acids, purines, and addi-
tional compounds needed for construction of cells are
formed in pathways, most of which branch from some
compound shown in Fig. 10-1 or from a point on one
of the pathways shown in the figure. In virtually every
instance biosynthesis is dependent upon a supply of
energy furnished by the cleavage to ATP. In many
cases it also requires one of the hydrogen carriers in a
reduced form. While Fig. 10-1 outlines in briefest form
a minute fraction of the metabolic pathways known,
the ones shown are of central importance.
D. Synthesis and Turnover of Macromolecules
Proteins make up the bulk of the catalytic machinery
of cells and together with other macromolecules most
of the structure. Therefore, the synthesis and degrada-
tion of proteins and the control of those processes are
of great importance to cells. Although the emphasis
BOX 10-B DISCOVERY OF THE CITRIC ACID CYCLE (KREBS TRICARBOXYLIC ACID CYCLE)
One of the first persons to study the oxidation
of organic compounds by animal tissues was T.
Thunberg, who between 1911 and 1920 discovered
about 40 organic compounds that could be oxidized
by animal tissues. Salts of succinate, fumarate,
malate, and citrate were oxidized the fastest. Well
aware of Knoops oxidation theory, Thunberg
proposed a cyclic mechanism for oxidation of acetate.
Two molecules of this two-carbon compound were
supposed to condense (with reduction) to succinate,
which was then oxidized as in the citric acid cycle
to oxaloacetate. The latter was decarboxylated to
pyruvate, which was oxidatively decarboxylated
to acetate to complete the cycle. One of the reactions
essential for this cycle could not be verified experimentally.
It is left to the reader to recognize which one.
In 1935, A. Szent-Gyrgyi discovered that all
of the carboxylic acids that we now recognize as
members of the citric acid cycle stimulated respira-
tion of animal tissues that were oxidizing other
substrates such as glucose. Drawing on this know-
ledge, Krebs and Johnson in 1937 proposed the citric
acid cycle.
a,b,c
Krebs provided further confirmation
in 1940 by the observation that malonate, a close
structural analog and competitive inhibitor of succi-
nate, in concentrations as low as 0.01 M blocked the
respiration of tissues by stopping the oxidation of
succinate to fumarate.
c

OOCCH
2
COO

Malonate

OOCCH
2
CH
2
COO

Succinate
In muscle, 90% of all respiration was inhibited
and succinate was shown to accumulate, powerful
proof of the importance of the citric acid cycle in the
respiration of animal tissues.
a
Krebs, H. A., and Johnson, W. A. (1937) Enzymologia 4, 148156
b
Krebs, H. A. (1981) Reminiscences and Reflections Clarendon
Press, Oxford
c
Fruton, J. S. (1999) Proteins, Enzymes, Genes: the Interplay of
Chemistry and Biology Yale Univ. Press, New Haven, CT
d
Quastel, J. H. (1978) Trends Biochem. Sci. 3, 6869
D. Synthesis and Turnover of Macromolecules
518 Chapter 10. An Introduction to Metabolism
in this section is on proteins, similar considerations
apply to nucleic acids, polysaccharides, and other
macromolecules.
With the exception of some antibiotics and other
short-chain molecules, all polypeptides are formed on
ribosomes, which assemble proteins according to the
sequences of nucleotides in the messenger RNA
(mRNA) molecules. The basic chemistry is simple.
The carboxyl groups of the amino acids are converted
to reactive acyl adenylates by reaction with ATP, just
as in Eq. 10-1. Each activated amino acid is carried
on a molecule of transfer RNA (tRNA) and is placed
in the reactive site of a ribosome when the appropriate
codon of the mRNA has moved into the site. The
growing peptide chain is then transferred by a dis-
placement reaction onto the amino group of the acti-
vated amino acid that is being added to the peptide
chain. In this manner, new amino acids are added one
at a time to the carboxyl end of the chain, which always
remains attached to a tRNA molecule. The process
continues until a stop signal in the mRNA ends the
process and the completed protein chain is released
from the ribosome. Details are given in Chapter 29.
1. Folding and Maturation of Proteins
A newly synthesized peptide chain probably folds
quickly. However, the cytosol provides an environ-
ment rich in other proteins and other macromolecules
that can interact with the new peptide and may cata-
lyze or inhibit folding. Among the most abundant
proteins of bacteria or eukaryotes are proteins known
as chaperonins. They apparently help polypeptide
chains to fold correctly, partly by chaperoning them
through the cytoplasm and across cell membranes,
protecting them from becoming entangled with other
proteins and macromolecules while they fold.
69
There
are several classes of chaperonins. Most are oligomers
made up of 10- to 90-kDa subunits. They have a variety
of names, which may be somewhat confusing. The
first chaperonins were identified only as heat shock
proteins (Hsp). They are produced in large amounts
by bacteria or other cells when the temperature is
raised quickly, and are designated Hsp70, Hsp90, etc.,
where the number is the subunit mass in kDa. Other
chaperonins were recognized as products of genes
needed for replication of the DNA of bacteriophage
in E. coli. Consequently, one major chaperonin of the
Hsp70 class is designated DnaK. The Hsp70 protein
of mitochondria was named binding protein or BiP.
10
Other workers have abbreviated chaperonins as Cpn70,
Cpn60, etc.
The abbreviations Hsp70, Cpn70, DnaK, and BiP
all refer to a group of similar 70-kDa proteins that are
apparently found in all organisms. Their role seems to
be to stabilize unfolded proteins prior to final folding
in the cytosol or after translocation into the endoplas-
mic reticulum (ER) or into mitochondria or other
organelles. Each of these proteins consists of two
functional domains. A 52-kDa domain at the C terminus
binds 7- or 8-residue segments of unfolded peptide
chains in an elongated conformation.
9
At some point
in its reaction cycle the N-terminal 40-kDa domain,
which binds ATP tightly, causes the ATP to be hydro-
lyzed to ADP and inorganic phosphate: ATP + H
2
O
ADP + P
i
. Binding and release of a polypeptide by
the E. coli DnaK protein is coupled tightly to this exer-
gonic ATPase reaction.
10a,10b
The reaction is depen-
dent upon potassium ions
11
and is regulated by two
co-chaperones, DnaJ (Hsp40)
11a-11c
and GrpE.
10a
Both ATP and extended polypeptides bind weakly to
DnaK, and the ATP in the DnaKpolypeptideATP
complex is hydrolyzed slowly to ADP and inorganic
phosphate.
10a,10b,12
Co-chaperone DnaJ, which shares
a largely -helical structural motif with J-domains in
various other proteins, binds to the complex.
11a, 11b, 11c
It probably induces a conformational change that leads
to rapid hydrolysis of ATP. In the resulting complex
both ADP and the polypeptide are bound tightly to
the DnaK protein and dissociate from it very slowly.
The E. coli co-chaperone GrpE acts as a nucleotide
exchange factor that catalyzes rapid loss of ADP from
the complex. If ATP binds to this DnaKpolypeptide
complex the polypeptide is released.
10a
This cycle,
which can be repeated, accomplishes the function of
DnaK in protecting extended polypeptides and releas-
ing them under appropriate conditions. There is also
evidence that DnaK participates in refolding of mis-
folded proteins.
11d
It cooperates with a ribosome-
associated prolylisomerase in bacteria.
11e
The three-
dimensional structure of the ATPase domain of Hsp70
is strikingly similar to that of the enzyme hexokinase
(Chapter 12) and to that of the muscle protein actin,
Fig. 7-10.
13,14
Archaeal chaperonins lack the Cpn10
ring but have lid-like extensions at the cylinder ends.
15a
The Cpn60 class of chaperonins are amazing cage-
like structures. Each oligomer is composed of two
rings, each made up of seven 60-kDa subunits stacked
back-to-back. Cpn60 structures from archaeobacteria
are similar but may have 8- or 9-subunit rings.
15,15a
The best known member of this group is the E. coli
protein known as GroEL whose three-dimensional
structure is depicted in Box 7-A.
16,17
This 14-subunit
oligomer of GroEL is a cylinder which is capped by a
smaller ring composed of seven 10-kDa subunits of
GroES, a Cpn10 protein.
17,18
Like Hsp70, GroEL has
ATPase activity and an ATP binding site in its large
equatorial domain.
16
Archaeal chaperonins lack the
Cpn10 ring but have lid-like extensions at the cylinder
ends.
15a
Within the GroELGroES cage polypeptide chains
can fold without becoming entangled with other pro-
teins or being cleaved by protein-hydrolyzing enzymes
519 D. Synthesis and Turnover of Macromolecules
insulin
(51 residues, two
disulfide-linked
chains)
proinsulin
(86 residues,
one chain)
preproinsulin
(110 residues)
(10-8)
50
C peptide, 29 residues
30
E R R
A
R Q K G
1 65 6
C C
C
C
A chain,
21 residues
B chain, 30 residues
1
24
F
7
C
S
S
19
C
S
S
N
COO
S
S
Pre-peptide,
24 residues
NH
3
20
+

Figure 10-7 Schematic diagram of the structure of human preproinsulin.


The 24-residue prepeptide, the 29-residue C-peptide and basic residues 31, 32,
64, and 65 are cut from the peptide upon conversion to insulin as indicated by
the small arrows. Some amino acid residues are identified using the one-
letter codes. See Fig. 7-17 for details of insulin structure.
of the cytoplasm. An unfolded or
partially folded protein may diffuse
into the open end of the complex
and bind temporarily via noncova-
lent interactions. The hydrophobic
inner surface of the complex may
favor the formation of helices within
the folding protein. Binding of the
polypeptide substrate and capping
with GroES causes a major coopera-
tive conformational change with
doubling of the internal volume
(Box 7-A). The character of the
inner wall also changes as a result of
exposure of hydrophilic groups.
18a,18b
Perhaps the expansion also stretches
segments of the unfolded poly-
peptide. Subsequent hydrolysis
of ATP may be coupled to release
of the bound polypeptide, which
may then leave the complex or bind
again for another chance to fold
correctly.
17,19
The two ends of the
GroELGroES complex may func-
tion alternately, with each end in
turn receiving the GroES cap.
17
Hsp90 is one of the most abundant cytosolic proteins
in eukaryotic cells, but it seems to chaperone only a few
proteins, among which are steroid-hormone receptors.
20,20a
Another group of over 30 different chaperone proteins
participate in assembly of external bacterial cell-surface
structures such as pili (Fig. 7-9). The chaperone PapD
consists of two domains, both having an immunoglobulin-
like fold.
20b
PapD binds to the pilin subunits, escorting
them to the site of pilus assembly. Folding of integral
membrane proteins may be facilitated by a chaperone
function of membrane lipids.
20c
For most proteins the initial synthesis is followed
by a sequence of processing or maturation reactions.
These reactions sometimes involve alteration of amino
acid side chains and very often include hydrolytic
cleavages by which pieces of the peptide chain are cut
off. The initially synthesized polypeptides are often
not functional and gain biological activity only after
one or more pieces have been removed. For example,
digestive enzymes are usually secreted as zymogens
or proenzymes which are activated by hydrolytic
cleavages only after secretion into the digestive tract.
Many proenzymes are components of the extremely
complex cascades of activating reactions involved in
blood clotting (Chapter 12) and in the defensive
complement system (Chapter 31).
Most peptide hormones are cut out from larger
proteins. For example, human insulin is synthesized
as a 110-residue preproinsulin which is converted in
stages to the active two-chain, 51-residue hormone
(Eq. 10-8 and Fig. 10-7).
20d,20e
The 24-residue prepeptide is cleaved from preproinsulin
within a few minutes of synthesis. Then, over a period
of about an hour additional cleavages occur to give the
final product.
21
This crystallizes as the zinc hexamer
(Fig. 7-18) within the dense cores of storage vesicles
from which it is released into the bloodstream as needed.
22
Other examples are described in Section 3.
2. Transport of Proteins to Their Destinations
within a Cell
The synthesis of peproinsulin on ribosomes in the
cytosol and the release of mature insulin from secretion
vesicles involve not only the chain cleavages but also
transport of the polypeptide across membranes of the
ER, Golgi compartments, and secretion vesicles.
Signal sequences and translocation. In 1971,
Blobel and Sabatini postulated that for some proteins
an N-terminal segment of newly synthesized poly-
peptide chains contains a signal sequence or leader
peptide of 1530 amino acids which carries informa-
tion concerning the location of the mature protein in
the cell.
2328b
The rather nonpolar signal sequences of
proteins destined for secretion from cells would interact
520 Chapter 10. An Introduction to Metabolism
6-8 residues 8-12 Hydrophobic core P AXA
(or G, S)
++
Cut
with and pass through the membrane of the ER. On
the other side of the membrane, a signal peptidase
would cut off the signal peptide. In many cases, glyco-
syltransferases would add sugar residues near the N
terminus to create a hydrophilic end which would
help to pull the rest of the peptide chain through the
membrane. A similar situation would hold for secre-
tion of proteins by bacteria.
The signal hypothesis has been proven correct but
with considerable added complexity. It now appears
that when the N terminus of a new polypeptide chain
carrying the proper signal sequence emerges from a
ribosome it is intercepted by a signal recognition
particle (SRP), a 250-kDa ribonucleoprotein consist-
ing of six polypeptide chains and a small 300-residue 7S
RNA chain.
2933
Binding to the ribosome, the SRP tem-
porarily blocks further elongation of the peptide chain
until it bumps against and binds to a 72-kDa mem-
brane protein called the SRP receptor or docking
protein. Then translation begins again and the pro-
tein moves into the ER, where it is cleaved by the
signal peptidase and is modified further. Although
the bacterial SRP is a single protein called Ffh (for 54
homolog),
33a
the basic machinery for signal recognition
and secretion is remarkably conserved from E. coli to
humans.
33b
Bacterial proteins that are destined for
secretion or for a function in the periplasmic space or
in external membranes also contain signal sequences
which are cut off by a signal peptidase embedded in
the plasma membrane.
33c
Signal sequences vary in structure but usually
have a net positive charge within the first 58 residues
at the N terminus. This region is followed by a hydro-
phobic core made up of 810 residues with a strong
tendency toward helix formation. This sequence
is often followed by one or a few proline, glycine, or
serine residues and then a sequence AXA that immedi-
ately precedes the cleavage site. Here, A is usually
alanine in prokaryotes but may also be glycine, serine,
or threonine in eukaryotes. Residue X is any amino
acid.
25,27,28a,28b
It had often been assumed that a hydrophobic
signal sequence, perhaps folded into a hairpin loop,
spontaneously inserts itself into an ER membrane to
initiate translocation. However, study of the genetics
of protein transport suggests otherwise. Over 50
different genetic loci affect the translocation of proteins
in yeast.
31,32,33d
Products of these secretory genes,
which are named SEC 61, SEC 62, SEC 63, etc., are
proteins with corresponding names, e.g., Sec 61 protein
(or Sec 61p). Study of these proteins suggested that the
Sec 61, Sec 62, and Sec 63 proteins are all directly
involved in transport into the ER.
31,34,35
Additional
proteins are needed for movement of vesicles out from
the Golgi and for delivery of secretory vesicles to the
plasma membrane.
33d
The protein Sec 61p has been
identified as homologous to the subunit of a similar
trimeric Sec 61p complex of mammalian tissues.
32
It also appears to correspond to Sec Y, a protein required
for secretion of proteins through the plasma mem-
brane of E. coli into the periplasmic space. Proteins
corresponding to the and subunits of mammalian
related proteins have been identified in yeast and E.
coli
36
and in Arabidopsis, where it is necessary to bring
proteins into the thylakoid membrane of the chloro-
plasts.
37
In E. coli the products of genes SecY, SecE, and
SecG are integral membrane proteins that, together
with additional proteins, form a proteinaceous pore
through which proteins pass.
3840a
Additional proteins
required for translocation of some bacterial proteins
are the chaperonin SecB, a tetramer of 17.3-kDa sub-
units,
41
and SecA, a soluble ATPase that may partici-
pate in docking and serve as an engine for transport
throught the pore.
38,40a,42
In eukaryotic cells a membrane-
bound Ca
2+
-dependent chaperonin called calnexin
assists in bringing the protein into the ER in a properly
folded state.
43,44
Unlike transport across the membranes of the ER,
transport across plasma membranes of bacteria often
requires both hydrolysis of ATP and energy provided
by the membrane electrical potential.
33,38,4448
Secre-
tion into the periplasmic space has been well charac-
terized but less is known about transport of proteins
into the external membranes of E. coli.
48
A 16 kDa
periplasmic chaperone may be required.
48a
Many bacteria have a second complete secretion
system.
48b
This multi-gene type II system is present
but usually inactive in E. coli. A third (type III) system
is present in many pathogenic bacteria, and has
evolved for delivery of specialized structural and
regulatory proteins into host cells.
48c,48d
Ticketing destinations. We have seen that
proteins that contain suitable signal sequences are
exported from the cytoplasm while other proteins
remain. Some proteins are secreted while others take
up residence as integral or peripheral membrane
proteins or as soluble proteins within an organelle.
All of the available evidence indicates that it is the
sequence of a protein that determines its destination.
Proteins targeted to pass through the inner mitochon-
drial or chloroplast membrane have 20- to 70-residue
presequences that are rich in arginine and lysine and
which are removed when the protein reaches its
521 D. Synthesis and Turnover of Macromolecules
destination.
4953
Proteins meant to go to the intermem-
brane space or the outer membrane of mitochondria
have a sequence containing basic amino acids followed
by a long stretch of uncharged residues.
54
Peroxisomal
preproteins may have signal sequences such as SKL at
the C terminus
5456
and proteins destined to become
attached to phosphatidylinositol glycan anchors (Fig.
8-13)
57
as well as some bacterial surface proteins
57a
have signal peptides at both N and C termini. Some
lysosomal membrane glycoproteins have an LE pair
in the N-terminal cytoplasmic tail
58
and proteins with
suitable dileucine and related pairs are often taken up
in lysosomes.
59
Soluble proteins that are resident in the
cisternae of the ER often have the C-terminal sequence
KDEL or HDEL.
29,6064
It may serve both as a retention
signal and as a retrieval signal for return of the protein
if it passes on into the Golgi vesicles.
65,66
Transport of proteins into mitochondria is depen-
dent upon both cytosolic chaperonins and mitochon-
drial chaperonins of the Hsp70 and Hsp60 classes.
6770
Entrance into mitochondria
7173
resembles passage
through membranes of the ER. However, entry to the
nucleus through the nuclear pore complex requires
other localization signals
73a
as well as specialized
proteins.
74,74a
The sorting of proteins into six different
compartments within a chloroplast requires a whole
series of recognition signals as well as chaperonins
and channel proteins.
7579
The various signal sequences
that determine a proteins interactions with chaperonins,
docking proteins, and proteinaceous pore complexes
may be complex and overlapping but evolution has
selected sequences that allow cells to live and function.
Computer programs for detecting sorting signals are
being developed.
79a
Not only proteins but also other
macromolecules are automatically sorted to their
correct destination.
Vesicular transport and the Golgi system.
Movement of secreted proteins through the cytosol
from the ER to the external surface occurs through the
formation and opening up of small vesicles about 70
nm in diameter. It occurs in steps that involve passage
from the ER to the various Golgi membranes. Figure
10-8 provides a sketch of the system of ER, Golgi,
secretion vesicles, and lysosomes.
63
Newly formed
secretory proteins flow from the ER into an intermediate
compartment where vesicles are formed and are carried
to the cis Golgi network (CGN). They move step-by-
step through the Golgi stack (GS) and into the trans
Golgi network (TGN). New vesicles are formed to
carry proteins between each pair of membranous
compartments. In each compartment new glycosylation
reactions or other modifications may occur. Finally,
vesicles carry the mature proteins to the plasma mem-
brane, lysosomes, or vacuoles.
79b
A related process is
uptake of proteins by endocytosis to form, consecutively,
early and late endosomes which fuse with lysosomes
(Chapter 8, Section C,6, Fig. 10-8).
At every step in these processes vesicles are formed
and are carried to the next destination where the
vesicles fuse with the new membrane and discharge
their contents.
8085a
This remarkable process is complex
and highly specific. Rothman proposed that the vesicles
to be transported are docked on appropriate receptor
molecules (called SNARES) on the destination mem-
brane. This is accomplished with the aid of specific
soluble marker proteins (called SNAPS) with surfaces
complementary to those of the receptors.
80
Proteins
are sorted in this way according to their destination
signals. The Golgi system is considered further in
Chapter 20.
Figure 10-8 Current version of protein synthesis and
processing via ER, Golgi, and secretory vesicles. CGN,
cis-Golgi network: C, T, M are the cis, medial, and trans
compartments of the Golgi stack; TGN, trans Golgi network.
Arrows indicate some of the movements of transport
vesicles.
Lysosome Endosomes
Secretion
vesicles
Nucleus
M C T TGN CGN
Plasma
membrane
3. Posttranslational Alterations
Chemical alterations to a protein begins as soon as
the peptide chain is formed. Proteins translocated into
the periplasmic space of bacteria or into the cisternae
of the ER of eukaryotic cells meet several important
enzymes. Peptidylprolyl isomerase (discussed in Box
9-F) assists the folding of the peptide chain and protein
disulfide isomerases help to form disulfide linkages.
The cytosol provides a reducing environment in which
many proteins that have cysteine side chains carry free
SH groups. In contrast, the periplasmic space and the
ER have more oxidizing environments in which disul-
fide bridges may form. As shown in Eq. 10-9, the
oxidizing agent is often assumed to be the disulfide
form of the tripeptide glutathione (see Box 11-B),
522 Chapter 10. An Introduction to Metabolism
which is commonly abbreviated GSSG. It can react
with proteins by disulfide exchange reactions to form
mixed disulfide groups with release of reduced gluta-
thione, GSH (Eq. 10-9, step a). The mixed disulfides
can be converted to disulfide bridges by a second
exchange reaction (Eq. 10-9, step b). However, in the
bacterial periplasm the dithiol protein DsbA appears
to be the major oxidant (see Box 15-C). It functions
together with other proteins, some of which carry
electrons from the electron transport chains of the
bacteria.
85a,85b
The rates of these reactions are greatly
increased by the protein disulfide isomerases.
85a85e,8691
These enzymes tend to promote formation of disul-
fide linkages between the correct pairs of SH groups.
If there are three or more SH groups in a chain some
incorrect pairing may, and often does, occur. The
protein disulfide isomerases break these bonds and
allow new ones to form.
92
The active sites of these
isomerases contain pairs of SH groups which can
be oxidized to internal SS bridges by NAD
+
-
dependent enzymes. These enzymes and their relatives
thioredoxin and glutaredoxin are discussed further
in Box 15-C. Glutathione and oxidationreduction
buffering are considered in Box 11-B.
Proteolytic processing. For proteins exported
from the cytosol, a signal peptidase (or leader peptidase)
is usually waiting in the bacterial plasma membrane or
in membranes of the ER. The E. coli leader peptidase
is an integral membrane protein with its catalytic
domain in the periplasm.
45,93
For insulin the 24-residue
prepiece serves as the signal sequence which is hydro-
lyzed off in the ER. However, the other cleavages
shown in Fig. 10-7 appear to take place in immature
secretory granules.
21,22
For many proteins proteolytic
processing begins earlier than this and may occur in
more than one location. Eukaryotic processing proteases
often resemble the bacterial protease subtilisin
20e
but
cut peptide chains preferentially after dibasic amino
acid pairs, e.g., Arg-Arg, Arg-Lys.
9496
There are also
other processing enzymes with differing specificities.
97
Although most polypeptides leave ribosomes as
single proteins, some are polyproteins, which give
rise to two or more functional peptides. Polyproteins
occur in all cells but are especially prevalent among
virally encoded peptides. For example, the polio
virus polyprotein is cut into at least ten pieces by
proteases, some present in the host cell normally and
some encoded by the virus.
98
Many neurohormones
arise from polyproteins that undergo processing as
they travel down an axon from the cell body before
being secreted into a synapse (Chapter 30). Within our
own brains the peptide prepro-opiomelanocortin
undergoes numerous cleavages to give rise to at least
seven different neurohormones (see Fig. 30-2).
96,99101
Altered ends. Proteins often contain blocked
end groups in place of free NH
3
+
or COO

. For
many of the cytosolic proteins, which do not carry
leader sequences, the N-terminal methionine that is
always used to initiate the ribosomal synthesis of
proteins in eukaryotes is removed. If the next residue
after the N-terminal methionine is small and uncharged,
the methionine is usually hydrolyzed off enzymatically
after 3040 residues have been added to the growing
peptide chain.
102105
An acetyl group is transferred
onto the NH
2
termini of about 85% of all cytosolic
proteins, whether or not the initiator methionine has
been removed.
106
In other cases fatty acyl groups may
be transferred to the terminal NH
2
group or to side
chain SH groups. For example, a myristoyl (tetradeca-
noyl) group is frequently combined in amide linkage
at the N terminus of cellular and virally encoded
proteins. A palmitoyl group is joined in thioester
linkage to a cysteine side chain near the N terminus
of the E. coli periplasmic lipoprotein (Fig. 8-28).
107
Together with the N-terminal myristoyl group it forms
a membrane anchor for this protein. Polyprenyl groups
are transferred onto cysteine SH groups at or near the
C termini of many eukaryotic proteins to form a differ-
ent membrane anchor.
108110
Some of the enzymes act
on the sequence CAAX, where X = M, S, Q, C but not
L or I. After transfer of a farnesyl or geranylgeranyl
group onto the cysteine the AAX is removed proteo-
lytically and the new terminal cysteinyl carboxyl
group is methylated. Surface proteins of gram-positive
bacteria are joined by amide linkage from their C
termini to a pentaglycine chain of the peptidoglycan
layer (Fig. 8-28)
57a
A pyroglutamyl N terminus is found in the thyro-
tropin-releasing hormone (Fig. 2-4) and in many other
peptide hormones and proteins. It presumably arises
by attack of the -NH
2
group of an N-terminal gluta-
mine on the side chain amide group with release of
NH
3
(Eq. 10-10).
111,112
Another frequent modification at the C terminus
of peptide hormones and of other proteins is amidation.
In this reaction a C-terminal glycine is oxidatively
removed as glyoxylate in an O
2

, copper- and ascorbate


Peptide
chain
CH
2
SH
CH
2
S GS
GS-SG GSH
GSH
CH
2
SH
CH
2
SH
CH
2
CH
2
S S
H
+
b
a
(10-9)
523 D. Synthesis and Turnover of Macromolecules
(vitamin C)-dependent process (Eq. 10-11).
113117
See
also Chapter 18, Section F,2.
An example of both of these modifications is forma-
tion of thyrotropin-releasing hormone shown in Fig. 2-4
from its immediate precursor:
GlnHisProGly pyroglutamylHisProNH
2
Proteins with long C-terminal hydrophobic signal
sequences may become attached to phosphatidylinositol-
glycan anchors embedded in the plasma membrane
(Fig. 8-13). An example is a human alkaline phospha-
tase in which the carboxyl of the terminal aspartate
residue forms an amide linkage with the ethanolamine
part of the anchor. Attachment may occur by a direct
attack of the NH
2
group of the ethanolanine on a pep-
tide linkage in a transacylation reaction that releases a
29-residue peptide from the C terminus.
118,119
(See
Chapter 29).
Many other covalent modifications of proteins are
dealt with in other sections of the book.
120122
A few
are described in Chapter 2, Eqs. 2-14 to 2-22. Reversible
alterations used to regulate enzymes are considered in
Chapter 11. Of these, the phosphorylation of OH
groups of serine, threonine, and tyrosine is the most
important. A large fraction of all cellular proteins appear
to be modified in this way. Protein glycosylation, the
transfer of glycosyl groups onto OH side chain groups
of serine and threonine (Chapter 4, and Chapter 20)
and nonenzymatic glycation (Eq. 4-8) also affect many
proteins, often at turns in the peptide chain. Hydroxy-
lation, glycosylation, and other modifications of col-
lagen are described in Chapter 8. Another common
reversible alteration is formation of sulfate esters of
tyrosine OH groups.
99,123
Reactions by which cofactors
become attached to proteins
124
are described in Chapters
1517.
Methylation,
125
hydroxylation, and other irreversible
modifications often affect specific residues in a protein.
Oxidative alterations occur during aging of proteins
(Chapter 18).
126
A few proteins even undergo splicing
that alters the amino acid sequence (Box 29-E).
127
All
of these reactions not only affect the properties of the
proteins but also participate in driving the turnover of
these macromolecules.
4. Intracellular Degradation of Proteins
Once a protein has reached its correct location and
has acquired its proper function, it usually has a limited
lifetime, which may average only a few hours or a few
days. The protein is then hydrolytically degraded
back to its constituent amino acids.
128
Defective and
damaged proteins are usually degraded much more
rapidly than are intact proteins.
129,130
Under condi-
tions of starvation, proteins are broken down more
rapidly than usual to supply the cell with energy.
Rapid degradation of proteins is often induced at
certain stages of differentiation. For example, spore-
forming bacteria contain a protease that becomes
activated upon germination of the spore.
131
Within
minutes this enzyme digests stored proteins to provide
amino acids for the synthesis of new proteins during
growth.
Eukaryotic cells degrade proteins within both the
cytosol and lysosomes. Lysosomes apparently take up
many proteins but have a preference for N-terminal
KFERQ
132
and also for particular types of glycosylation
(Chapter 20). Lysosomes act on many long-lived
proteins.
133,133a
Once within the lysosomes, the proteins
are broken down into amino acids with a half-life of
~8 minutes. During nutritional deprivation, the rate
of uptake of proteins by lysosomes increases markedly.
The same is true during certain developmental changes,
for example, when a tadpole loses its tail.
Many short-lived proteins are degraded within the
cytosol in ATP-dependent processes. A major process
involves the small protein ubiquitin (Box 10-C).
134
Once labeled by formation of an isopeptide linkage
to ubiquitin, a peptide is attacked by proteases in the
proteasome complexes (Box 7-A, Chapter 12). There
it is quickly degraded. Other proteases, most of which
do not require ATP, are also present in the cytoplasm
(Chapter 12). How do these enzymes as well as those
within the lysosomes work together to produce
a harmonious turnover of the very substance of our
tissues? How is it possible that one protein has a long
half life of many days while another lasts only an hour
or two in the same cell? The answer seems to be that
(10-12)
H
2
C CH
2
C C
H
2
N
O
+
H
C N Peptide
O
H
H
2
N
H
2
C CH
2
C C
C N Peptide
O
H
H
H
O
N
NH
3
H
(10-10)
Peptide C
O
N CH
2
COO

Peptide C
O
NH
2
H
Gly residue
O
2
H C
O
COO

H
2
O
H
2
A
(Ascorbate)
A
Glyoxylate
(10-11)
524 Chapter 10. An Introduction to Metabolism
BOX 10-C UBIQUITIN
The small 76-residue protein called ubiquitin
a-c
is probably present in all eukaryotic cells. Found
in the nucleus, cytoplasm, cell surface membranes,
and extracellular fluids, ubiquitin is often joined by
isopeptide linkages from its C-terminal glycine to
the -amino groups of lysine side chains of other
proteins. Ubiquitin has one of the most conserved
of all known amino acid sequences. No amino acid
substitutions have been found among animal species
and only three differences distinguish plant ubiqui-
tins from that of humans. In its three-dimensional
structure ubiquitin is compact, tightly hydrogen
bonded, and roughly spherical. It contains an
helix, a mixed sheet, and a distinct hydrophobic
core.
Linkage of ubiquitin to other proteins occurs
through the action of a ligase system which catalyzes
four sequential reactions as shown in the accom-
panying equation. In step a a ubiquitin activating
enzyme (E
1
) forms a C-terminal acyl adenylate by
reaction of ubiquitin (Ub) and ATP.
d,e
In step b a
sulfhydryl group of the same enzyme then displaces
the AMP part to form a thioester linkage to ubiquitin.
The chemistry of the reaction is the same as that in
Eq. 10-1. The activated ubiquitin is next transferred
(step c) by transacylation to several ubiquitin-
conjugating enzymes (E
2
), also called ubiquitin-
carrier proteins.
f-i
These in turn (step d) transfer the
ubiquitin to amino groups of lysine side chains of
target proteins (Prot-NH
2
). A third protein, ubiquitin-
protein ligase (E
3
), is sometimes required for this
last step. Most E
2
enzymes, acting
without E
3
, couple ubiquitin with
amines or with small basic proteins
whose cellular functions are still
unclear. For example, an E
2
which
is the product of the yeast gene
CDC34 appears to function in the
cell cycle (Fig. 11-15). Its absence
from yeast is lethal
f,i,j
and it is
clear that ubiquitin-mediated
hydrolysis of the specialized pro-
teins called cyclins is essential to
operation of the cycle.
k,l
The best understood function
of ubiquitin is in nonlysosomal
degradation of proteins.
j,m,n
Pro-
tein E
3
appears to select the proteins for degrada-
tion, binding them and catalyzing the formation of
the isopeptide linkage to substrate. After one ubiquit-
in molecule has been attached, and while still held
by E
3
, a second activated ubiquitin is coupled to Lys
48 of the first ubiquitin. This process may continue
until several molecules of ubiquitin are joined by
Gly-Lys isopeptide linkages to form a polyubiquitin
chain. Sometimes more than one lysine of the sub-
strate becomes polyubiquitinated to form branched
chains. A free -NH
3
+
group on the protein being
degraded is essential for rapid conjugation with
ubiquitin and certain N-terminal residues such as
arginine favor the conjugation and subsequent
hydrolytic breakdown. Some proteins become
attached to ubiquitin only after arginine is transferred
Stereoscopic drawing of human ubiquitin. The C terminus is at the top.
From Vijay-Kumar et al.
c
Courtesy of William J. Cook
Ub C O P
O
O
O
Adenosyl
O
Ub COO
Ub C S
O
E
1
Ub C S
O
E
2
Ub C N
O
H
Protein
E
1
E
2
E
2
NH
2
Protein
E
3
AMP
HS-E
1
AMP

ATP PP
i
E
1
a
b
c
d
525
BOX 10-C (continued)
onto their N-termini from an aminoacyl-tRNA.
o
Polyubiquitin chains serve as recognition markers
that induce rapid hydrolysis of the marked proteins
in the 2000 kDa 26S proteasome or multicatalytic
protease.
ps
This complex is discussed in Box 7-A
and again in Chapter 12. During hydrolytic destruc-
tion of the protein the ubiquitin is released for reuse
by ubiquitin carboxyl-terminal hydrolases or
isopeptidases which cleave the thioester or isopep-
tide linkages that tie ubiquitin to proteins.
t,u
While proteins may be modified to favor rapid
ubiquitination, others may be altered to protect
them from ubiquitination. For example, calmodulin
produced from a cloned gene in bacteria is a good
substrate for ubiquitination but within cells it appears
to be protected by the posttranslational conversion
of Lys 115 to trimethyllysine.
v
About 10% of the histone H2A present in higher
eukaryotes is ubiquitinated at Lys 119.
w
In the slime
mold Physarum the content of ubiquitinated histones
H2A and H2B changes rapidly during the various
stages of mitosis. Apparently, ubiquitin must be
cleaved from the histones to permit packaging of
DNA into metaphase chromosomes and must become
attached to the histones in some regions of the
chromosomes to allow unfolding of the highly
packed nucleosomes. A yeast enzyme that attaches
ubiquitin to histones is encoded by the gene RAD6,
which is required for DNA repair, sporulation, and
a
Vijay-Kumar, S., Bugg, C. E., Wilkinson, K. D., Vierstra, R. D.,
Hatfield, P. M., and Cook, W. J. (1987) J. Biol. Chem. 262,
63966399
b
Vierstra, R. D., Langan, S. M., and Schaller, G. E. (1986)
Biochemistry 25, 31053108
c
Vijay-Kumar, S., Bugg, C. E., and Cook, W. J. (1987) J. Mol. Biol.
194, 531544
d
Hershko, A. (1991) Trends Biochem. Sci. 16, 265268
e
Pickart, C. M., Kasperek, E. M., Beal, R., and Kim, A. (1994)
J. Biol. Chem. 269, 71157123
f
Jentsch, S., Seufert, W., Sommer, T., and Reins, H.-A. (1990)
Trends Biochem. Sci. 15, 195198
g
Cook, W. J., Jeffrey, L. C., Sullivan, M. L., and Vierstra, R. D.
(1992) J. Biol. Chem. 267, 1511615121
h
Cook, W. J., Jeffrey, L. C., Xu, Y., and Chau, V. (1993) Biochemis-
try 32, 1380913817
i
Blumenfeld, N., Gonen, H., Mayer, A., Smith, C. E., Siegel, N.
R., Schwartz, A. L., and Ciechanover, A. (1994) J. Biol. Chem.
269, 95749581
j
Ciechanover, A., and Schwartz, A. L. (1994) FASEB J. 8,
182191
k
Barinaga, M. (1995) Science 269, 631632
l
Dore, M., and Galas, S. (1994) FASEB J. 8, 11141121
m
Pickart, C. M. (2000) Trends Biochem. Sci. 25, 544548
n
Johnson, E. S., Ma, P. C. M., Ota, I. M., and Varshavsky, A.
(1995) J. Biol. Chem. 270, 1744217456
o
Ciechanover, A., Ferber, S., Ganoth, D., Elias, S., Hershko, A.,
and Arfin, S. (1988) J. Biol. Chem. 263, 1115511167
p
Goldberg, A. L. (1995) Science 268, 522523
q
Lwe, J., Stock, D., Jap, B., Zwickl, P., Baumeister, W., and
Huber, R. (1995) Science 268, 533539
r
Thrower, J. S., Hoffman, L., Rechsteiner, M., and Pickart, C. M.
(2000) EMBO J. 19, 94102
s
Peters, J.-M. (1994) Trends Biochem. Sci. 19, 377382
t
Stein, R. L., Chen, Z., and Melandri, F. (1995) Biochemistry 34,
1261612623
u
Wilkinson, K. D., Tashayev, V. L., OConnor, L. B., Larsen, C. N.,
Kasperek, E., and Pickart, C. M. (1995) Biochemistry 34, 14535
14546
v
Johnston, S. C., Riddle, S. M., Cohen, R. E., and Hill, C. P. (1999)
EMBO J. 18, 38773887
w
Davie, J. R., and Murphy, L. C. (1990) Biochemistry 29,
47524757
x
Mori, H., Kondo, J., and Ihara, Y. (1987) Science 235,
16411644
y
Finley, D., and Varshavsky, A. (1985) Trends Biochem. Sci. 10,
343347
z
Baker, R. T., Tobias, J. W., and Varshavsky, A. (1992) J. Biol.
Chem. 267, 2336423375
aa
Tyers, M., and Willems, A. R. (1999) Science 284, 601604
bb
Turner, G. C., Du, F., and Varshavsky, A. (2000) Nature (London)
405, 579583
cc
Hochstrasser, M. (2000) Science 289, 563564
dd
Ellgaard, L., Molinari, M., and Helenius, A. (1999) Science 286,
18821888
ee
Plemper, R. K., and Wolf, D. H. (1999) Trends Biochem. Sci. 24,
266270
other cellular processes. Terminal differentiation
of reticulocytes to form erythrocytes involves loss
of specific enzymes as well as of entire mitochon-
dria. These processes also depend upon ubiquitin.
Ubiquitin is one of the components of the paired
helical filaments present in brains of persons with
Alzheimers disease.
x
In most organisms there are two arrangements
for ubiquitin genes. There is a cluster of up to 100
tandomly repeated genes whose transcription gives
rise to polyubiquitin, a chain of ubiquitin molecules
joined by Gly-Met linkages. These must be cleaved,
perhaps by the same ubiquitin C-terminal hydrolase
that releases ubiquitin from its conjugates.
y
Other
ubiquitin genes are fused to genes encoding riboso-
mal proteins. The resulting polyproteins have the
ribosomal peptides fused to the C termini of the
ubiquitin sequences and must be proteolytically
cleaved to give mature proteins.
z
Recently a variety of modifiers of ubiquitin
ligases have been discovered
aa,bb
as have ubiquitin-
like domains in other proteins.
cc
These findings
elucidate the complexity of the sorting of proteins
and removal of improperly folded and otherwise
defective proteins from the secretory pathway and
return to the proteasomes in the cytosol.
dd,ee
They
also suggest important roles for ubiquitination in a
broad range of metabolic controls.
D. Synthesis and Turnover of Macromolecules
526 Chapter 10. An Introduction to Metabolism
TABLE 10-1
Types of Biochemical Reactions with Ionic Mechanisms
Nucleophilic displacement, often via an
additionelimination sequence
(Examples: phosphatases, kinases,
phosphomutases, ribozymes)
Addition
A.
D.
(Examples: methyl-, glycosyl-transferases)
B.
(Examples: esterases, peptidases)
C.
(Examples: sulfatases, sulfotransferases)
and
(This reaction most often occurs as a step in an enzymatic
process e.g., formation of hemiacetals, hemiketals,
hemimercaptals, carbinolamines)
(Example: mutarotase)
B. To double bond conjugated with
(Examples: acyl-CoA, fumarate hydratases, aspartase)
C. To isolated double bonds (Example: oleate hydratase)
(see p. 689)
Elimination
A. and B. Precisely the opposite of addition
C. Decarboxylative elimination
(Eliminations that are the reverse of type 2A are frequent
steps in more complex enzyme mechanisms)
Formation of stabilized enolate anions and
enamines
A.
B.
Isomerization reactions
C.
(Example: sugar isomerases)
D.
(Example: oxosteroid isomerases)
A. To polarized double bonds such as
1. 3.
4.
2.
or
C Y C B + H + YH
+
O
C Y
O
C B + + H YH
+
O
P Y
O
P B
O O O O
+ H + YH

+
O
S Y
O
S B
O O
O
O + H + YH

+
C O
C N
C O C B OH + H
+
C O
C N
C C C B C C O C
+
H
H
+
Y C C COO C C Y + CO
2
+

C C C C C C
O O
H
O

C
H N
C C
H N
C C
HN
C
H
+ +
C
O
C
OH
C
HO
C
OH
C
OH
C
O
H H
enediol
C
O
C C C
H
C
O
C C C C
O
C C C

H
B: +

B: +

B: +

B: +

B: +

B: +

527
TABLE 10-1
(continued)
Stabilized enolate anions as nucleophiles: formation of
carboncarbon bonds ( condensation)
A. Displacement on a carbonyl group
(Example: 3-Oxoacyl CoA transferase (Y = SCoA)
Some isomerization and rearrangement reactions
A. Allylic rearrangement (1,3-proton shift)
B. Allylic rearrangement with condensation
(Example: condensation of dimethylallyl pyrophosphate with
isopentenyl pyrophosphate)
C. Rearrangements with alkyl or hydride ion shift
Glyoxalase
(Examples: biosynthesis of leucine and valine,
xylose isomerase, glyoxalase)
(Examples: phosphoenolpyruvate carboxylase, oxaloacetate
decarboxylases)
C. Addition to a carbon dioxide ( carboxylation); decarboxylation
B. Addition to a carbonyl group: aldol condensation
(Examples: aldolases, citrate synthases)
5.
6.
C
O
C C
O
Y C
O
C C
O
YH + + H +

+ +
C
O
C C O C
O
C C OH + H +

+
C
O
C C C
O
C C
O
O
O
+
O

CH C C
H
H
CH
2
C C
H
H
+
R C
Y
H
2
C C C
H
R CH
2
CH
2
C C +
H H
HY
C
R
O
H
C
O
C
O
C
R
OH
C
OH
C
R
OH
H
NADH
the turnover rate of a protein is determined
in large part by its sequence. Some proteins
are tightly folded and have few bends on
the outside that have sequences meeting
the specificity requirements of intracellular
proteolytic enzymes. These have long half
lives. Other proteins may have external
loops with sequences susceptible to attack
or sequences that favor rapid reaction
with the ubiquitin system or uptake into
lysosomes. Prematurely terminated pro-
teins and peptide fragments from partial
degradation of proteins may tend to be
unfolded at the N terminus and to be
attacked rapidly.
128,135
Proteins that have
undergone covalent modifications or oxida-
tive damage also seem to be hydrolyzed
rapidly.
136
Regions rich in proline, glutamate,
serine, and threonine (PEST regions) may
be good substrates for Ca
2+
-activated cyto-
solic proteases.
137
The ubiquitin system
appears to act most slowly on a protein
when the normal initiation amino acid
methionine is present at the N terminus.
For example, the half-life of -galactosidase
in yeast is over 20 hours. Replacement
of the methionine with S, A, T, V, or G
has little effect. However, replacement
with other amino acids shortens the half-
life as follows: I and E, 30 min.; Y and Q,
10 min.; F, L, D, and K, 3 min.; and R, 2
min.
138140a
5. Turnover of Nucleic Acids
Because of its special role in carrying
genetic information, DNA is relatively
stable. An elaborate system of repair
enzymes (Chapter 27) act to correct errors
and to help DNA to preserve its genetic
information. However, in some special-
ized cells such as those forming immuno-
globulin, the DNA too undergoes major
rearrangements (Chapter 31). RNA mole-
cules are subject to extensive processing.
This includes the conversion of RNA
bases to modified forms, chain cleavages
during maturation of ribosomal and trans-
fer RNAs, cutting and splicing of gene
transcripts to form mRNAs and finally
degradation of the mRNA (Chapter 28).
Proteins that serve as RNA chaperones
assist in folding these molecules.
141
D. Synthesis and Turnover of Macromolecules
528 Chapter 10. An Introduction to Metabolism
BOX 10-D DRAWING THOSE LITTLE ARROWS
a

O
C
O
CH
2
C CH
3
NH
+
Enzyme
C CH
3
N H
Enzyme
CH
2
H
+
CH
3
C CH
3
N
Enzyme
CH
3
C CH
3
O
+ CO
2
Schiff base
H
2
NEnzyme
b
c
H
2
O
Organic mechanisms are often indicated by arrows that show the flow of electrons in individual steps of
a reaction. Many errors are made by students on exams and even in published research papers. The arrows
are often drawn backward, are too numerous, or do not clearly indicate electron flow. Here are some tips.
Groups with nucleophilic centers indicated by
unshared electron pairs and/or negative charge.
Identify the nucleophilic and electrophilic
centers before starting to write a mechanism.
Oxygen, sulfur, and nitrogen atoms are
usually nucleophilic, e.g., those in OH,
NH
2
, COO

, SH, OPO
3
2
, and enolate
anions. The weak nucleophiles OH and
SH may be converted into strong nucleo-
philes O

by removal of a proton by a
basic group of an enzyme. The nonnucleo-
philic NH
3
+
becomes the good nucleophile
NH
2
by loss of a proton. Nucleophilic
centers contain unshared electron pairs.
Nucleophiles are basic but the basicity, as
indicated by proton binding (by the pK
a
), is
not necessarily proportional to nucleophilic
strength (nucleophilicity). Enolate anions
and enamines provide nucleophilic centers
on carbon atoms, important in formation and
cleavage of C C bonds.
Electrophilic centers include acidic hydrogen
atoms, metal ions, the carbon atoms of car-
bonyl groups, and the -carbon atoms of ,
unsaturated acids, ketones, or acyl-CoA
derivatives. Highly polarized groups such as
carbonyl and enamine generate electrophilic
centers as indicated by the positive charges.
They also affect more distant positions in
conjugated systems, e.g., in , -unsaturated
acyl-CoA derivatives, and in intermediates
formed from thiamin diphosphate and pyri-
doxal phosphate.
Always write a mechanism step-
by-step. Never combine two
steps (e.g., a and b at right) in
which electron flow occurs in
opposite directions. Notice that
step c, the hydrolysis of a Schiff
base, is also a two-step process.
The reaction is a familiar one that
is commonly indicated as shown
here. However, this scheme does
not show a detailed mechanism
for step c.
1
2
Protonated imine
Carbonyl
Enamine
Some electrophilic centers are indicated by + or
+
,-Unsaturated acyl-CoA
Enolate anion
O
H
N
H
C
O
,
O
,

H
C C O C C O

C C N C C
H
+
N
H

C O C O

+
C C C S C C C S
O

O
CoA
+
CoA

N
+

+
C N
H
+
C N
H
+
529
3
4
6
BOX 10-D (continued)
When an ionic organic reaction (the kind
catalyzed by most enzymes) occurs a
nucleophilic center joins with an electro-
philic center. We use arrows to show the
movement of pairs of electrons. The move-
ment is always away from the nucleophile
which can be thought of as attacking an
electrophilic center. Notice the first step in
the second example at right. The unsatur-
ated ketone is polarized initially. However,
this is not shown as a separate step. Rather,
the flow of electrons from the double bond,
between the - and -carbons into the
electron-accepting C = O groups, is coordi-
nated with the attack by the nucleophile.
Dotted lines are often used to indicate
bonds that will be formed in a reaction
step, e.g., in an aldol condensation (right).
Dashed or dotted lines are often used to
indicate partially formed and partially
broken bonds in a transition state, e.g., for
the aldol condensation (with prior protona-
tion of the aldehyde). However, do not put
arrows on transition state structures.
For reactions of radicals (homolytic reactions) arrows are used to indicate motion of single electrons
rather than of electron pairs. It is desirable to use arrows with half-heads. For example, reaction of a
superoxide radical
(O
2

) with FADH
2
could occur as
follows:
In a given reaction step all of the arrows must
point the same way. The arrows point into bonds
that are forming or toward atoms that will carry
an unshared electron pair in the product. Arrows
originate from unshared electron pairs or from bonds
that are breaking.
Never start an arrow from the same bond that
another arrow is forming; i.e., electrons flow
out of alternate bonds.
Polarized unsaturated ketone
Aldol condensation
All arrows point the same way
Transition state
5
C C C
O O
H

+
H
H
C C C
O

O
+
E. Classifying Enzymatic Reactions
NH
2
H
+
NH
3
C C C
O
C C C
H
O

HO
C C C
O
+
HO
(Enolate anion) HO

H
+
C C C
O
C C C
O

O
C
O
C C N H C C N CO
2
+
+
H
H
3
C
H
3
C N
N
NH
N
O
O
Ribityl
H
O
O

H
3
C
H
3
C N
N
NH
N
O
O
Ribityl
H
O
OH
H
FADH
2
Half-heads on arrows indicate one-electron movement
530 Chapter 10. An Introduction to Metabolism
1. Iwata, S., Ostermeier, C., Ludwig, B., and
Michel, H. (1995) Nature 376, 660669
2. Tsukihara, T., Aoyama, H., Yamashita, E.,
Tomizaki, T., Yamaguchi, H., Shinzawa-Itoh,
K., Nakashima, R., Yaono, R., and Yoshikawa,
S. (1995) Science 269, 10691074
3. Mitchell, P. (1966) Biol. Rev. Cambridge Philos.
Soc. 41, 445502
4. Mitchell, P. (1979) Science 206, 11481159
5. Nicholls, D. G., and Ferguson, S. J. (1992)
Bioenergetics 2, Academic Press, London
6. Gething, M.-J., and Sambrook, J. (1992) Science
355, 3345
7. Frydman, J., Nimmesgern, E., Ohtsuka, K.,
and Hartl, F. U. (1994) Nature 370, 111117
8. Buchner, J. (1994) Trends Biochem. Sci. 19, 559
9. Hartl, F.-U., Hlodan, R., and Langer, T. (1994)
Trends Biochem. Sci. 19, 2025
10. Flynn, G. C., Pohl, J., Flocco, M. T., and
Rothman, J. E. (1991) Nature 353, 726730
10a. Russell, R., Jordan, R., and McMacken, R.
(1998) Biochemistry 37, 596 607
10b. Harrison, C. J., Hayer-Hartl, M., Di Liberto,
M., Hartl, F.-U., and Kuriyan, J. (1997) Science
276, 431 435
11. Wilbanks, S. M., and McKay, D. B. (1995) J.
Biol. Chem. 270, 22512257
11a. Kelley, W. L. (1998) Trends Biochem. Sci. 23,
222 227
11b. Martinez-Yamout, M., Legge, G. B., Zhang,
O., Wright, P. E., and Dyson, H. J. (2000) J.
Mol. Biol. 300, 805 818
11c. Westermann, B., and Neupert, W. (1997) J.
Mol. Biol. 272, 477483
11d. Mogk, A., Tomoyasu, T., Goloubinoff, P.,
Rdiger, S., Rder, D., Langen, H., and
Bukau, B. (1999) EMBO J. 18, 6934 6949
11e. Deuerling, E., Schulze-Specking, A.,
Tomoyasu, T., Mogk, A., and Bukau, B. (1999)
Nature (London) 400, 693 696
12. McCarty, J. S., Buchberger, A., Reinstein, J.,
and Bukau, B. (1995) J. Mol. Biol. 249, 126137
13. Flaherty, K. M., DeLuca-Flaherty, C., and
McKay, D. B. (1990) Nature 346, 623628
14. Flaherty, K. M., Wilbanks, S. M., DeLuca-
Flaherty, C., and McKay, D. B. (1994) J. Biol.
Chem. 269, 1289912907
15. Phipps, B. M., Typke, D., Hegeri, R., Volker,
S., Hoffmann, A., Stetter, K. O., and
Baumeister, W. (1993) Nature 361, 475477
15a. Schoehn, G., Hayes, M., Cliff, M., Clarke, A.
R., and Saibil, H. R. (2000) J. Mol. Biol. 301,
323332
16. Braig, D., Otwinowski, Z., Hegde, R.,
Boisvert, D. C., Joachimiak, A., Horwich, A.
L., and Sigler, P. B. (1994) Nature 371, 578586
17. Hayer-Hartl, M. K., Martin, J., and Hartl, F. U.
(1995) Science 269, 836841
18. Chen, S., Roseman, A. M., Hunter, A. S.,
Wood, S. P., Burston, S. G., Ranson, N. A.,
Clarke, A. R., and Saibil, H. R. (1994) Nature
371, 261264
18a. Betancourt, M. R., and Thirumalai, D. (1999)
J. Mol. Biol. 287, 627 644
References
18b. Wang, Z., Feng, H.-p, Landry, S. J., Maxwell,
J., and Gierasch, L. M. (1999) Biochemistry 38,
1253712546
19. Shtilerman, M., Lorimer, G. H., and
Englander, S. W. (1999) Science 284, 822 825
20. Weaver, A. J., Sullivan, W. P., Felts, S. J.,
Owen, B. A. L., and Toft, D. O. (2000) J. Biol.
Chem. 275, 23045 23052
20a. Buchner, J. (1999) Trends Biochem. Sci. 24, 136
141
20b. Sauer, F. G., Ftterer, K., Pinkner, J. S.,
Dodson, K. W., Hultgren, S. J., and Waksman,
G. (1999) Science 285, 1058 1061
20c. Bogdanov, M., and Dowhan, W. (1999) J. Biol.
Chem. 274, 3682736830
20d. Orci, L., Vassalli, J.-D., and Perrelet, A. (1988)
Sci. Am. 259(Sep), 8594
20e. Zhou, A., Webb, G., Zhu, X., and Steiner, D. F.
(1999) J. Biol. Chem. 274, 20745 20748
21. Smeekens, S. P., Montag, A. G., Thomas, G.,
Albiges-Rizo, C., Carroll, R., Benig, M.,
Phillips, L. A., Martin, S., Ohagi, S., Gardner,
P., Swift, H. H., and Steiner, D. F. (1992) Proc.
Natl. Acad. Sci. U.S.A. 89, 88228826
22. Huang, X. F., and Arvan, P. (1995) J. Biol.
Chem. 270, 2041720423
23. Blobel, G., and Sabatini, D. D. (1971) in
Biomembranes, Vol. 2 (Manson, L. A., ed), pp.
193195, Plenum, New York
24. Blobel, G., and Dobberstein, B. (1975) J. Cell
Biol. 67, 852
E. Classifying Enzymatic Reactions
The majority of enzymes appear to contain, in their
active centers, only the side chains of amino acids.
Most of the reactions catalyzed by these enzymes can
be classified into a small number of types as is indicated
in Table 10-1: (Type 1) displacement or substitution
reactions in which one base or nucleophile replaces
another, (Type 2) addition reactions in which a reagent
adds to a double bond, and (Type 3) elimination
reactions by which groups are removed from a substrate
to create double bonds. Note that the latter are the
reverse of addition reactions. Two other groups of
reactions depend upon formation of transient enolic
forms. These include (Type 4) isomerases and
(Type 5) lyases, a large and important group of
reactions that form or cleave carboncarbon bonds.
Finally, there is a group (Type 6) of isomerization
and rearrangement reactions that do not appear to
fit any of the foregoing categories. Another quite differ-
ent group of enzyme-catalyzed reactions, which are
considered in Chapter 16, function with the participa-
tion of free radical intermediates.
Biochemical displacement reactions include all
of the hydrolytic reactions by which biopolymers are
broken down to monomers as well as most of the
reactions by which the monomers are linked together
to form polymers. Addition reactions are used to
introduce oxygen, nitrogen, and sulfur atoms into
biochemical compounds and elimination reactions often
provide the driving force for biosynthetic sequences.
Complex enzymatic processes are often combinations
of several steps involving displacement, addition, or
elimination. The reactions involving formation or
cleavage of CC bonds are essential to biosynthesis
and degradation of the various carbon skeletons found
in biomolecules, while the isomerization reactions
provide connecting links between the other kinds of
reactions in the establishment of metabolic pathways.
In Chapters 12 and 13 the individual reactions
of metabolism are classified into these types and the
enzymes that catalyze them are described in some detail.
The chemistry of coenzymes and metalloenzymes are
presented systematically in Chapters 14 to 16, and in
Chapter 17 the logic of the combining of individual
reactions into metabolic sequences is considered. It is
not necessary to read Chapters 1216 in their entirety
since much of their content is reference material. In
the later chapters on metabolism, cross-references
point out the discussions of individual enzymes in
Chapters 1216.
The following are topics that may be especially
valuable to the student and which might be read
initially: in Chapter 12, lysozyme (Section B,5), chymo-
trypsin (Section C,1), kinases (Section D,9), multiple
displacement, reactions (Section G); in Chapter 13,
imines (Section A,2), addition to C=C bonds (Section
A, 4,5), beta cleavage and condensation (Section C); in
Chapter 14, thiamin diphosphate (Section D), pyridoxal
phosphate (Section E); in Chapter 15, NAD (Section A).
531
References
25. Landry, S. J., and Gierasch, L. M. (1991) Trends
Biochem. Sci. 16, 159163
26. Gierasch, L. M. (1989) Biochemistry 28, 923930
27. Jain, R. G., Rusch, S. L., and Kendall, D. A.
(1994) J. Biol. Chem. 269, 1630516310
28. Andersson, H., and von Heijne, G. (1993) J.
Biol. Chem. 268, 2138921393
28a. von Heijne, G. (1998) Nature (London) 396, 1
11 113
28b. Emanuelsson, O., Nielsen, H., Brunak, S., and
von Heijne, G. (2000) J. Mol. Biol. 300, 10051016
29. Verner, K., and Schatz, G. (1988) Science 241,
13071313
30. Rapoport, T. A. (1992) Science 258, 931935
30a. Batey, R. T., Rambo, R. P., Lucast, L., Rha, B.,
and Doudna, J. A. (2000) Science 287, 12321239
31. Sanders, S. L., and Schekman, R. (1992) J. Biol.
Chem. 267, 1379113794
32. Hartmann, E., Sommer, T., Prehn, S., Grlich,
D., Jentsch, S., and Rapoport, T. A. (1994)
Nature 367, 654657
32a. Clemons, W. M., Jr., Gowda, K., Black, S. D.,
Zwieb, C., and Ramakrishnan, V. (1999) J. Mol.
Biol. 292, 697 705
33. Wickner, W. T. (1994) Science 266, 11971198
33a. Moser, C., Mol, O., Goody, R. S., and Sinning,
I. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 11339
11344
33b. Samuelson, J. C., Chen, M., Jiang, F., Mller, I.,
Wiedmann, M., Kuhn, A., Phillips, G. J., and
Dalbey, R. E. (2000) Nature (London) 406, 637
640
33c. Paetzel, M., Strynadka, N. C. J., Tschantz, W.
R., Casareno, R., Bullinger, P. R., and Dalbey,
R. E. (1997) J. Biol. Chem. 272, 9994 10003
33d. Guo, W., Grant, A., and Novick, P. (1999) J.
Biol. Chem. 274, 2355823564
34. Nol, P. J., and Cartwright, I. L. (1994) EMBO
J. 13, 52535261
35. Brodsky, J. L., Goeckeler, J., and Schekman, R.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 96439646
36. Dobberstein, B. (1994) Nature 367, 599600
37. Laidler, V., Chaddock, A. M., Knott, T. G.,
Walker, D., and Robinson, C. (1995) J. Biol.
Chem. 270, 1766417667
38. Dalbey, R. E., and Robinson, C. (1999) Trends
Biochem. Sci. 24, 17 22
39. Douville, K., Price, A., Eichler, J., Economou,
A., and Wickner, W. (1995) J. Biol. Chem. 270,
2010620111
39a. Eichler, J., Brunner, J., and Wickner, W. (1997)
EMBO J. 16, 2188 2196
39b. Matsumoto, G., Yoshihisa, T., and Ito, K. (1997)
EMBO J. 16, 6384 6393
39c. Scotti, P. A., Urbanus, M. L., Brunner, J., de
Gier, J.-W. L., von Heijne, G., van der Does,
C., Driessen, A. J. M., Oudega, B., and
Luirink, J. (2000) EMBO J. 19, 542549
40. Meyer, T. H., Mntret, J.-F., Breitling, R.,
Miller, K. R., Akey, C. W., and Rapoport, T. A.
(1999) J. Mol. Biol. 285, 17891800
40a. Yahr, T. L., and Wickner, W. T. (2000) EMBO J.
19, 43934401
41. Randall, L. L., and Hardy, S. J. S. (1995) Trends
Biochem. Sci. 20, 6569
42. Ulbrandt, N. D., London, E., and Oliver, D. B.
(1992) J. Biol. Chem. 267, 1518415192
43. Bergeron, J. J. M., Brenner, M. B., Thomas, D.
Y., and Williams, D. B. (1994) Trends Biochem.
Sci. 19, 124128
44. Jungery, M., Pasvol, G., Newbold, C. I., and
Weatherall, D. J. (1983) Proc. Natl. Acad. Sci.
U.S.A. 80, 10181022
45. Cao, G., Kuhn, A., and Dalbey, R. E. (1995)
EMBO J. 14, 866875
46. Kawasaki, S., Mizushima, S., and Tokuda, H.
(1993) J. Biol. Chem. 268, 81938198
47. Wickner, W., Driessen, A. J. M., and Hartl, F.-
U. (1991) Ann. Rev. Biochem. 60, 101124
48. Matsuyama, S.-i, Tajima, T., and Tokuda, H.
(1995) EMBO J. 14, 33653372
48a. Schfer, U., Beck, K., and Mller, M. (1999) J.
Biol. Chem. 274, 2456724574
48b. Russel, M. (1998) J. Mol. Biol. 279, 485499
48c. Galn, J. E., and Collmer, A. (1999) Science
284, 13221328
48d. Tamano, K., Aizawa, S.-I., Katayama, E.,
Nonaka, T., Imajoh-Ohmi, S., Kuwae, A.,
Nagai, S., and Sasakawa, C. (2000) EMBO J.
19, 38763887
49. Schatz, G. (1993) Protein Sci. 2, 141146
50. Pfeffer, S. R., and Rothman, J. E. (1987) Ann.
Rev. Biochem. 56, 829852
51. Lodish, H. F. (1988) J. Biol. Chem. 263, 21072110
52. Smeekens, S., Bauerle, C., Hageman, J., Keegstra,
K., and Weisbeek, P. (1986) Cell 46, 365375
53. Roise, D., and Schatz, G. (1988) J. Biol. Chem.
263, 45094511
54. Hurt, E. C., and van Loon, A. P. G. M. (1986)
Trends Biochem. Sci. 11, 204207
55. Aitchison, J. D., Murray, W. W., and
Rachubinski, R. A. (1991) J. Biol. Chem. 266,
2319723203
56. Wolins, N. E., and Donaldson, R. P. (1994) J.
Biol. Chem. 269, 11491153
57. Takeda, J., and Kinoshita, T. (1995) Trends
Biochem. Sci. 20, 367371
57a. Mazmanian, S. K., Liu, G., Ton-That, H., and
Schneewind, O. (1999) Science 285, 760 762
58. Ogata, S., and Fukuda, M. (1994) J. Biol. Chem.
269, 52105217
59. Pond, L., Kuhn, L. A., Teyton, L., Schutze, M.-
P., Tainer, J. A., Jackson, M. R., and Peterson,
P. A. (1995) J. Biol. Chem. 270, 1998919997
60. Pelham, H. R. B. (1990) Trends Biochem. Sci. 15,
483486
61. Wilson, D. W., Lewis, M. J., and Pelham, H. R.
B. (1993) J. Biol. Chem. 268, 74657468
62. Mallabiabarrena, A., Jimnez, M. A., Rico, M.,
and Alarcn, B. (1995) EMBO J. 14, 22572268
63. Luzio, J. P., and Banting, G. (1993) Trends
Biochem. Sci. 18, 395398
64. Peter, F., Van, P. N., and Soling, H.-D. (1992) J.
Biol. Chem. 267, 1063110637
65. Beh, C. T., and Rose, M. D. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 98209823
66. Hoe, M. H., Slusarewicz, P., Misteli, T.,
Watson, R., and Warren, G. (1995) J. Biol.
Chem. 270, 2505725063
67. Schneider, H.-C., Berthold, J., Bauer, M. F.,
Dietmeier, K., Guiard, B., Brunner, M., and
Neupert, W. (1994) Nature 371, 768773
68. Bhattacharyya, T., Karnezis, A. N., Murphy,
S. P., Hoang, T., Freeman, B. C., Phillips, B.,
and Morimoto, R. I. (1995) J. Biol. Chem. 270,
17051710
69. Schmitt, M., Neupert, W., and Langer, T.
(1995) EMBO J. 14, 34343444
70. Stuart, R. A., Cyr, D. M., Craig, E. A., and Neupert,
W. (1994) Trends Biochem. Sci. 19, 8792
71. Lithgow, T., Glick, B. S., and Schatz, G. (1995)
Trends Biochem. Sci. 20, 98101
72. Pfanner, N., Craig, E. A., and Meijer, M. (1994)
Trends Biochem. Sci. 19, 368372
73. Mayer, A., Nargang, F. E., Neupert, W., and
Lill, R. (1995) EMBO J. 14, 42044211
73a. Heard, T. S., and Weiner, H. (1998) J. Biol.
Chem. 273, 2938929393
74. Grlich, D., Vogel, F., Mills, A. D., Hartmann, E.,
and Laskey, R. A. (1995) Nature 377, 246248
74a. Koehler, C. M., Merchant, S., and Schatz, G.
(1999) Trends Biochem. Sci. 24, 428432
75. Smeekens, S., Weisbeek, P., and Robinson, C.
(1990) Trends Biochem. Sci. 15, 7376
76. Pilon, M., Wienk, H., Sips, W., de Swaaf, M.,
Talboom, I., van t Hof, R., de Korte-Kool, G.,
Demel, R., Weisbeek, P., and de Kruijff, B.
(1995) J. Biol. Chem. 270, 38823893
77. Schnell, D. J., Kessler, F., and Blobel, G. (1994)
Science 266, 10071011
78. Voelker, R., and Barkan, A. (1995) EMBO J. 14,
39053914
79. Viitanen, P. V., Schmidt, M., Buchner, J.,
Suzuki, T., Vierling, E., Dickson, R., Lorimer,
G. H., Gatenby, A., and Soll, J. (1995) J. Biol.
Chem. 270, 1815818164
79a. Nakai, K., and Horton, P. (1999) Trends
Biochem. Sci. 24, 3435
79b. Klionsky, D. J. (1998) J. Biol. Chem. 273, 10807
10810
80. Rothman, J. E. (1994) Nature 372, 5563
81. Pryer, N. K., Wuestehube, L. J., and
Schekman, R. (1992) Ann. Rev. Biochem. 61,
471516
82. Bennett, M. K., and Scheller, R. H. (1993) Proc.
Natl. Acad. Sci. U.S.A. 90, 25592563
83. Duden, R., Hosobuchi, M., Hamamoto, S.,
Winey, M., Byers, B., and Schekman, R. (1994)
J. Biol. Chem. 269, 2448624495
84. Edelmann, L., Hanson, P. I., Chapman, E. R.,
and Jahn, R. (1995) EMBO J. 14, 224231
84a. Dulubova, I., Sugita, S., Hill, S., Hosaka, M.,
Fernandez, I., Sdhof, T. C., and Rizo, J.
(1999) EMBO J. 18, 43724382
84b. Katz, L., Hanson, P. I., Heuser, J. E., and
Brennwald, P. (1998) EMBO J. 17, 62006209
84c. Gerona, R. R. L., Larsen, E. C., Kowalchyk, J.
A., and Martin, T. F. J. (2000) J. Biol. Chem. 275,
63286336
85. Morgan, A., and Burgoyne, R. D. (1995)
EMBO J. 14, 232239
85a. Glockshuber, R. (1999) Nature (London) 401,
30 31
85b. Bolhuis, A., Venema, G., Quax, W. J., Bron, S.,
and van Dijl, J. M. (1999) J. Biol. Chem. 274,
24531 24538
85c. Ban, N., Nissen, P., Hansen, J., Moore, P. B.,
and Steitz, T. A. (2000) Science 289, 905 920
85d. Berardi, M. J., and Bushweller, J. H. (1999)
J. Mol. Biol. 292, 151161
85e. van den Berg, B., Chung, E. W., Robinson, C.
V., Mateo, P. L., and Dobson, C. M. (1999)
EMBO J. 18, 4794 4803
86. Freedman, R. B., Hirst, T. R., and Tuite, M. F.
(1994) Trends Biochem. Sci. 19, 331335
87. Martin, J. L., Bardwell, J. C. A., and Kuriyan,
J. (1993) Nature 365, 465468
88. Kishigami, S., Kanaya, E., Kikuchi, M., and
Ito, K. (1995) J. Biol. Chem. 270, 1707217074
89. Jander, G., Martin, N. L., and Beckwith, J.
(1994) EMBO J. 13, 51215127
90. Kanaya, E., Anaguchi, H., and Kikuchi, M.
(1994) J. Biol. Chem. 269, 42734278
91. Frech, C., and Schmid, F. X. (1995) J. Biol.
Chem. 270, 53675374
92. Chivers, P. T., Laboissire, M. C. A., and
Raines, R. T. (1996) EMBO J. 15, 2659 2667
93. Tschantz, W. R., Paetzel, M., Cao, G., Suciu,
D., Inouye, M., and Dalbey, R. E. (1995)
Biochemistry 34, 39353941
94. Steiner, D. F., Smeekens, S. P., Ohagi, S., and
Chan, S. J. (1992) J. Biol. Chem. 267, 23435
23438
95. De Bie, I., Savaria, D., Roebroek, A. J. M., Day,
R., Lazure, C., Van de Ven, W. J. M., and Seidah,
N. G. (1995) J. Biol. Chem. 270, 10201028
96. Fisher, J. M., and Scheller, R. H. (1988) J. Biol.
Chem. 263, 1651516518
97. Rehfeld, J. F., Hansen, C. P., and Johnsen, A.
H. (1995) EMBO J. 14, 389396
98. Ypma-Wong, M. F., Filman, D. J., Hogle, J. M.,
and Semler, B. L. (1988) J. Biol. Chem. 263,
1784617856
99. Bateman, A., Solomon, S., and Bennett, H. P. J.
(1990) J. Biol. Chem. 265, 2213022136
100. Richter, D. (1983) Trends Biochem. Sci. 8, 278280
532 Chapter 10. An Introduction to Metabolism
Study Questions
101. Douglass, J., Civelli, O., and Herbert, E. (1984)
Ann. Rev. Biochem. 53, 665715
102. Arfin, S. M., and Bradshaw, R. A. (1988)
Biochemistry 27, 79797984
103. Tolan, D. R., Amsden, A. B., Putney, S. D.,
Urdea, M. S., and Penhoet, E. E. (1984) J. Biol.
Chem. 259, 11271131
104. Rubenstein, P. A., and Martin, D. J. (1983) J.
Biol. Chem. 258, 39613966
105. Sheff, D. R., and Rubenstein, P. A. (1992) J.
Biol. Chem. 267, 2021720224
106. Kulkarni, M. S., and Sherman, F. (1994) J. Biol.
Chem. 269, 1314113147
107. Gan, K., Gupta, S. D., Sankaran, K., Schmidt,
M. B., and Wu, H. C. (1993) J. Biol. Chem. 268,
1654416550
108. Vogt, A., Sun, J., Qian, Y., Tan-Chiu, E.,
Hamilton, A. D., and Sebti, S. M. (1995)
Biochemistry 34, 1239812403
109. Parish, C. A., and Rando, R. R. (1994)
Biochemistry 33, 99869991
110. Pompliano, D. L., Rands, E., Schaber, M. D.,
Mosser, S. D., Anthony, N. J., and Gibbs, J. B.
(1992) Biochemistry 31, 38003807
111. Busby, W. H., Jr., Quackenbush, G. E., Humm,
J., Youngblood, W. W., and Kizer, J. S. (1987) J.
Biol. Chem. 262, 85328536
112. Fischer, W. H., and Spiess, J. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 36283632
113. Katopodis, A. G., Ping, D., Smith, C. E., and
May, S. W. (1991) Biochemistry 30, 61896194
References
114. Merkler, D. J., Kulathila, R., Consalvo, A. P.,
Young, S. D., and Ash, D. E. (1992) Biochemis-
try 31, 72827288
115. Ping, D., Katopodis, A. G., and May, S. W.
(1992) J. Am. Chem. Soc. 114, 39984000
116. Eipper, B. A., Milgram, S. L., Husten, E. J.,
Yun, H.-Y., and Mains, R. E. (1993) Protein Sci.
2, 489497
117. Bradbury, A. F., and Smyth, D. G. (1991)
Trends Biochem. Sci. 16, 112115
118. Low, M. G., and Saltiel, A. R. (1988) Science
239, 268275
119. Lisanti, M. P., and Rodriguez-Boulan, E.
(1990) Trends Biochem. Sci. 15, 113118
120. Graves, D. J., Martin, B. L., and Wang, J. H.
(1994) Co-and post-translational modification of
proteins, Oxford Univ. Press, New York
121. Tuboi, S., Taniguchi, N., and Katunuma, N.,
eds. (1992) Post-Translation Modification of
Proteins, CRC Press, Boca Raton, Florida
122. Barrett, G. C., ed. (1985) Chemistry and
Biochemistry of the Amino Acids, Chapman and
Hall, London; New York
123. Niehrs, C., Kraft, M., Lee, R. W. H., and Huttner,
W. B. (1990) J. Biol. Chem. 265, 85258532
124. Rucker, R. B., and Wold, F. (1988) FASEB J. 2,
22522261
125. Klotz, A. V., Leary, J. A., and Glazer, A. N.
(1986) J. Biol. Chem. 261, 1589115894
126. Oliver, C. N., Ahn, B., Moerman, E. J.,
Goldstein, S., and Stadtman, E. R. (1987) J.
Biol. Chem. 262, 54885491
1. Outline in detail, using structural formulas, the
enzyme-catalyzed reactions by which cells in the
human body convert glyceraldehyde 3-phosphate
into pyruvate.
2. Describe, using chemical structural formulas, the
reactions involved in the breakdown of glycogen
to glucose 1-phosphate and the synthesis of glyco-
gen from glucose 1-phosphate.
3. Describe the reaction steps in gluconeogenesis by
which pyruvate is converted into glyceraldehyde
3-phosphate.
4. Compare the reactions of pyruvate that give rise
to the following three compounds. List coenzymes
or electron-carriers involved in each case and indi-
cate any intermediate compounds.
a) Ethanol
b) Lactic acid
c) Acetyl-Coenzyme A
5. Mammalian sperm cells metabolize D-fructose
preferentially as a source of energy. Fructose is
formed in cells of the seminal vesicle from D-glucose
via reduction to the sugar alcohol sorbitol using
NADPH, followed by oxidation of sorbitol to
fructose using NAD
+
. The fructose concentration
in human semen is about 12 mM, whereas the
glucose concentration within cells is usually less
than 1 mM. If the ratio [ NADPH ] / [ NADP
+
] is
10
4
times higher than the ratio [ NADH ] / [ NAD
+
],
what is the minimum glucose concentration in
cells that could allow formation of 12 mM fruc-
tose? The standard Gibbs energies of formation
from the elements G
o
f
in kJ/ mol at 25C are:
D-glucose 917, D-fructose 915.
6. Outline the reactions by which glyceraldehyde
3-phosphate is converted to 3-phosphoglycerate
with coupled synthesis of ATP in the glycolysis
pathway. Show important mechanistic details.
7. Why cant acetyl CoA be converted to glucose in
animals?
8. Describe the parallel reaction sequences between
the citric acid cycle and the oxidation pathway.
9. Contrary to legend, camels do not store water in
their humps, which actually consist of a large fat
deposit. How can these fat deposits serve as a
source of water? Calculate the amount of water
(liters) that can be produced by the camel from
500 g of fat. Assume for simplicity that the fat
consists entirely of tripalmitin.
127. Cooper, A. A., and Stevens, T. H. (1995) Trends
Biochem. Sci. 20, 351356
128. Rechsteiner, M., Rogers, S., and Rote, K.
(1987) Trends Biochem. Sci. 12, 390394
129. Stadtman, E. R. (1986) Trends Biochem. Sci. 11,
1112
130. Rivett, A. J. (1985) J. Biol. Chem. 260, 300 305
131. Loshon, C. A., Swerdlow, B. M., and Setlow, P.
(1982) J. Biol. Chem. 257, 1083810845
132. Olden, K., Parent, J. B., and White, S. C. (1982)
Biochim. Biophys. Acta. 650, 209232
133. Chiang, H.-L., and Dice, J. F. (1988) J. Biol.
Chem. 263, 67976805
133a. DellAngelica, E. C., Mullins, C., Caplan, S., and
Bonifacino, J. S. (2000) FASEB J. 14, 12651278
134. Hershko, A., and Ciechanover, A. (1992) Ann.
Rev. Biochem. 61, 761807
135. Dice, J. F. (1987) FASEB J. 1, 349357
136. Stadtman, E. R. (1990) Biochemistry 29, 63236331
137. Rogers, S., Wells, R., and Rechsteiner, M.
(1986) Science 234, 364368
138. Bachmair, A., Finley, D., and Varshavsky, A.
(1986) Science 234, 179186
139. Gonda, D. K., Bachmair, A., Wnning, I.,
Tobias, J. W., Lane, W. S., and Varshavsky, A.
(1989) J. Biol. Chem. 264, 1670016712
140. Madura, K., and Varshavsky, A. (1994) Science
265, 14541458
140a. Davydov, I. V., and Varchavsky, A. (2000) J.
Biol. Chem. 275, 2293122941
141. Herschlag, D. (1995) J. Biol. Chem. 270, 20871
20874
533
Study Questions
10. A little-known microorganism carries out the
following net reaction in a series of enzymatic steps:
heptanoyl CoA + CoASH + NAD
+
+
FAD + H
2
O pentanoyl CoA +
acetyl CoA + NADH + FADH
2
+ H
+
The net result is exactly the same as that of the
oxidation pathway, but for this microorganism,
the pathway is demonstrably different in that
pentanal, CH
3
(CH
2
)
3
CHO, is an intermediate in
the sequence. Which enzyme(s) of the oxidation
pathway does this microorganism lack? Propose
an enzymatic reaction to account for the formation
of this intermediate and a series for its conversion
to the acyl CoA. ATP is not required.
11. Most natural fatty acids are even-numbered. What
is the product of the final thiolase reaction to form
an odd-chain fatty acid? Give a brief explanation.
12. It has been calculated that an average man takes
in 1.5 kg of solid food and 1.4 kg of water per day.
He gives off 3.5 kg of waste and 0.75 kg of sweat.
It would thus appear that he should be losing over
1 kg per day through normal activites. How do
you account for the fact that his weight remains
relatively constant?
13. Suppose that fatty acids, instead of being broken
down in two carbon fragments, were metabolized
in three carbon units. What product(s) would
Dr. Knoop have observed in the urine of his
experimental dogs?
14. A renowned pharmacologist has announced the
discovery of a new drug that specifically inhibits
the fatty acid oxidation pathway within minutes
of ingestion. The effects last for several hours only.
The drug has no other effects on the subjects. The
pharmacologist argues that this will increase athletic
performance by shifting oxidative metabolism
entirely to the more rapidly mobilized carbohydrate
degradation pathway. Assume that the drug does
work exactly as he suggests. Explain in a sentence or
two how the drug would affect the performance of
a) a sprinter in the 100 meter dash
b) a long-distance runner in a marathon
15. Some bacteria use a dicarboxylic acid cycle to
oxidize glyoxylate OHC COO

to CO
2
. The
regenerating substrate for this cycle is acetyl-CoA.
It is synthesized from glyoxylate by a complex
pathway that begins with conversion of two mole-
cules of glyoxylate to tartronic semialdehyde:

OOC CHOH CHO. The latter is then dehydro-


genated to D-glycerate. Write out a detailed scheme
for the dicarboxylate cycle. Also indicate how
glucose and other cell constituents can be formed
from intermediates created in this biosynthetic
pathway.
16. Write a balanced fermentation sequence by which
glycogen can be converted rapidly to sn-glycerol
3-phosphate and pyruvate in insect flight muscle.
How many molecules of ATP per glucose unit of
glycogen will be formed?
17. Show which parts (if any) of the citric acid cycle
are utilized in each of the following reactions and
what if any additional enzymes are needed in each
case.
a) Oxidation of acetyl-CoA to CO
2
b) Catabolism of glutamate to CO
2
c) Biosynthesis of glutamate from pyruvate
d) Formation of propionate from pyruvate
18. a) If the Gibbs energy change G (pH 7) for the
reaction A B is + 25 kJ/ mol at 25C, what would
the ratio of [ B ] / [ A ] be at equilibrium?
b) Suppose that the reaction were coupled to the
cleavage of ATP as follows:
Suppose further that the group transfer potential
( G ) for the phospho group of A P at 25C, pH
7 is 12 kJ/mol and that the equilibrium constant
for conversion of A P to B P is the same as that
for conversion of A to B. Calculate the concentra-
tions of A, B, A P and B P at equilibrium if the
phosphorylation state ratio R
p
is 10
4
M
1
.
19. This problem refers to the 13 meter climb described
in Chapter 6, Study Question 14.
a) Assuming that muscle accounts for 30% of total
body mass, estimate the amounts (mmol / kg,
total mmol, and total grams) of each of the
following in their coenzyme forms: nicotinamide,
pantothenic acid, thiamin. You may be able to
obtain rough estimates from the vitamin content
of pork.
b) Calculate how many times, on the average,
each molecule of nicotinamide would undergo
reduction and re-oxidation (i.e., turn over)
during the climb. Do the same for pantothenic
acid (cycling between acyl-CoA and free CoA
forms) and for thiamin through its catalytic
cycles.
A AP BP B
ATP ADP H
2
O P
i
534 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Contents
535
538
538
538
539
539
539
540
541
541
541
544
545
545
549
549
552
553
553
556
557
558
559
559
561
561
563
563
563
566
566
567
567
568
568
569
571
571
576
576
578
580
581
587
A. Pacemakers and the Control of Metabolic Flux
B. Genetic Control of Enzyme Synthesis
1. One Gene or Many?
2. Repression, Induction, and Turnover
3. Differences among Species
C. Regulation of the Activity of Enzymes
1. Allosteric Control
Regulatory subunits
Glycolysis and gluconeogenesis
2. Covalent Modification by Phosphorylation and
Dephosphorylation
Protein kinases and cyclic AMP
Protein phophatases
Phophorylation in bacteria
3. Other Modification Reactions Involving Group
Transfer
4. Thiol disulfide Equilibria
5. Regulatory Effects of H
+
, Ca
2+
, and Other
Specific Ions
6. Compartments and Organized Assemblies
D. Hormones and Their Receptors
1. Beta Adrenergic Receptors and Related
Seven-Helix Proteins
2. Adenylate Cyclases (Adenylyl Cyclases)
3. Guanine Nucleotide-Binding Proteins
(G Proteins)
Monomeric G proteins
Acylation and prenylation
Three-dimensional structures
4. Guanylate Cyclase (Guanylyl Cyclase), Nitric
Oxide, and the Sensing of Light
5. Bacterial Chemoreceptors
E. Calcium, Inositol Polyphosphates, and
Diacylglycerols
1. Alpha Adrenergic Receptors
2. Phosphatidylinositol and the Release of
Calcium Ions
F. Regulatory Cascades
1. Advantages of Regulatory Cascades
2. Substrate Cycles
G. Insulin and Related Growth-Regulating
Hormones
1. Metabolic Effects of Insulin
2. Insulin Receptors
3. A Second Messenger for Insulin?
H. Growth Factors, Oncogenes, and the Cell Cycle
1. Oncogenes and Proto-oncogenes
The ras oncogenes
Transcription factors
2. The MAP Kinase Cascade
3. The Cell Cycle and Control of Growth
References
Study Questions
546
550
557
573
543
554
572
577
Boxes
Box 11-A Cholera Toxin and Other Dangerous
Proteins
Box 11-B Glutathione, Intracellular Oxidation
Reduction Buffer
Box 11-C The Attraction of Dictyostelium to
Cyclic AMP
Box 11-D Cancer
Tables
Table 11-1 A Few Covalent Modification Reactions
Utilized to Control Metabolism
Table 11-2 Some Molecules (Second Messengers)
That Carry Intracellular Signals
Table 11-3 A Few Oncogenes That Have Interested
Biochemists
Table 11-4 A Few Abbreviations Used in Discussions
of Cell Signaling
Intracellular
Extracellular
7 1
2
3
4
5
6
S S
C
C O
O
O
C
H O O
N
1
2
3
4
7
5
6
S S S165
D113
D79
Y316
S207
S204
O
O
O
O O
H
H
H
H H
H O
H
HN H
H
+
Much of the control of cellular metabolism is accomplished by hormones and other molecules
that bind to receptor proteins embedded in the plasma membrane. Left: Many receptors are
seven-helix transmembrane proteins, the best known being the light receptors of the rhodopsin
family (Chapter 23). Center: A related adrenergic receptor, viewed here from the extracellular
side, has a molecule of the hormone adrenaline (green) bound in the center (Fig. 11-6). Right: These
receptors interact with GTP-hydrolyzing G proteins, which pass signals, often for short periods
of time, to enzymes and other proteins. One structural domain of a G protein (Fig. 11-7C) shown
here contains a propeller domain which binds to the Ras-like domain of the subunit. A few
residues of the latter are visible in the center of this image.
535
Living cells must operate with controls that provide
a stable environment and a relatively constant supply
of materials needed for biosynthesis and for meeting
the energy needs of cells. They must also be responsive
to changes in their environment and must be able to
undergo mitosis and reproduce when appropriate.
The necessary control of metabolism and of growth
is accomplished largely through mechanisms that
regulate the locations, the amounts, and the catalytic
activities of enzymes. The purpose of this chapter is
to summarize these control mechanisms and to intro-
duce terminology and shorthand notations that will
be used throughout this book. Many of the control
elements considered are summarized in Fig. 11-1.
A. Pacemakers and the Control of Metabolic
Flux
Metabolic control can be understood to some
extent by focusing attention on those enzymes that
catalyze rate-limiting steps in a reaction sequence.
Such pacemaker enzymes
14
are often involved in
reactions that determine the overall respiration rate of
a cell, reactions that initiate major metabolic sequences,
or reactions that initiate branch pathways in metabolism.
Often the first step in a unique biosynthetic pathway
for a compound acts as the pacemaker reaction. Such
a reaction may be described as the committed step of
the pathway. It usually proceeds with a large decrease
in Gibbs energy and tends to be tightly controlled.
Both the rate of synthesis of the enzyme protein and
the activity of the enzyme, once it is formed, may be
inhibited by feedback inhibition which occurs when
an end product of a biosynthetic pathway accumulates
and inhibits the enzyme. Enzyme activity may also be
turned on or off by the effect of a hormone, by some
other external stimulus, or by internal mechanisms
that sense the metabolic state of the cell. Enzymes,
other than the pacemaker, that catalyze reactions in a
pathway may not be regulated and may operate at a
steady state close to equilibrium.
If conditions within a cell change, a pacemaker
reaction may cease to be rate limiting. A reactant
plentiful in one circumstance may, in another, be de-
pleted to the point that the rate of its formation from
a preceding reaction determines the overall rate. Thus,
as we have seen in Chapter 10, metabolism of glucose
in our bodies occurs through the rapidly interconvertible
phosphate esters glucose 6-phosphate and fructose
6-phosphate. The pacemaker enzyme in utilization of
glucose or of glycogen is often phosphofructokinase
(Fig. 11-2, step b) which catalyzes further metabolism
of fructose 6-phosphate. However, if metabolism by
this route is sufficiently rapid, the rate of formation
of glucose 6-phosphate from glucose catalyzed by
hexokinase (Fig. 11-2, step a) may become rate limiting.
Some catabolic reactions depend upon ADP, but
under most conditions its concentration is very low
because it is nearly all phosphorylated to ATP. Reactions
utilizing ADP may then become the rate-limiting pace-
makers in reaction sequences. Depletion of a reactant
sometimes has the effect of changing the whole pattern
of metabolism. Thus, if oxygen is unavailable to a yeast,
the reduced coenzyme NADH accumulates and reduces
pyruvate to ethanol plus CO
2
(Fig. 10-3). The result is
a shift from oxidative metabolism to fermentation.
Pacemaker enzymes are often identified by the
fact that the measured mass action ratio, e.g., for the
reaction A + B P + Q ,the ratio [P][Q]/[A][B], is far
The Regulation of Enzymatic
Activity and Metabolism
11
Intracellular
Extracellular
7
1
2
3
4
5
6
S S
C
C
O
O

O
C
H O
O
N
1
2
3
4
7
5
6
S S S165
D113
D79
Y316
S207
S204
O
O
O
O
O
H
H
H
H
H
H
O
H
H
N
H
H
+
536 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
(11-1)
from that predicted by the known equilibrium constant
for the reaction. Another approach to identifying a
pacemaker enzyme is to inhibit it and observe resulting
changes in steady-state metabolite concentrations.
5
If
the pacemaker enzyme is inhibited, its substrate and
other compounds preceding the step catalyzed by this
enzyme will accumulate. At the same time the concen-
trations of products of the pacemaker reaction will
drop as a result of their relatively more rapid removal
by enzymes catalyzing subsequent steps. However,
conclusions drawn by this approach may sometimes
be erroneous.
6
In spite of its usefulness, the pacemaker concept
is oversimplified. It is often impossible to identify a
specific pacemaker enzyme. When both catabolism
and biosynthesis occur (e.g., as in the scheme in Fig.
11-2) it may be more useful to model the entire system
with a computer than to try to identify pacemakers.
7,8
It is also important to realize that reaction rates may be
determined by the rate of diffusion of a compound
through a membrane. Thus, membrane transport
processes can serve as pacemakers.
A general approach to analysis of complex meta-
bolic pathways or to cell growth was introduced by
Savageau,
911
and similar approaches have been followed
by others.
3,12 19
They emphasize the flux of material
through a pathway under steady-state conditions and
recognize that every enzyme in a sequence can have
some effect on the overall rate. Consider a chain of
enzymes E
1
to E
n
acting on substrate S
0
, which is
converted via substrates S
1
, S
2
... S
n1
into product S
n
:
The flux F = d[S
n
]/dt is a constant. If the concentration
of E
i
were to change by an infinitesimal amount a
corresponding change in F might be observed. The
Figure 11-1 Some control elements for metabolic reactions. Throughout the book modulation of the activity of an enzyme by allosteric
effectors or of transcription and translation of genes is indicated by dotted lines from the appropriate metabolite. The lines terminate in
a minus sign for inhibition or repression and in a plus sign for activation or derepression. Circles indicate direct effects on
enzymes, while boxes indicate repression or induction of enzyme synthesis.
Precursor
molecule, A A B C D E P
cAMP
second messenger
ATP
C
e
l
l

m
e
m
b
r
a
n
e
Binding
protein
Complex
Complex
Binding
protein
Activated
nucleotides
Transcription
Translation
Proteins
mRNA
+
+

Feedback
repression
Catabolism
Further
biosynthesis
(Product)
Isoenzymes
with differing
regulatory
properties
Enzymes as well
as structural and
regulatory proteins
affect every step
in metabolism.
Feedback inhibition
Precursor
activation
+

C' D' P'


EP (active)
P
i
H
2
O
Phosphatase
E (inactive)
+
ADP
ATP
Activated
amino acids
Steroid
hormone
Hormone
messenger
Hormone
receptor
E
1
E
2
E
3
S
0
S
1
S
2
S
3
S
n1
E
n1
E
n
S
n
537
(11-4)
ratio of the change in flux to the change in [E
i
] is the
sensitivity coefficient Z
i
:
The sensitivity coefficient can range from 1 for a pace-
maker enzyme to almost zero for a very active enzyme
that does not significantly limit the flux. The sum of
Z
1
to Z
n
is equal to 1:
Not only can the concentration [E
i
] change but also allo-
steric effectors can alter the activity. Kacser and Burns
defined this in terms of a controllability coefficient ,
the ratio of the logarithmic change in velocity to the
change in the concentration of an effector:
= d ln v/d ln [P]
It follows that the logarithmic change in flux d ln F can
be related to the change in effector concentration [P] in
the following way:
From this equation we see that for the flux to be sensi-
tively dependent upon [P], both Z
i
and must be rea-
sonably large. This is exactly the case for many of the
enzymes that have been identified as pacemakers.
This approach can be applied to many aspects of
metabolic control including cell growth.
20
Figure 11-2 Roles of phosphofructose kinase and fructose 1,6-bisphosphatase in the control of the breakdown (

) and
storage (

) of glycogen in muscle. The uptake of glucose from blood and its release from tissues is also illustrated. The
allosteric effector fructose 2,6-bisphosphate (Fru-2,6-P
2
) regulates both phosphofructokinase and fructose 2,6-bisphosphatase.
These enzymes are also regulated by AMP if it accumulates. The activity of phosphofructokinase-2 (which synthesizes
Fru-2,6-P
2
) is controlled by a cyclic AMP-dependent kinase and by dephosphorylation by a phosphatase.
O
CH
2
OPO
3
2
OH
OH
HO
2
O
3
POCH
2

O
OPO
3
2
CH
2
OH
OH
HO
2
O
3
POCH
2

Stored glycogen
+
Glucose 6-phosphate
Fructose 6-phosphate
ATP
ADP
-Fructose 2,6-
bisphosphate
Phosphofructokinase
AMP
Fructose 1,6-
bisphosphatase
H
2
O
P
i
+

Fructose 1,6-bisphosphate
From
lactate
and
amino acid
metabolism
Further
metabolism
ATP ADP
Glucose
Hexokinase
Glucose
6-phosphatase
P
i
H
2
O
Phosphofructokinase-2
a
d
e
b c

f
(11-2)
(11-3)
(11-5)
A. Pacemakers and the Control of Metabolic Flux
Z
dF F
d
d F
d
i
i i i
= =
[ ] [ ]
ln
ln[ ] E E E
Z
i
n
i =

=
1
1
d F Z d
i
ln ln [ ] = P
538 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
B. Genetic Control of Enzyme Synthesis
All cellular regulatory mechanisms depend upon
the synthesis of proteins, that is, upon the expression
of genetic information. Within a given cell many
genes are transcribed continuously but others may
remain unexpressed. The rates of both transcription
and degradation of mRNA are regulated, as are the
rates of synthesis of enzymes on the ribosomes in the
cytoplasm and the rates of protein turnover. Although
a single copy of a gene in each member of a pair of
chromosomes is often adequate, there are situations
in which extra copies of part of the DNA of a cell are
formed. Such gene amplification, which is dealt
with further in Chapter 27, provides the possibility for
very rapid synthesis of an enzyme or other protein. It
often happens in highly specialized cells such as those
of the silkworms silk glands which must make enor-
mous amounts of a small number of proteins.
21
1. One Gene or Many?
Many enzymes exist within a cell as two or more
isoenzymes, enzymes that catalyze the same chemical
reaction and have similar substrate specificities. They
are not isomers but are distinctly different proteins
which are usually encoded by different genes.
22,23
An
example is provided by aspartate aminotransferase
(Fig. 2-6) which occurs in eukaryotes as a pair of cyto-
solic and mitochondrial isoenzymes with different
amino acid sequences and different isoelectric points.
Although these isoenzymes share less than 50% se-
quence identity, their internal structures are nearly
identical.
24 27
The two isoenzymes, which also share
structural homology with that of E. coli,
28
may have
evolved separately in the cytosol and mitochondria,
respectively, from an ancient common precursor. The
differences between them are concentrated on the
external surface and may be important to as yet un-
known interactions with other protein molecules.
Isoenzymes are designated in various ways. They
are often numbered in the order of decreasing electrophoretic
mobility at pH 7 to 9: Most enzymes are negatively
charged in this pH range and the one that migrates
most rapidly toward the anode is numbered one. This
is the same convention used in the electrophoresis of
blood proteins (e.g. see Box 2-A).
Lactate dehydrogenase exists in the cytoplasm
of humans and most animals as five forms which are
easily separable by electrophoresis and are evenly
spaced on electropherograms.
8
This enzyme is a tetra-
mer made of two kinds of subunits. Isoenzyme 1, which
has the highest electrophoretic mobility, consists of
four identical type B subunits. The slowest moving
tetramer (isoenzyme 5) consists of four type A subunits,
while the other three forms, AB
3
, A
2
B
2
, and A
3
B, contain
both subunits in different proportions. The two sub-
units are encoded by separate genes which are expressed
to different extents in different tissues. Thus, heart
muscle and liver produce mainly subunit B, while
skeletal muscle produces principally subunit A. A
third subunit type (C) is found only in the testes.
29
Why do cells produce isoenzymes? One reason
may be that enzymes with differing kinetic parameters
are needed.
30
Substrate concentrations may vary greatly
between different tissues; between different subcellu-
lar compartments; and at different developmental
stages of an organism. While the need for various
isoenzymes of lactate dehydrogenase is not well un-
derstood,
31
it is easier to understand the roles of the
multiple forms of hexokinase, the enzyme that catalyzes
the reaction of step a in Fig. 11-2. The brain enzyme
has a high affinity for glucose (K
m
= 0.05 mM). Thus,
it is able to phosphorylate glucose and to make that
substrate available to the brain for metabolism, even
if the glucose concentration in the tissues falls to low
values.
31a
On the other hand, glucokinase, the hexo-
kinase isozyme found in liver, has a much higher K
m
of ~10 mM. It functions to remove excess glucose
from blood, whose normal glucose content is ~5.5 mM.
Glucokinase reaches its maximal activity only when
the glucose concentration becomes much higher.
32
This happens after a meal when the absorbed glucose
passes through the portal circulation directly to the
liver.
Another important source of variation in enzymes
as well as in other proteins is alternative splicing
of mRNA.
33
For example, transcription of the mouse
-amylase gene in the salivary gland starts at a different
site (promoter) than does transcription in the liver.
The two common isoforms of the insulin receptor (Fig.
11-11) arise because a 36-nucleotide (12-amino acid)
exon is spliced out of the mRNA for the shorter protein.
Isoenzymes of aldolase
34
and of many other proteins
are formed in a similar manner. Frame-shifting
during protein synthesis (Chapter 29) and also post-
translational alterations may give rise to additional
modified forms. They are often synthesized in rela-
tively small amounts but may be essential to the life
of the cell. In addition, genetic variants of almost any
protein will be found in any population. These often
differ in sequence by a single amino acid.
2. Repression, Induction, and Turnover
The synthesis of some enzymes is referred to as
constitutive, implying that the enzyme is formed no
matter what the environmental conditions of the cell.
For example, many bacteria synthesize the enzymes
required to catabolize glucose under all conditions of
growth. Other enzymes, known as inducible, are
often produced only in small amounts. However, if
539
cells are grown in specific inducing conditions for
these enzymes, they are synthesized in larger quanti-
ties. For example, when E. coli is cultivated in the
presence of lactose, several of the enzymes required
for the catabolism of that disaccharide are formed.
Synthesis of these enzymes is normally repressed.
The genes which code for them are kept turned off
through the action of protein repressors which bind
to specific sites on the DNA and block transcription of
the genes that they control (Fig. 11-1). Repressors have
allosteric properties; in one conformation they bind
tightly to DNA but in another they do not bind. For
example, the free tryptophan repressor binds to DNA
only weakly, but if a high concentration of tryptophan
develops within the cell the tryptophan binds to an
allosteric site on the repressor protein (Fig. 5-35). This
changes the conformation to one that binds tightly to
the appropriate control sequence in the DNA. In the
case of lactose catabolism the free repressor binds to
control sequences in the DNA until the inducer, allo-
lactose (Chapters 4 and 28), binds at an allosteric site.
This decreases the affinity of the repressor for DNA
and the controlled genes are derepressed. There are
also many protein transcription factors that have a
positive effect, binding to DNA and promoting tran-
scription of specific genes.
Synthesis of many enzymes is repressed most of
the time. The appearance of an enzyme at a particular
stage in the life of an organism as well as the differing
distributions of isoenzymes within differentiated tissue
result from derepression. The control of enzyme syn-
thesis may also be exerted during the splicing of tran-
scripts and at the translational level as well. These
control mechanisms are often relatively slow, with
response times of hours or even days. However,
effects on the synthesis of some hormones, such as
insulin (Section G), may be observed within a few
minutes.
Genetic factors influence the rate of not only
synthesis of proteins but also their breakdown, i.e.,
the rate of turnover. As we have seen in Chapter 10,
some enzymes are synthesized as inactive proenzymes
which are later modified to active forms, and active
enzymes are destroyed, both by accident and via
deliberate hydrolytic pathways. Protein antienzymes
may not only inhibit enzymes but may promote their
breakdown.
35
An example is the antienzyme that
controls ornithine decarboxylase, a key enzyme in
the synthesis of the polyamines that are essential to
growth.
36,37
As with all cell constituents, the synthesis of
enzymes and other proteins is balanced by degradation.
3. Differences among Species
Catalytic mechanisms of enzymes have usually
been conserved throughout evolution, and certain
residues in an enzyme may be invariant among many
species. However, there are usually many differences
in the distribution of amino acid residues on the
surface of the proteins. Since changes in the surface
shape of a protein molecule may alter the sensitivity to
a potential allosteric effector, very different regulatory
properties are sometimes found between species.
C. Regulation of the Activity of Enzymes
Some regulation of metabolism is provided by the
kinetic properties of the enzymes. Thus, the value
(mol/l) of the Michaelis constant K
m
for an enzyme is
usually low if the substrate normally occurs in very
low concentration. It is likely to be higher if the enzyme
acts on an abundant substrate. A tightly bound product
of an enzymatic reaction will be released slowly from
an enzyme if the product concentration within the cell
is too high. However, while some regulation of meta-
bolism is provided in such simple ways, the rapid
changes that result from stimulation by hormones or
by nerve impulses depend upon additional specific
regulatory mechanisms that are discussed in the fol-
lowing sections.
1. Allosteric Control
Probably the most common and widespread control
mechanisms in cells are allosteric inhibition and
allosteric activation. These mechanisms are incorpo-
rated into metabolic pathways in many ways, the most
frequent being feedback inhibition. This occurs when
an end product of a metabolic sequence accumulates
and turns off one or more enzymes needed for its own
formation. It is often the first enzyme unique to the specific
biosynthetic pathway for the product that is inhibited. When
a cell makes two or more isoenzymes, only one of
them may be inhibited by a particular product. For
example, in Fig. 11-1 product P inhibits just one of the
two isoenzymes that catalyzes conversion of A to B;
the other is controlled by an enzyme modification
reaction. In bacteria such as E. coli, three isoenzymes,
which are labeled I, II, and III in Fig. 11-3, convert
aspartate to -aspartyl phosphate, the precursor to the
end products threonine, isoleucine, methionine, and
lysine. Each product inhibits only one of the isoenzymes
as shown in the figure.
Feedback can also be positive. Since AMP is a
product of the hydrolysis of ATP, its accumulation is a
signal to activate key enzymes in metabolic pathways
that generate ATP. For example, AMP causes allosteric
activation of glycogen phosphorylase, which catalyzes
the first step in the catabolism of glycogen. As is shown
in Fig. 11-5, the allosteric site for AMP or IMP binding
is more than 3 nm away from the active site. Only a
C. Regulation of the Activity of Enzymes
540 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
subtle conformational change accom-
panies binding of IMP.
38
However,
31
P
NMR studies indicate that binding of
the activator may induce a change in the
state of protonation of the phosphate
group of the coenzyme pyridoxal phos-
phate at the active site as explained in
Chapter 12.
A second pattern of allosteric con-
trol may be referred to as precursor
activation or feed-forward control.
A metabolite acting as an allosteric
effector turns on an enzyme that either
acts directly on that metabolite or acts
on a product that lies further ahead in
the sequence. For example, in Fig. 11-1,
metabolite C activates the enzyme that
catalyzes an essentially irreversible
reaction of compound D. An actual
example is provided by glycogen synthase,
whose inactive dependent or D form
is activated allosterically by the glycogen
precursor glucose 6-phosphate.
39
See
also phosphorylase kinase (Section 2).
Regulatory subunits. Some
enzymes consist not only of catalytic
subunits, which contain the active sites,
but also of regulatory subunits. The
latter bind to the catalytic subunits and
serve as allosteric modifiers. The bind-
ing of inhibitors or activators to specific
sites on the regulatory subunits induces
conformational changes in these subunits,
altering their interaction with the cata-
lytic subunits. A well-known example is
aspartate carbamoyltransferase (ACTase)
from E. coli (Fig. 7-20).
40 42
Its regulatory
subunits carry binding sites for cytidine
triphosphate (CTP), which acts as a
specific allosteric inhibitor of the en-
zyme. The significance of this fact is
that ACTase catalyzes the first reaction
specific to the pathway of synthesis of
pyrimidine nucleotides (Chapter 25).
CTP is an end product of that pathway
and exerts feedback inhibition on the
enzyme that initiates its synthesis. ATP
also binds to the regulatory subunits
and causes inhibition of the enzyme.
Figure 11-3 Feedback inhibition of enzymes involved in the biosynthesis of threonine, isoleucine, methionine, and lysine in
E. coli. These amino acids all arise from L-aspartate, which is formed from oxaloacetate generated by the biosynthetic reactions
of the citric acid cycle (Fig. 10-6). Allosteric inhibition. Repression of transcription of the enzyme or of its synthesis on
ribosomes.
L-Homoserine
H NH
3
+
CH
2
COO

HO
L-Aspartate
Aspartate semialdehyde
I II III
NADH + H
+
NAD
+
+ P
i
2-Oxobutyrate L-Isoleucine
L-Methionine
L-Lysine
HOOC
COO

H NH
3
+
H NH
3
+
C
COO

H
O
L-Threonine
H HO
H NH
3
+
H
3
C
COO

H
3
C
CH
2
COO

O
Aspartyl phosphate
H NH
3
+
C
COO

O
O

HO
3
P
H NH
3
+
OH
2
C
COO

HO
3
P
541
In the presence of CTP the binding of the substrates is
cooperative, as would be anticipated if there is a two-
state conformational equilibrium involving the catalytic
subunits. This would be similar to the case depicted
in Fig. 9-13 except that trimers rather than dimers are
involved and the inhibitor is a part of the regulatory
subunit and is controlled by binding of CTP.
Binding of ATP to ACTase decreases the coopera-
tivity in substrate binding, again as predicted for a two-
state model. However, as suggested by its structure,
this enzyme system is more complex, as also indicated
by the observed anticooperative binding of the activator
CTP. X-ray studies show that binding of ligands causes
a movement along subunit interfaces as well as local-
ized conformational changes within the subunits. These
are reminiscent of the changes seen upon binding of
oxygen to hemoglobin (Fig. 7-25).
Glycolysis and gluconeogenesis. The highly
regulated enzymes phosphofructokinase and fructose
1,6-bisphosphatase catalyze steps b and c of the reactions
in Fig. 11-2, reactions that control glucose metabolism
in cells. These enzymes have been studied for many
years but the important allosteric effector -fructose
2,6-bisphosphate was not discovered until 1980.
4346
This compound, which is formed from fructose 6-phos-
phate by a new enzyme, phosphofructokinase-2 (also
called fructose 6-phosphate 2-kinase) in step d, Fig. 11-2
activates phosphofructokinase allosterically. At the
same time it inhibits fructose 1,6-bisphosphatase, an
enzyme required for reversal of glycogen breakdown,
that is, for the conversion of various metabolites arising
from amino acids into glycogen (Fig. 11-2, step c).
46a
These same two key regulated enzymes are also affected
by many other metabolites. For example, ATP in excess
inhibits phosphofructokinase, decreasing the overall
rate of glucose metabolism and consequently of ATP
production. Citrate, which is exported from mito-
chondria when carbohydrate metabolism is excessive,
inhibits the same enzyme. On the other hand, AMP
acts together with fructose 2,6-bisphosphate to activate
the pathway for glycogen breakdown and to inhibit
that for its synthesis (Fig. 11-2). The concentration of
the regulator fructose 2,6-bisphosphate is controlled
by mechanisms that are discussed in the following
section.
O
OH
HO
OPO
3
2
CH
2
OH
2
O
3
POCH
2
-Fructose
2,6-bisphosphate
2. Covalent Modification by Phosphorylation
and Dephosphorylation
Rapid alteration of the activities of enzymes is often
accomplished by reversible covalent modification.
39,47
Many different modification reactions are known
(Table 11-1) and there are doubtless many more to be
discovered. Probably the most widespread and certainly
the most studied is phosphorylation, the transfer of a
phospho group from ATP or other suitable donor to a
side chain group on the enzyme. An example is the
phosphorylation by ATP of hydroxyl groups of specific
serine residues in the two enzymes glycogen phospho-
rylase and glycogen synthase. These modifications are
accomplished through a series or cascade of reactions
initiated by the binding of hormones to cell surface
receptors or by nerve impulses as is shown in Fig. 11-4.
In the absence of such a stimulus, glycogen phospho-
rylase is present in its unphosphorylated or b form.
Although this form can be activated allosterically by
AMP, it is normally nearly inactive. When an appro-
priate hormone binds to the cell surface a cascade of
reactions, as described in Section D, leads to activation
of an enzyme called phosphorylase kinase.
47a
This
enzyme transfers a phospho group from ATP to the
OH group of the side chain of Ser 14 in each subunit
(Fig. 11-4, left center), converting the enzyme into the
active glycogen-degrading phosphorylase a. This
switches the cellular metabolism from that designed
to deposit the storage polysaccharide glycogen to one
that degrades glycogen to provide the cell with energy.
Serine 14 of glycogen phosphorylase is located adjacent
to the allosteric AMP binding site (Fig. 11-5) and is
surrounded by positively charged arginine and lysine
side chains. Phosphorylation induces a rearrangement
of hydrogen bonds involving these residues and in
some way sends an appropriate signal to the active site.
It also increases the affinity for AMP in the allosteric
sites.
48,48a
Phosphorylase kinase is allosterically acti-
vated by AMP, a product of its action a feed-forward
activator.
The control of glycogen phosphorylase by the
phosphorylationdephosphorylation cycle was dis-
covered in 1955 by Edmond Fischer and Edwin Krebs
50
and was at first regarded as peculiar to glycogen break-
down. However, it is now abundantly clear that similar
reactions control most aspects of metabolism.
51
Phos-
phorylation of proteins is involved in control of carbo-
hydrate, lipid, and amino acid metabolism; in control
of muscular contraction, regulation of photosynthesis
in plants,
52
transcription of genes,
51
protein syntheses,
53
and cell division; and in mediating most effects of
hormones.
Protein kinases and cyclic AMP. Phosphorylase
kinase is one of hundreds of different protein kinases
which differ in specificity toward their substrates, in
C. Regulation of the Activity of Enzymes
542 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Figure 11-4 Cascades of phosphorylation and dephosphorylation reactions involved in the control of the metabolism of
glycogen. Heavy arrows (

) show pathways by which glucosyl units of glycogen are converted into free glucose or enter
the glycolytic pathway. Green arrows (

) trace the corresponding biosynthetic pathways. Gray arrows (

) trace the
pathway of activation of glycogen phosphorylase by a hormone such as adrenaline (epinephrine) or glucagon and by the
action of protein kinases. A few of the related pathways in the network of reactions that affect glycogen metabolism are also
shown. This includes protein phosphatases, which remove phospho groups from proteins and allow cells to relax to the state
that preceded activation. One of these (protein phosphatase 1) is activated by phosphorylation by an insulin-stimulated
protein kinase.
49
However, the significance is uncertain; control of glycogen synthase is complex.
49a
Glucagon
Adrenaline
(epinephrine)
PI
Phosphatidyl
inositol
Vasopressin
Angiotensinogen II
Glucagon
receptor
ATP cAMP
+ +
Inositol trisphosphate (Ins-P
3
)
+ +
adrenergic
receptor
adrenergic
receptor
Nerve
impulse
+
+
Ca
2+
,
sequestered
Free Ca
2+
Inactive
protein kinase A
R
2
C
2
(R-cAMP)
2
2C (Active protein kinase A)
Active
phosphorylase
kinase
Inactive
phosphorylase
kinase
Glycogen
phosphorylase b
(inactive)
Glycogen
phosphorylase a
(active)
Inactive
glycogen synthase
Active
glycogen synthase
ATP
ATP ADP
ADP
P
i
P
i
H
2
O
H
2
O
Glycogen
(n residues
of glucose)
Glycogen
(n-1 residues
of glucose)
P
i UDP
UDP-Glc
Glc-1-P
Glc-1-P
Glc-6-P
Glycolytic pathway
Glucose
PP
i
UTP
H
2
O
2P
i
P
i
H
2
O
Active protein
phosphatase 1
Active protein
phosphatase -2
Membrane
+
Adrenaline
Many
kinases
Glycogen
synthesis
Glycogen
breakdown
543
Reaction Example Location of discussion
A. Phosphorylationdephosphorylation
Phosphorylation of Ser, Thr Glycogen phosphorylase This section
Phosphorylation of Tyr Insulin receptor Section G
Adenylylation, Uridylylation Glutamine synthetase Chapter 25
ADP-ribosylation This section
B. Methylation of carboxyl groups Bacterial glutamyl Section D,5
Aspartyl Box 12-A
Protein phosphatase 2A Section C,3
Ras Section D,3
C. Formation of carbamino groups
In hemoglobin Eq. 7-23
In ribulose bisphosphate carboxylase Chapters 13,23
D. Acylation
Acetylation Histones Chapter 27
Palmitoylation Ras Section D,3
Prenylation Ras Chapter 22
E. Disulfide formation and cleavage Chapters 10,15
Section C,4
TABLE 11-1
A Few Covalent Modification Reactions Utilized to Control Metabolism
Figure 11-5 Schematic diagram of the
glycogen phosphorylase dimer. The
view is down the twofold axis of the
dimer with the allosteric and Ser-P
sites toward the viewer. Access to the
catalytic site is from the far side of the
molecule. The diagram shows the
major change in conformation of the
amino-terminal residues on phospho-
rylation. Residues 10 23 of glycogen
phosphorylase b are shown as a thick
solid line. These residues are not well
ordered and make intrasubunit con-
tacts. Upon phosphorylation, residues
10 23 change their conformation and
are shown dark crosshatched with the
position of Ser14-P indicated at the
intersubunit interface. The fold of
residues 24 80 through the 1 helix,
the cap, and the 2 helix is shown
lightly cross-hatched. The AMP allo-
steric effector site is located between
the 2 helix and the cap region of the
other subunit. The glycogen storage
site is located on the surface of the
subunit and is associated with a long
helix. The catalytic site is at the
center of the subunit where the two
domains come together. Courtesy of
Louise N. Johnson.
48
C. Regulation of the Activity of Enzymes
Catalytic site
Allosteric
effector site
Cap
Allosteric
effector site
Catalytic site
Glycogen
storage site
Glycogen
storage site
544 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
the functional groups phosphorylated, and in their
allosteric activators.
39,51,54 56
There may be more than
1000 different protein kinase genes in vertebrate animals,
accounting for ~2% of the genome.
57
Many cytosolic
protein kinases transfer a phospho group from ATP to
either a serine or threonine side chain at a bend or
other surface feature of the substrate protein. Some
sites of phosphorylation in the substrate proteins
contain lysine or arginine residues, separated from
the serine or threonine by only one residue but many
other sequences may also surround phosphorylation
sites.
51,58
In the case of the 750-residue glycogen
synthase, seven serine residues in different parts of
the chain are phosphorylated by the action of at least
five different kinases.
49,49a,59,60
The various kinases
phosphorylate groups at different sites and their effects
are roughly additive.
Some of the best known protein kinases (designated
PKA or cAPK) are those that depend upon 3', 5'-cyclic
AMP (cAMP) as an allosteric effector. They are oligo-
meric proteins of composition R
2
C
2
, where R is a regula-
tory subunit and C is a catalytic subunit. Unless
cAMP is present, the regulatory subunits interact with
the catalytic subunits, keeping them in an inactive
inhibited form. However, when cAMP is present it
binds to the regulatory subunit dimer, releasing the
two active catalytic units (Eq. 11-6). This reaction is
R
2
C
2
+ 4 cAMP R
2
(cAMP)
4
+ 2C
reversible and as the concentration of cAMP is reduced
by hydrolysis (see Eq. 11-8) the regulatory units recom-
bine with the catalytic subunits and again inhibit them.
There are two prominent types of mammalian
cAMP-dependent protein kinases.
51,61
The catalytic
subunit is identical for both; the 41-kDa peptide as
isolated from beef heart has 350 residues and an N
terminus blocked by a myristoyl (tetradecanoyl) group.
62
One phosphoserine and one phosphothreonine are also
present.
51
The ~50-kDa regulatory subunits vary in
size and may also be subject to additional regulation by
phosphorylation.
63
Three-dimensional structures are
known for both the catalytic
62,64,65
and the regulatory
66
subunits. A cyclic GMP (cGMP)-activated protein
kinase is present in some mammalian tissues
67,68
and is
widespread in invertebrates.
51,67
The cyclic nucleotide-
binding domains of these kinases have structures
similar to that of the E. coli catabolite activator protein
(Fig. 28-6)
51,68
and the catalytic domains are structurally
similar to those of many other kinases.
Among the kinases that phosphorylate glycogen
synthase is a casein kinase, named for the fact that
the milk protein casein is also a good substrate. A
family of casein kinases are found in the cytoplasm and
nuclei of all eukaryotic cells. They phosphorylate serine
and threonine side chains but have structures distinct
from those of the cAMP-dependent kinases.
69 72
Casein
kinase-2 (CK2) phosphorylates many proteins, including
several that function in gene replication, transcription,
and cell growth and division.
72a
Theileriosis, a para-
sitic disease of cattle in Africa, is caused by the tick-
borne protist Theileria parva. The condition is often
fatal as a result of a leukemia-like condition resulting
from overexpression of the casein kinase-2 gene.
72
Phosphorylase kinase is one of a large group of
specialized protein kinases, each of which acts on a
small number of proteins. It is regulated both by
covalent modification and allosterically by calcium
ions.
73 78
It contains four different kinds of subunits
ranging in size from 17 kDa to about 145 kDa and
has the composition ()
4
. Phosphorylation of one
serine on each of the 145-kDa and 120-kDa regula-
tory subunits is catalyzed by a cAMP-dependent
protein kinase. The subunit is the Ca
2+
-binding
protein calmodulin (Fig. 6-8) and serves as a regula-
tory subunit sensitive to Ca
2+
. The 45-kDa subunit
contains the catalytic domain as well as a calmodulin-
binding domain.
79
Other Ca
2+
-dependent protein
kinases include the protein kinase C family, which
is discussed further in Section E, Ca
2+
/calmodulin-
dependent protein kinases,
80,81
and a plant kinase
with a regulatory domain similiar to calmodulin.
82
Protein tyrosine kinases (PTKs) place phospho
groups on the phenolic oxygen atoms of tyrosyl residues
of some proteins.
83 85
The resulting phosphotyrosine
accounts for only about 1/3000 of the phospho groups
in proteins but has aroused interest for two reasons.
First, binding of growth hormones, such as epidermal
growth factor (EGF), platelet-derived growth factor
(PDGF), and insulin, to their receptors stimulates
tyrosine-specific protein kinase activity of the receptor
proteins (see Figs. 11-11 to 11-13).
69,83,86
Second, a
number of cancer-causing oncogenes encode similar
kinases (Section H). Tyrosine protein kinases are essen-
tial components of the cell division cycle (see Fig. 11-15).
Protein phosphatases. Most regulatory alter-
ations in enzymatic activity are spontaneously revers-
ible. The concentration of the allosteric effector soon
drops and the covalent modifications are reversed so
that the system relaxes to a state approximating the
N
N
N
N
NH
2
O CH
2
O
P
O
O
O
OH
3',5'-cyclic AMP
(11-6)
545
original one. Among the enzymes required for this
reversal are the protein phosphatases that remove the
phospho groups placed on amino acid side chains by
kinases.
54,86 91
For example, four different phosphatases
act on the (inactive) phosphorylated glycogen synthase
and on the phosphorylated forms of glycogen phos-
phorylase and phosphorylase kinase. At least one of
these (a protein phosphatase 1; see Fig. 11-4) is regulated
by phosphorylation that is stimulated by insulin.
49,49a
The matter is made yet more complex by the presence
in tissues of small protein inhibitors which can prevent
the action of these phosphatases.
88,92 94
Another natu-
rally occurring inhibitor is the polyether fatty acid
okadaic acid, a shellfish poison, actually produced
by dinoflagellates but which accumulates in sponges
and in mussels and other bivalves.
94
See also Chapter 12,
D,4.
Most protein phosphatases are specific toward
phosphoserine and phosphothreonine
89,95
or toward
phosphotyrosine residues.
90,96 98
Some have a dual
specificity.
99
There are Ca
2+
-activated phosphatases
100
and phosphatases with transmembrane segments
connected to receptors on the external cell surface.
Some phosphorylationdephosphorylation regulatory
systems utilize bifunctional enzymes consisting of a
protein kinase fused to a protein phosphatase. For
example, the same E. coli enzyme that phosphorylates
isocitrate dehydrogenase also hydrolyzes off the phos-
pho group that it has put on.
101
Another bifunctional
kinasephosphatase catalyzes both the formation
(reaction d of Fig. 11-2) and breakdown (reaction e) of
fructose 2,6bisphosphate in eukaryotic tissues. This
kinasephosphatase enzyme is, in turn, controlled by
phosphorylation by a cAMP-dependent protein kinase.
In the liver, this latter phosphorylation strongly inhibits
the kinase activity preventing buildup of fructose
2,6-bisphosphate, the allosteric activator of phospho-
fructokinase-1, which catalyzes reaction b of Fig. 11-2.
Consequently, when a pulse of cAMP is generated within
a hepatocyte, glycolysis is blocked but glycogenolysis,
the breakdown of glycogen by phosphorylase, is stim-
ulated. The glucose 6-phosphate that is formed is
hydrolyzed by glucose-6-phosphatase releasing glucose
into the bloodstream. On the other hand, in heart and
probably in most other tissues, a different isoenzyme
form of the bifunctional enzyme is present. Phospho-
rylation by cAMP activates the kinase and inhibits the
phosphatase. Consequently, in these tissues cAMP
induces the activation of glycogenolysis and glycolysis
coordinately.
102 105
In brain a different allosteric activator,
ribose 1, 5-bisphosphate, may regulate phospho-
fructokinase-1.
106
Phosphorylation in bacteria. A bacterial enzyme
whose activity is controlled by phosphorylation is
isocitrate dehydrogenase. Transfer of a phospho
group to the OH of Ser 113 completely inactivates the
E. coli enzyme,
107,108
causing isocitrate to build up in
the citric acid cycle (Fig. 10-6) and to be diverted into
the glyoxylate pathway, which is depicted in Fig. 17-16.
In this instance, it appears likely that the negative
charge of the added phospho group causes electrostatic
repulsion of the substrate isocitrate. In agreement
with this concept, mutation of Ser 113 of this enzyme
to Asp (mutant S113 D) also inactivates the enzyme.
109
In addition to kinasephosphatase cycles, bacteria
use at least two other ATP-dependent regulatory mech-
anisms.
110
In the sensor kinase/response regulator
(or two-component) systems
110- 111
a sensor protein,
upon being allosterically activated, phosphorylates
itself (autophosphorylation) on a specific histidyl
residue to form an N-phosphohistidine derivative.
The phosphohistidine then transfers its phospho
group onto a specific aspartyl group in the response
regulator causing the regulator to bind to its target
protein and to exert its regulatory effect. The best
known example involves the control of the motion of
bacterial flagella which is discussed briefly in Section
D,5 and further in Chapter 19.
The third bacterial regulatory device is the phospho-
enolpyruvate:sugar phosphotransferase system (Eq. 8-4).
It is involved not only in transport but also in control-
ling a variety of physiological processes.
110,112,113
3. Other Modification Reactions Involving
Group Transfer
Nucleotidylation, the transfer of an entire nucleo-
tidyl unit, rather than just a phospho group, to a protein
is sometimes utilized for regulation. For example, gluta-
mine synthetase of E. coli is modified by adenylylation,
transfer of an adenylyl group from ATP to a tyrosine
side chain.
114 116
Relaxation to the unmodified enzyme
is catalyzed by a deadenylylating enzyme. A regulatory
protein that undergoes reversible uridylylation
117
also
functions in this system (see Fig. 24-7). Several mito-
chondrial and cytoplasmic proteins are modified by
attachment of ADP-ribosyl groups to specific guanidino
groups of arginine and other side chain groups.
114,118 123
This ADPribosylation requires the coenzyme NAD
+
as the ADP-ribosyl donor and is also catalyzed by
bacterial toxins such as cholera toxin (Box 11-A) and
by diphtheria toxin (Box 29-A). A second kind of ADP
ribosylation occurs within nuclei where the enzyme
poly(ADPribose) synthase catalyzes both an initial
ADPribosylation of an amino acid side chain group
and also addition of more ADP-ribosyl units to form
a polymer
124
(see Chapter 27).
Like inorganic phosphate, inorganic sulfate can be
converted into an activated form in which the sulfate
resembles the terminal phospho group of ATP (Eq. 17-38).
The resulting activated sulfo group can be transferred
to other compounds including enzymes. Formation of
C. Regulation of the Activity of Enzymes
546 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
BOX 11-A CHOLERA TOXIN AND OTHER DANGEROUS PROTEINS
As recently as 1980 it was estimated that there
were ~ 100 million cases of acute diarrhea in Asia,
Africa, and Latin America;
a
in 1991 there were four
million deaths among children under five years of
age.
b
The causative agents are bacteria and one of
the most dangerous is Vibrio cholerae, which multiplies
in the small intestine and secretes an exotoxin.
Cholera toxin causes such a rapid loss of fluid and
salts from the body that death occurs very quickly,
even in adults. There is little cellular damage and
almost all deaths can be prevented by intravenous
administration of water, salts, and the antibiotic
tetracycline. Fluids can also be given orally if
glucose, which promotes intestinal absorption, is
included with the Na
+
, K
+
, Cl

, and HCO
3

salts.
b
Cholera toxin consists of one 240-residue A sub-
unit and a ring of five 103-residue B subunits.
ce
The
latter is a targeting complex, a bacterial lectin each
subunit of which binds to the galactose and sialic acid
termini of a single molecule of ganglioside G
M1
.
Gal13Gal14Gal14Glc1 Ceramide
3

NeuNAc2
Ganglioside G
M1
(See also Fig. 20-11)
The nonpolar ceramide chains of the ganglioside
are buried in the membrane bilayer. The initial tight
binding (K
f
~ 2 x 10
7
M
1
) of the toxin is followed by
a poorly understood process in which the A subunit
enters the cell. The peptide chain of this subunit is
cut by bacterial proteases to give A
1
and A
2
fragments
joined by an S S bridge. Within the victims cells
the disulfide is reduced to generate the active toxin,
the 192- to 194- residue A
1
fragment.
f,g
This toxin subunit is an enzyme, an ADP-ribo-
syltransferase which catalyzes transfer of ADP-
ribosyl units from the coenzyme NAD
+
to specific
arginine side chains to form N-ADP-ribosyl deriva-
tives of various proteins. Of the proteins modified
by cholera toxin, the most significant is the guanyl
nucleotide regulatory protein G
s
of the adenylate
cyclase system.
c,f,h
ADP ribosylation of arginine
201 of the subunit of protein G
s
inhibits the GTP
hydrolysis that normally allows the protein to relax
to an unactivated form.
e
The ADP-ribosylated G
s
keeps adenylate cyclase activated continuously and
The three-dimensional structure of cholera toxin.
e
Side
view of the subunit pentamer as a ribbon drawing.
Bound noncovalently to it are five molecules of the gan-
glioside G
M1
(compare with the structure in Fig. 7-5). The
diacyl glycerol parts of the gangliosides are buried in the
membrane that lies below the toxin molecule. Courtesy
of W. G. J. Hol.
Arginine side chain
Nicotinamide
NAD
+
Cholera toxin
H
2
N N Protein
NH
2
H
+
N
N
N
N
O
NH
2
H
2
C
O
P
O
P
O
CH
2
O O

O
O
NH
N
Protein
H
OH OH
OH HO
2
+
N
H
547
BOX 11-A (continued)
(A) Stereoscopic view of the G
M1
pentasaccharide binding site,
showing both direct and solvent-
mediated hydrogen bonding inter-
actions between sugar and protein
residues. The viewpoint is from
underneath the membrane bind-
ing surface of the B pentamer.
Starred residues are from an adja-
cent monomer. The terminal galac-
tose residue of the pentasaccharide
(upper right) is most deeply inserted
into the binding site and is involved
in the greatest number of identifiable
binding interactions. The sialic acid
residue, near the bottom of the figure,
is also involved in several hydro-
gen-bonding interactions. Hydro-
phobic interactions include the
approach of the sialic acid acetyl
group to the edge of the Tyr 12
phenyl ring and the positioning
of the terminal galactose sugar ring
parallel to the indole ring of Trp 88.
Figures generated using program
MolScript (Kraulis, 1991). From
Merritt et al.
e
(B) Side view of the
intact AB
5
toxin as an -carbon
trace. The nicked A chain is at the
top. The nick can be seen in the
upper right corner, as can a disulfide
bridge connecting the A
1
and A
2
segments. A single helix extends
into the pore in the center of the B
pentamer. The side chains of Trp 88
of the B subunits have been added
to mark the ganglioside binding
sites. These side chains are also
seen in (A). (C) View of the AB
5
molecule from the bottom showing
the helix from the A
2
fragment
surrounded by a tight cage of five
long helices from the B subunits.
Residues 237240 of the A
2
frag-
ment have the KDEL sequence and
may extend from the pore to contact
the membrane. (B) and (C) courtesy
of Edwin M. Westbrook. See Zhang
et al.
c
generating abnormally high concentrations of cAMP.
It is the effect of the cAMP on proteins of the intestinal
mucosal cell membranes that causes the disastrous
excessive secretion of water and salts that are charac-
teristic of cholera.
Most strains of V. cholerae are relatively harm-
less but they may suddenly be transformed into a
virulent toxin-producing form by infection with a
bacterial virus similar to M13 (Chapter 5), which
carries the toxin gene. Entrance into the Vibrio cell
occurs with the help of pili which are present in
many strains.
i,j
Many other bacterial toxins have AB
n
structures
similar to that of cholera toxin. For example, toxin-
producing strains of E. coli are also important causes
of diarrhea in humans and in domestic animals.
km
C. Regulation of the Activity of Enzymes
Glu 11
Trp 88
Tyr 12
Asn 14
Gly 33*
Glu 51
Gln 61
Lys 91
Asn 90
His 13
His 57
Gln 56
Glu 11
Trp 88
Tyr 12
Asn 14
Gly 33*
Lys 91
His 13
Glu 51
Gln 61
Asn 90
His 57
Gln 56
A
B
C
548 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
a
Holmgren, J. (1981) Nature (London) 292, 413417
b
Hirschhorn, N., and Greenough, W. B., III (1991) Sci. Am.
264(May), 5056
c
Zhang, R. -G., Scott, D. L., Westbrook, M. L., Nance, S.,
Spangler, B. D., Shipley, G. G., and Westbrook, E. M. (1995) J.
Mol. Biol. 251, 563573
d
Zhang, R. -G., Westbrook, M. L., Westbrook, E. M., Scott, D. L.,
Otwinowski, Z., Maulik, P. R., Reed, R. A., and Shipley, G. G.
(1995) J. Mol. Biol. 251, 550562
e
Merritt, E. A., Sarfaty, S., van den Akker, F., LHoir, C., Martial,
J. A., and Hol, W. G. J. (1994) Protein Sci. 3, 166175
f
Moss, J., Stanley, S. J., Morin, J. E., and Dixon, J. E. (1980) J. Biol.
Chem. 255, 1108511087
g
Janicot, M., Fouque, F., and Desbuquois, B. (1991) J. Biol. Chem.
266, 1285812865
h
Tsai, S.-C., Adamik, R., Moss, J., and Vaughan, M. (1991)
Biochemistry 30, 36973703
i
Williams, N. (1996) Science 272, 1869 1870
j
Waldor, M. K., and Mekalanos, J. J. (1996) Science 272, 1910
1914
k
Moss, J., and Vaugham, M., eds. (1990) ADP Ribosylating Toxins
and G Proteins: Insights into Signal Transduction, American
Society for Microbiology, Washington, DC
l
Sato, T., Ozaki, H., Hata, Y., Kitagawa, Y., Katsube, Y., and
Shimonishi, Y. (1994) Biochemistry 33, 86418650
m
van den Akker, F., Merritt, E. A., Pizza, M. G., Domenighini,
M., Rappuoli, R., and Hol, W. G. J. (1995) Biochemistry 34,
1099611004
n
Sixma, T. K., Pronk, S. E., Kalk, K. H., van Zanten, B. A. M.,
Berghuis, A. M., and Hol, W. G. J. (1992) Nature (London) 355,
561564
o
Sixma, T. K., Kalk, K. H., van Zanten, B. A. M., Dauter, Z.,
Kingma, J., Witholt, B., and Hol, W. G. J. (1993) J. Mol. Biol. 230,
890918
p
Antoine, R., and Locht, C. (1994) J. Biol. Chem. 269, 64506457
q
Goodemote, K. A., Mattie, M. E., Berger, A., and Spiegel, S.
(1995) J. Biol. Chem. 270, 1027010277
r
Ohtsuka, T., Nagata, K. -i, Iiri, T., Nozawa, Y., Ueno, K., Ui, M.,
and Katada, T. (1989) J. Biol. Chem. 264, 1500015005
s
Arora, N., Klimpel, K. R., Singh, Y., and Leppla, S. H. (1992) J.
Biol. Chem. 267, 15542 15548
t
Benz, R., Maier, E., Ladant, D., Ullmann, A., and Sebo, P. (1994)
J. Biol. Chem. 269, 2723127239
u
Heveker, N., Bonnaff, D., and Ullmann, A. (1994) J. Biol. Chem.
269, 32844 32847
v
Otero, A. S., Yi, X. B., Gray, M. C., Szabo, G., and Hewlett, E. L.
(1995) J. Biol. Chem. 270, 96959697
w
Hackett, M., Guo, L., Shabanowitz, J., Hunt, D. F., and Hewlett,
E. L. (1994) Science 266, 433435
x
Ozaki, H., Sato, T., Kubota, H., Hata, Y., Katsube, Y., and
Shimonishi, Y. (1991) J. Biol. Chem. 266, 59345941
y
Moss, J., and Vaughan, M. (1995) J. Biol. Chem. 270, 1232712330
The heat-labile E. coli enterotoxin, whose gene is
carried on a plasmid, is a close relative of cholera
toxin
n,o
and also catalyzes ADP ribosylation of argi-
nine 201 of the G
s
subunit.
m
Bordetella pertussis,
which causes whooping cough, forms a similar
toxin that attacks the inhibitory regulatory protein
G
i
p,q
as well as transducin and inactivates them by
ADP ribosylation. Diphtheria toxin (Box 29-A),
the exotoxin from Pseudomonas aeruginosa, and
the toxin from Clostridium botulinum also catalyze
ADP-ribosylation reactions.
k,r
Some bacteria produce effects similar to those
of cholera toxin in different ways. For example,
among a variety of toxic proteins produced by
Bacillus anthracis, the causative agent of anthrax,
is an adenylate cyclase that is able to enter the
hosts cells.
s
Similarly, B. pertussis, in addition to
its ADP-ribosylating toxin, produces a calmodulin-
stimulated adenylate cyclase which, when taken up
by phagocytic cells, disrupts their function in the
bodys defense system.
t w
In addition to the heat-
labile enterotoxin, some strains of E. coli produce
heat-stable toxins, small 18-residue peptides related
in structure to the intestinal peptide guanylin.
l,x
These peptides bind to a membrane-bound guanylate
cyclase activating fluid secretion into the intestine.
Why do our cells obligingly provide both initial
receptors and means of uptake for these dangerous
toxic proteins? Some ADP-ribosylation reactions
are a natural part of cell function
y
and some hor-
mones, for example thyrotropin, seem to stimulate
their activity. It may be that the toxins use mecha-
nisms designed to respond to normal hormonal
stimulation.
tyrosine-O-sulfate residues may be a widespread
regulatory mechanism.
125 127
One of the proteins
known to contain a tyrosine sulfate residue is the blood
protein fibrinogen. Many polysaccharides and oligo-
saccharides on glycoproteins exist in part as sulfate
esters (Fig. 4-11).
128,129
Carboxyl groups of certain glutamate side chains
in proteins that control bacterial chemotaxis are
methylated reversibly to form methyl esters.
130
This
carboxylmethylation occurs as part of a reaction
sequence by which the bacteria sense compounds that
can serve as food or that indicate the presence of food
(Section D,5, Fig. 11-8, and Chapter 19). Demethylation
occurs by hydrolysis, which is catalyzed by esterases.
Carboxylmethylation also occurs in eukaryotic cells
but is often substoichiometric and part of a mechanism
for repair of isomerized or racemized aspartyl residues
in aged proteins (Box 12-A). However, the major eukary-
otic protein phosphatase 2A is carboxylmethylated at
its C terminus,
131
as are the Ras proteins discussed in
Section D,3.
It was pointed out in Chapter 4 that many proteins,
especially those secreted from cells or taking up resi-
dence within membranes, are glycosylated. Specific
BOX 11-A CHOLERA TOXIN AND OTHER DANGEROUS PROTEINS (continued)
549 C. Regulation of the Activity of Enzymes
glycosyl groups may be removed or added to a protein.
Such alterations may all be regarded as parts of control
mechanisms that direct the proteins to their proper
locations or determine the length of time that they remain
active. The nuclear histones undergo extensive and
reversible acetylation
132,133
which is thought to be
important to replication of DNA and to transcription
(Chapters 27 and 28).
4. Thiol-Disulfide Equilibria
The activity of enzymes is sometimes controlled
by the formation or the reductive cleavage of disulfide
linkages between cysteine residues within the protein.
An example is provided by the effects of light on en-
zymes of chloroplasts. Light absorbed by the photo-
synthetic reaction centers generates NADPH, which
in turn reduces thioredoxin. This is a small protein
containing a readily accessible S S bridge that can be
reduced to a pair of SH groups (Box 15-C). Within
illuminated chloroplasts, these newly formed SH groups
reduce disulfide bridges to activate a series of enzymes
including fructose 1,6-bisphosphatase that participate in
the photosynthetic incorporation of CO
2
into sugars
134,135
(see Chapter 23). Thioredoxins also function within
bacteria, fungi, and animals, serving as electron carriers
for processes, some of which are involved in metabolic
control. A number of known enzymes and other
proteins, including insulin, contain reducible S S
bridges within peptide loops as in the thioredoxins.
Another possible control mechanism for SH-containing
enzymes depends upon formation of mixed disulfides
with small SH-containing metabolites. For example,
cysteamine disulfide, a minor constituent of cells,
cysteine, or some small disulfide, could be released
H
3
+
NCH
2
CH
2
SSCH
2
CH
2
NH
3
+
Cysteamine disulfide (cystamine)
following hormonal or other stimulation and could
react by disulfide exchange (Eq. 11-7) to either inactivate
or activate an enzyme.
Almost all cells contain a high concentration (39
mM) of the thiol-containing tripeptide glutathione
(G SH, Box 11-B). In its disulfide form it participates
in forming disulfide bridges in secreted extracellular
proteins (Eq. 10-9) via intermediate mixed disulfides.
Mixed disulfides with glutathione as well as with other
thiols can also be formed within cells by oxidative
reactions. Reduction of these disulfides by reduced
glutathione will return the enzymes to their reduced
states. The small protein glutaredoxin, whose eukary-
otic forms are also called thioltransferase, resembles
thioredoxins. It undergoes reduction by glutathione
and, in turn, reduces S S linkages in a different set of
proteins than those reduced by thioredoxin.
Low-molecular-mass thiols such as coenzyme A
and protein-bound thiol cofactors such as phospho-
pantetheine are present in all cells. Their SH groups
can also be oxidized to disulfides and it is of interest
that in resting bacterial spores these compounds exist
largely as disulfides or mixed disulfides. Upon germi-
nation of the spores special enzymes reduce the disul-
fides.
136
Some proteins involved in control of protein
synthesis contain SH groups that add covalently to
C-6 atoms of a uracil ring in specific mRNA molecules.
Control of their state of reduction may also be impor-
tant.
137
5. Regulatory Effects of H
+
, Ca
2+
, and Other
Specific Ions
The pH of the cytoplasm is controlled tightly. Yet
transient changes can occur and may affect many aspects
of metabolism. For example, rapid glycolysis leads to
lactic acid formation with an associated drop in internal
pH. Both increases and decreases in pH have been
associated with successive stages in embryonic or
larval development.
138
Cytoplasmic pH changes may
serve as regulatory signals. A well-understood example
is the Bohr effect on oxygenation of hemoglobin
(Chapter 7). Another is the protein kinase C-stimulated
H
+
/Na
+
exchange through membranes. Because the
Na
+
concentration is high outside cells and low inside,
the exchange leads to an increase in cytosolic pH with
many resultant effects on metabolism.
139,140
Exchange
of external Cl

for internal HCO


3

also affects pH.


141
Uptake of Ca
2+
into cells, or release of this ion
from intracellular stores, is a major regulatory mecha-
nism in many if not all cells (see Section E). Mn
2+
activates phosphoenolpyruvate carboxykinase (Eq.
13-46) and may be a regulator of gluconeogenesis.
142
Iron controls the synthesis of ferritin and of transferrin
receptors
137
(Chapter 16). The specific metal ions
present in many biological macromolecules are likely
to participate in additional regulatory processes.
Phosphate and bicarbonate ions are important
substrates for many enzymatic processes and as such
have regulatory functions. Bicarbonate controls the key
enzyme of photosynthesis, ribulose bisphosphate
carboxylase, by carbamate formation (Fig. 13-12).
Chloride ions activate amylases and may affect the
action of G proteins that mediate hormone actions.
Other observed effects of ions are too numerous to
mention.
(11-7)
ESH ESSR
RSSR
Cystamine
(Cystine)
RSH
Cysteamine
(Cysteine)
550 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
BOX 11-B GLUTATHIONE, INTRACELLULAR OXIDATIONREDUCTION BUFFER
In 1888, de Rey-Pailhade discovered the sulfur-
containing tripeptide that we now know as gluta-
thione (G SH).
a
By 1929 it had been characterized
by F. G. Hopkins and others as an unusual tripeptide
present in most, if not all, eukaryotic cells. Within
animal cells the concentration is typically 15 mM.
Lower levels are found in many bacteria.
b g
The most interesting chemical characteristics of
glutathione are the -glutamyl linkage and the pres-
ence of a free SH group. The latter can be oxidized
to form a disulfide bridge linking two glutathione
molecules.
2G SH 2[H] G S S G
E (pH 7) = 0.25 V
It is this redox reaction that has focused attention on
glutathione as an intracellular reducing agent whose
primary function may be to keep the SH groups
of proteins reduced (see Section C). Glutathione
is usually maintained in its reduced form by the
flavoprotein glutathione reductase (Fig. 15-12).
Although it is primarily an intracellular com-
pound, glutathione is secreted by epithelial and
other cells. It may regulate the redox state of pro-
teins in plasma and other extracellular fluids as well
as within cells. In addition, glutathione released
from the liver may be an important source of cysteine
for other tissues. In the endoplasmic reticulum and
the periplasm of bacteria glutathione functions in
crosslinking thiol groups in newly formed proteins
(Eq. 10-9).
Glutathione also has a series of protective
functions. It reduces peroxides via the selenium-
containing glutathione peroxidase (Box 15-H) and
is part of a system for trapping and detoxifying harm-
ful free radicals. The importance of this function is
suggested by the fact that Entamoeba histolytica, an
organism that lacks both mitochondria and aerobic
respiration, produces no glutathione.
h
It may be
that the primary function of glutathione in eukaryotes
is to protect cells against oxygen toxicity associated
with their mitochondria.
i
The renewal of free
radicals and regeneration of protein SH groups
may involve the cooperation of glutathione, glutare-
doxin (Box 15 - C), and ascorbic acid.
j
Another protective function is fulfilled by the
formation of soluble mercaptides and other conju-
gates of glutathione with many foreign substances
(xenobiotics). These conjugates are made by the action
of a large family of glutathione transferases
e,k,l
(see also Chapter 13) which catalyze addition reactions
of the thiolate group of glutathione with epoxides,
alkylating compounds, and chlorinated aromatic
hydrocarbons. The addition step is often followed
by elimination, e.g., of chloride ion, as in the follow-
ing example. Two steps of hydrolysis and an acety-
lation by acetyl-CoA form a mercapturic acid:
O
2
N
NO
2
S C
N
COO

C CH
3
O
H
H
O
2
N
NO
2
S C
NH
3
+
COO

H
O
2
N
NO
2
S CH
2
C C
O
NH
H
C
CH
2
O CH
2
C
H
NH
3
+
COO

CH
2
COO

N
H
H
2
O
Glutamate
H
2
O
Glycine
CoA-SH
Acetyl-S-CoA
G-SH
Cl

Mercapturic acid
Hydrolysis
Addition and
elimination
O
2
N
NO
2
Cl
OOC
H
N
N
H
COO
H NH
3
O
O
H
H
2
C
SH
+

-L-Glutamyl-L-cysteinylglycine Glutathione ( )
551
BOX 11-B (continued)
The mercapturic acids and related compounds
can then be exported from cells by an ATP-dependent
export pump.
m
Glutathione is a coenzyme for
glyoxalase (Eq. 13-33), maleylacetoacetate
isomerase (Eq. 13-20), and DDT dehydrochlorinase.
The latter enzyme catalyzes elimination of HCl from
molecules of the insecticide and is especially active
in DDT-resistant flies.
a
Glutathione is said to be the
specific factor eliciting the feeding reaction of Hydra;
that is, the release of glutathione from injured cells
causes the little animal to engulf food.
Synthesis of glutathione occurs within cells
via the ATP-dependent reactions in the following
scheme.
n,o
Much more -glutamylcysteine is synthe-
sized than is converted to glutathione and the excess
is degraded by -glutamyl cyclotransferase to form
the cyclic amide 5-oxoproline. Cleavage of ATP
is required to reopen the ring to form glutamate.
Although biosynthesis is exclusively intracellular,
most glutathione appears to be secreted and degraded
by extracellular enzymes. The membrane-bound
-glutamyl transferase catalyzes hydrolysis of
Scheme illustrating interrelationships of the biosynthesis and protective, coenzymatic, and transport functions of
glutathione. See also Meister.
c
Oxidized glutathione
(GSSG)
Free
radicals
Peroxides
Trans-
hydro-
genase
NAD(P)H + H
+
Glutathione reductase
NAD(P)
+
Mercaptides
leukotrienes
prostaglandins
estrogens
melanins
etc.
-Glutamyl-
amino acid
Amino acid
inside cell
Uptake and
recycling H
2
O
-Glutamyl
transferase
on membrane
outer surface
Glycine
Cysteine
Glutamate
BIOSYNTHESIS
Cys-Gly
Cysteine
ADP + P
i
-Glutamyl
cysteine
synthetase
GluCys (or AA)
Glutamate
-Glutamyl
cyclotransferase
Recycle
excess
-Glu-Cys
Amino
acid
outside
of cell
ATP
ADP + P
i
2H
2
O
5-Oxoprolinase
ADP + P
i
Glutathione synthetase
GluCysGly
S
Electrophiles, X
Glutathione (GSH)
GluCysGly
SH
X
5-Oxoproline
PROTECTIVE
FUNCTIONS
COENZYME
FUNCTIONS
ATP
ATP
O
HN
COO

C. Regulation of the Activity of Enzymes


552 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
BOX 11-B GLUTATHIONE, INTRACELLULAR OXIDATIONREDUCTION BUFFER (continued)
glutathione to cysteinylglycine which is further
cleaved by a peptidase. The activity of -glutamyl-
transferase varies among tissues and is especially
high in cells of the kidney tubules. The cysteine and
glycine released by the peptidase may reenter the
cells by a Na
+
-dependent process. Meister proposed
that the -glutamyltransferase, acting on amino
acids, forms -glutamyl amino acids (see scheme)
which are released within cells and are cleaved by
-glutamyl cyclotransferase.
c
The cleavage of gluta-
thione would provide the driving force for amino
acid uptake. However, this is probably a minor
pathway.
p
Trypanosomes contain little glutathione but a
large amount of trypanothione [N
1
,N
8
-bis (gluta-
thionyl) spermidine].
q
This diamide of spermidine
(Chapter 24) is in equilibrium with its disulfide, a
24-membered macrocyclic structure, and appears
to have functions similar to those of glutathione.
Trypanothione reductase, which is unique to trypa-
nosomes, is a potential target for antitrypanosomal
drugs,
q s
as is trypanothione synthetase.
t
Bacteria
that do not synthesize glutathione usually accumulate
some other thiol, e.g., -glutamylcysteine or coen-
zyme A, in high concentrations.
u,v
a
Meister, A. (1988) Trends Biochem. Sci. 13, 185188
b
Bernofsky, C., and Wanda, S.-Y. C. (1982) J. Biol. Chem. 257,
68096817
c
Meister, A. (1988) J. Biol. Chem. 263, 1720517208
d
Inoue, M. (1985) in Renal Biochemistry (Kinne, R. K. H., ed), pp.
225269, Elsevier, Amsterdam
e
Orrenius, S., Ormstad, K., Thor, H., and Jewell, S. A. (1983) Fed.
Proc. 42, 31773188
f
Via, J., ed. (1990) Glutathione: Metabolism and Physiological
Functions, CRC Press, Inc., Boca Raton, Florida
g
Dolphin, D., Poulson, R., and Avramovic, O., eds. (1989) Glu-
tathione - Chemical, Biochemical and Medical Aspects (Coenzymes
and Cofactors), Vol. 3, Wiley, New York (Parts A & B)
h
Fahey, R. C., Newton, G. L., Arrick, B., Overdank-Bogart, T., and
Aley, S. B. (1984) Science 224, 7072
i
Garcia de la Asuncion, J., Millan, A., Pla, R., Bruseghini, L.,
Esteras, A., Pallardo, F. V., Sastre, J., and Via, J. (1996) FASEB J.
10, 333338
j
Meister, A. (1994) J. Biol. Chem. 269, 93979400
k
Ji, X., Johnson, W. W., Sesay, M. A., Dickert, L., Prasad, S. M.,
Ammon, H. L., Armstrong, R. N., and Gilliland, G. L. (1994)
Biochemistry 33, 10431052
l
Hebert, H., Schmidt-Krey, I., and Morgenstern, R. (1995) EMBO
J. 14, 38643869
m
Ishikawa, T. (1992) Trends Biochem. Sci. 17, 463468
n
Polekhina, G., Board, P. G., Gali, R. R., Rossjohn, J., and Parker,
M. W. (1999) EMBO J. 18, 32043213
o
Lu, S. C. (1999) FASEB J. 13, 11691183
p
Lee, W., Hawkins, R., Peterson, D., and Via, J. (1996) J. Biol.
Chem. 271, 1912919133
q
Henderson, G. B., Ulrich, P., Fairlamb, A. H., Rosenberg, I.,
Pereira, M., Sela, M., and Cerami, A. (1988) Proc. Natl. Acad. Sci.
U.S.A. 85, 53745378
r
Bollinger, J. M., Jr., Kwon, D. S., Huisman, G. W., Kolter, R., and
Walsh, C. T. (1995) J. Biol. Chem. 270, 1403114041
s
Sullivan, F. X., Sobolov, S. B., Bradley, M., and Walsh, C. T.
(1991) Biochemistry 30, 27612767
t
Henderson, G. B., Yamaguchi, M., Novoa, L., Fairlamb, A. H.,
and Cerami, A. (1990) Biochemistry 29, 39243929
u
Sundquist, A. R., and Fahey, R. C. (1989) J. Biol. Chem. 264, 719
725
v
Swerdlow, R. D., Green, C. L., Setlow, B., and Setlow, P. (1979) J.
Biol. Chem. 254, 68356837
6. Compartments and Organized Assemblies
The geometry of cell construction provides another
important aspect of metabolic control. In a bacterium,
the periplasmic space (Fig. 8-28) provides a compart-
ment that is separate from the cytosol. Some enzymes
are localized in this space and do not mix with those
within the cell. Other enzymes are fixed within or
attached to the membrane. Eukaryotic cells have more
compartments: nuclei, mitochondria (containing both
matrix and intermembrane spaces), lysosomes, micro-
bodies, plastids, and vacuoles. Within the cytosol the
tubules and vesicles of the endoplasmic reticulum (ER)
separate off other membrane-bounded compartments.
The rate of transport of metabolites through the mem-
branes between compartments is limited and often is
controlled tightly.
While many enzymes appear to be dissolved in
the cytosol and have no long-term association with
other proteins, enzymes that catalyze a series of con-
secutive reactions may form complexes within which
substrates are channeled.
143147
Many enzymes are
attached to membranes where they may be held close
together as organized assemblies.
146
This appears to
be the case for oxidative enzymes of mitochondria
(Chapter 18) and for the cytoplasmic fatty acid synthe-
tases (Chapter 21). In bacterial fatty acid synthesis,
the product of the first enzyme is covalently attached
to a carrier and, while so attached, is subjected to
the action of a series of other enzymes. In eukaryotes
several enzymes form domains of a single fatty acid
synthase. Efficient substrate channeling results. Tryp-
tophan synthase (Fig. 25-3) passes indole through a
tunnel between subunits.
146a
Both NH
3
and carbamate
ions pass through tunnels between subunits of E. coli
carbamoyl phosphate synthetase (Eq. 24-22). The
product carbamoyl phosphate may then be passed
directly to aspartate carbamoyltransferase for synthesis
of carbamoylaspartate in the pyrimidine biosynthetic
pathway.
146b
Channeling is sometimes difficult to
553
prove. Geck and Kirsch have provided a generally
useful technique for testing. A large amount of a
genetically modified, inactive form of an enzyme is
added. Unless channeling occurs, this will decrease
the rate of a reaction whose rate is limited by diffusion
or by instability of an intermediate.
146c
Proteasomes (Box 7-A) have enzymatic sites within
a protected box which limits the escape of long peptide
fragments. Membrane anchors (Chapter 8), often con-
sisting of acyl or polyprenyl groups, hold many proteins
to cell surfaces and strong protein protein bonds hold
many others.
146d
Enzymes involved in cell signaling,
and discussed in the following section, are often
anchored close together on membrane surfaces.
148,149
D. Hormones and Their Receptors
A major element in the control of the metabolism
of a cell is provided by chemical messages sent from
other cells and sensed by receptors on the cell mem-
brane or in the cytoplasm. Hormones such as insulin,
adrenaline, and the sex hormones are released from an
organ and travel through the blood, affecting tissues
throughout the body. There are many such hormones.
A large number of other hormones have more local
effects, influencing mostly adjacent cells. When re-
leased at nerve endings these substances are called
neurotransmitters. Chemical messages are probably
sent from virtually all cells to their immediate neighbors,
affecting both their growth and their behavior. In
recent years the very reactive and toxic compounds
nitric oxide (NO) and carbon monoxide (CO) have
been identified as important hormones. These com-
pounds can diffuse rapidly and react with many com-
pounds within cells. Their actions are more rapid,
more rapidly ended, and probably less specific than
those of most hormones.
Hormones bind to receptors on their target
cells. The receptors are often integral membrane
proteins on cell surfaces. Binding of the hormone
often causes a conformational alteration that activates
the receptor. In some cases the receptor is an enzyme
and the hormone an allosteric activator. In others, the
activated receptor interacts with an enzyme in the
cytoplasm or on the membrane facing the cytoplasm.
This enzyme may generate a second messenger, a
substance that can diffuse throughout the cell and alter
metabolism by exerting allosteric effects on various
other enzymes. The best known second messenger is
cyclic AMP but there are many others, a few of which
are listed in Table 11-2.
Some hormonereceptor complexes enter the cell
via endocytosis in coated vesicles. Within the cell both
the receptor and the bound hormone, if a peptide, may
be degraded by proteases. The initial binding of the
hormone may induce the release of a second messenger,
while the degradation of the receptor complex at a later
time releases peptides that may be additional second
messengers. This is one way in which hormones may
elicit a rapid response followed by delayed responses.
While many hormones bind to surface receptors
the steroid hormones, which are lipid in nature, pass
through the cell membrane and bind to receptor pro-
teins in the nucleus. The resulting hormoneprotein
complexes induce changes in gene expression through
regulation of transcription (Fig. 11-1, top). These
receptors are considered in Chapter 22 and hormones
are considered further in Chapter 30.
1. Beta Adrenergic Receptors and Related
Seven-Helix Proteins
Sites that bind adrenaline (epinephrine),
noradrenaline (norepinephrine), and related
catecholamines (see Chapter 30) to almost all cell
surfaces are classified as either adrenergic or
adrenergic receptors. The receptors, which have
been studied the most,
150
occur as two major types.
The
1
receptors have approximately equal affinity for
adrenaline and noradrenaline, whereas the
2
receptors,
the most common type, are more nearly specific for
adrenaline. Binding of the hormone or other agonist
such as isoproterenol to any of the receptors stimu-
lates cAMP formation within the cell.
153
The receptors
belong to a family of integral membrane proteins
related in sequence to the opsins of the retina and
the light-operated proton pump bacteriorhodopsin
(Fig. 23-45) and probably have a very similar three-
dimensional structure. Based upon the known
three-dimensional structures of rhodopsin and bacterio-
rhodopsin (Chapter 23), sequence comparisons, and
much other evidence, all of these receptors are probably
folded into groups of seven hydrophobic membrane-
spanning helices arranged as closely packed bundles
with folded loops protruding into the cytoplasm and
into the extracellular space. The N termini of the proteins
are thought to be in the extracellular space and the C
termini in the cytoplasm as shown in Fig. 11-6.
153a,b,c
Defects in structure or functioning of human receptors
have been associated with both asthma
154
and heart
failure.
155,155a,b
Among the proteins phosphorylated in response
to formation of cAMP are the adrenergic receptors
themselves. Phosphorylation occurs within a cluster
of serine and threonine residues near the C terminus as
is indicated in Fig. 11-6A by the action of various kinases
including a receptor kinase
156,156a
which may be an-
chored nearby.
157
The effect of this C-terminal phospho-
rylation is to decrease the sensitivity of the receptor so
that after a few minutes it conveys a diminished response.
Thus, cAMP exerts feedback inhibition of its own
synthesis. Desensitization upon continuous occupancy
D. Hormones and Their Receptors
554 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
by an agonist is a property of many other receptors as well.
When a receptor is no longer occupied, phosphatases
remove the phospho groups added by the receptor
kinase permitting the sensitivity to rise again. This
phosphorylationdephosphorylation cycle appears
to be only one of several mechanisms by which cells
regulate receptor sensitivity.
155b,158,158a,158b
Many other receptors also have a seven-helix
structure similar to that of the adrenergic receptors.
These include receptors for the following: glucagon,
one of the pancreatic hormones regulating glucose
metabolism;
159,160
vasopressin (Fig. 2-4);
161,162
lutropin,
another pituitary hormone;
163
other gonadotropins;
164
the thyrotropin-releasing factor (TRF; Fig. 2-4);
165
a receptor for KDEL peptide sequences. The KDEL
receptor functions in the return of soluble proteins
containing the KDEL motif from the Golgi to the endo-
plasmic reticulum.
152
HO
HO
C
H
OH
CH
2
N H
CH
3
H
HO
HO
C
H
OH
CH
2
NH
3
HO
HO
C
H
OH
CH
2
N
C
O
CH
2
C
H
OH
CH
2
Noradrenaline
N +
Adrenaline (epinephrine)
+
H
Propranolol
+
H
C
Isoproterenol
+
CH
3
H
CH
3
H
CH
3
CH
3
H
H
Compound Metabolic state Response Location of discussion
Cyclic AMP Stimulation of Increased glycogenolysis, Sections C,2 and D,2;
adrenergic receptors glycolysis
Cyclic GMP Visual stimulation Neuronal signal Chapter 23, Section D
Ca
2+
Ca
2+
channels open Muscular contraction, Box 6-D, Section E
others Chapter 19, Section B,4
Inositol-1,4,5- Stimulation of Opens Ca
2+
channels in ER Section E
trisphosphate and adrenergic receptors,
related compounds various other stimuli
Diadenosine 5'- Oxidative or heat Chapters 28, 29
tetraphosphate (Ap
4
A) stress
Guanosine 5'- Nutritional stress Chapter 29
diphosphate,
3-diphosphate (ppGpp)
Mn
2+
Low glucose Increased PECK activity Chapter 17, Section L
Increased arginase activity Chapter 16, Section E
Cyclic ADP-ribose Chapter 15, Section E
(cADPR) and
2'-P-cADPR
TABLE 11-2
Some Molecules (Second Messengers) That Carry Intracellular Signals
555
G
E
Q
H
W
Y
R
E V H Y G S Q E G T N G N S S
Y
G
Q
E
K
E
N
K L L C E
A
T
H
A
I
N
C
Y
A
N
E
T
C
C
D
F
F
T
N
P
I
Q
M
S
F
L
S
G
L
T
W
I
V
I
L
M
V
R
V
I
A
A
I
T
S
P
F
K Y
Q
S
L
L
T
K
N
K
F
Y
G
M
G
S
M
V
I
V
I
L
V
I
L
A
N
G
F
I
V
L
V
A
I
A
T
H
I
L
F
G
A
A
P
V
V
M
G
L
A
D
L
V
L
A
A
C
F
I
T
S
W
F
E
D
I
S
T
C
L
V
S
A
T
V
T
E
I
I
V
C
L
D
V
A
R
Y
A
Q
S
A
I
A
V
I
S
V
Y
F
S
V
L
P
F
V
M
I
R
S
Y
V
H
V
I
V
N
I
V
F
F
I
C
W
L
P
F
T
L
I
M
G
T
L
G
I
Y
V
E
N
L
L
I
G
I
W
S
N
V
Y
N
F
G
Y
I
L
P
S
R
C
P
Q
D
N
L I
R
K
C
106
S
S
184
V
V
W
V
E
D
R
Q
Q
T
V
D
H
D
A
H
S
R N
P
A L
L
F
A
S
G
N G P Q G M
M
K
M
W
T F G
N
F
W
K
F
E
R
L
Q T
V
T
N
Y
V
F
Q
E
A K R Q L Q
K
I
D
K S E G R F H
V
Q
N
L S Q V E Q D G R T G H G L
R
R
S
S
T
K
L
A
K
H
E
K
L
K
F
C
D
F
R I
A
F
Q
E
L
L
R
R
S
S
L
K
A
Y
G
N
L
D L P G T E D F V
G
H
O
T
V P S D N I D S Q G R N C S T N D S L L
Phosphorylation sites for
adreneric receptor kinase
Cytoplasmic surface
Segments involved in coupling to G
s
1 2 3 4 5 6 7
341
C
Palmitoyl
anchor
NH
2
P
h
o
s
p
h
o
r
y
l
a
t
i
o
n

s
i
t
e
s

f
o
r

p
r
o
t
e
i
n

k
i
n
a
s
e

A
P
Extracellular surface
HOOC
113
79
Figure 11-6 (A) Proposed organization of the human
2
adrenergic receptor. The 413-residue polypeptide chain is arranged
according to the model for rhodopsin as seven transmembrane helices. The two N-glycosylation sites, which may carry very large
oligosaccharides, are indicated by Y. The palmitoylated cysteine 341 is shown with its alkyl side chain embedded in the membrane.
Aspartates 79 and 113, also shown in (C), are shaded. After Strosberg
151
and Scheel and Pelham.
152
(B) Arrangement of the seven
helices suggested by the rhodopsin structure. (C) Hypothetical view of the receptor from the external membrane surface showing
a molecule of noradrenaline bound to hydrophilic residues deep in the cleft between the helices. From Strosberg.
151
Intracellular
Extracellular
7
1
2
3
4
5
6
S S
C
C
O
O

O
C
H O
O
N
1
2
3
4
7
5
6
S S S165
D113
D79
Y316
S207
S204
O
O
O
O
O
H
H
H
H
H
H
O
H
H
N
H
H
+
D. Hormones and Their Receptors
A
B
C
556 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
2. Adenylate Cyclases (Adenylyl Cyclases)
All of the effects of the catecholamines bound to
adrenergic receptors and of glucagon, ACTH, and
many other hormones appear to be mediated by
adenylate cyclase. This integral membrane protein
catalyzes the formation of cAMP from ATP (Eq. 11-8,
step a). The reaction, whose mechanism is considered
in Chapter 12, also produces inorganic pyrophosphate.
The released cAMP acts as the second messenger and
diffuses rapidly throughout the cell to activate the
cAMP-dependent protein kinases and thereby to
stimulate phosphorylation of a selected group of
proteins (Fig. 11-4). Subsequent relaxation to a low
level of cytosolic cAMP is accomplished by hydrolysis
of the cAMP by a phosphodiesterase (Eq. 11-8, step
b).
166,167
In the absence of phosphodiesterase cAMP
is extremely stable kinetically. However, it is thermo-
dynamically unstable with respect to hydrolysis.
The existence of cAMP as a compound mediating
the action of adrenaline and glucagon on glycogen
phosphorylase was first recognized in 1956 by Suther-
land.
168,169
However, for many years most biochemists
regarded cAMP as a curiosity and the regulatory chem-
istry of phosphorylase as an unusual specialization.
That view was altered drastically when cAMP was
found to function as a second messenger in the action
of over 20 different hormones. Phosphorylation by
cAMP-dependent protein kinases regulates several
enzymes concerned with energy metabolism or with
the control of cell division. Phosphorylation reactions
can lead to different responses in different specialized
cells. Proteins of membranes, microtubules, and ribo-
somes are phosphorylated, as are nuclear histones.
Cyclic AMP mediates the action of some neurotrans-
mitters released at synapses and functions as a signal
between cells of some slime molds (Box 11-C). Tran-
scription of genes can be either stimulated or inhibited
by cAMP.
170
Even in E. coli cAMP is generated and acts
as a positive effector for transcription of some genes
(Chapter 28). Cyclic AMP may function in signaling
stress in plants.
171
The isolation and characterization of adenylate
cyclases have been difficult because of their location
within membranes and because there is so little (usually
only 0.001 0.01%) of the membrane protein.
172
The
1060 - to 1250-residue, 120 kDa proteins are also easily
denatured. However, the sequences of cloned adenylate
cyclase genes and the observed patterns of synthesis
of the corresponding mRNAs have been revealing.
There are at least eight mammalian adenylate cyclase
genes with complex regulatory properties.
173 177
The sequences suggest that most of the isoenzymes
are integral membrane proteins, each with 12 transmem-
brane helices organized as two sets of six with a large
~ 40-kDa cytoplasmic domain between the sets. This
is similar to the organization of the cystic fibrosis
transmembrane conductance regulator (Box 26-A) and
some other membrane transporters. However, there is
no firm evidence that adenylate cyclases contain ion
channels. These enzymes are also discussed in Chapter
12, Section D,9.
A quantitative indication of the importance of
the cAMP system within cells can be derived from
measurement of the kinetics of the incorporation of
18
O from water into the -phospho groups of AMP,
ADP, and ATP. This incorporation will result from
hydrolysis of cAMP by the phosphodiesterases that
allow relaxation to a low cAMP level (Eq. 11-8). It is
thought that in human blood platelets this represents
the major pathway of this labeling (Eq. 11-9), which
occurs at a rate of about 1.1 mol of
18
O kg
1
s
1
.
(11-9)
O
O P
O
O
O
OH
Adenine
O
HO OH
Adenine
O
P

O
O
18
OH
18
OH
2
(11-8)
O
O
OH
Adenine
N
N
N
N
NH
2
O
O
O
OH
P
O

O
HO
O
P

O O
P
O
O
HO
P
O
O
P P
i
Adenylate cyclase
H
2
O
Mg
2+
Phosphodiesterase
O
O
O
HO
HO
Adenine
OH
P

O
5'-Adenylic acid (AMP)
3',5'-Cyclic AMP (cAMP)
H
a
b
5'
3'
557
In higher animals cyclic AMP (cAMP) is an
intracellular second messenger, but in the cellular
slime mold Dictyostelium discoideum it serves to
convey signals between cells.
a c
As was mentioned
on p. 20, the organism exists as individual amebas
until the food supply is exhausted. Then the cells
begin to signal their lack of food by secreting pulses
of cAMP. Because they also secrete a phosphodiester-
ase, the cAMP is short-lived.
d
However, it is present
long enough for any other nearby ameba to sense
the gradient of cAMP concentration from one end
of the cell to the other. As little as a 2% difference in
concentration can induce chemotaxis.
e
The amebas
move toward the source of cAMP and emit pulses
of the compound. This results in the formation of
aggregation centers in which the concentration of
cAMP oscillates spontaneously as the cAMP moves
outward in waves. The cells move up the concentra-
tion gradient until the peak of a wave reaches them.
Then they move in a random direction until the next
wave reaches them and again orients their motion.
After ~50 movement steps an aggregation center
contains ~10
5
cells which now follow a development
program. The amebas adhere to each other to form
1- to 2 mm-long multicellular slugs in which all of
the cells move forward together. About 30 h after
aggregation begins the slugs stop and form stalks
with spore-containing fruiting bodies on top. The
BOX 11-C THE ATTRACTION OF DICTYOSTELIUM TO CYCLIC AMP
a
Gerisch, G. (1987) Ann. Rev. Biochem. 56, 853 879
b
Devreotes, P. (1989) Science 245, 1054 1058
c
Alberts, B., Bray, D., Lewis, J., Raff, M., Roberts, K., and Watson,
J. D. (1994) Molecular Biology of the Cell, 3rd ed., Garland, New
York
d
Levine, H., Aranson, I., Tsimring, L., and Truong, T. V. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 6382 6386
e
Kuwayama, H., and Van Haastert, P. J. M. (1996) J. Biol. Chem.
271, 23718 23724
f
Schapp, P., Konijn, T. M., and van Haastert, P. J. M. (1984) Proc.
Natl. Acad. Sci. U.S.A. 81, 2122 2126
g
Shimomura, O., Suthers, H. L. B., and Bonner, J. T. (1982) Proc.
Natl. Acad. Sci. U.S.A. 79, 7376 7379
lead cells in a slug become the stalk.
a,c
Two types of G protein-coupled cAMP receptors
in the cell membranes of the amebas have been
identified. One leads to activation of adenylate
cyclase and the release of new pulses of cAMP and
the other to activation of guanylate cyclase.
e
This
enzyme causes a rapid 7- to 10-fold increase in intra-
cellular cGMP which plays an important role in
controlling chemotaxis.
Some species of cellular slime molds use other
chemical attractants (acrasins). For example, D.
minutum secretes an analog of folic acid
f
and cells
of Polysphondylium violaceum are attracted by the
ethyl ester of N-propionyl--L-glutamyl-L-ornithine-
-lactam.
g
D. Hormones and Their Receptors
Stimulation of the platelets by prostacyclin (prosta-
glandin I
2
; see Chapter 22) leads to a 10- to 40- fold
increase in cAMP concentration and a 4- to 5-fold
increase in the rate of
18
O incorporation.
178
A quite
different soluble adenylate cyclase is present in sper-
matozoa. It is stimulated directly by bicarbonate ions.
It may be a bicarbonate sensor in sperm cells and in
some other bicarbonate-responsive tissues as well as
in cyanobacteria.
178a,178b
3. Guanine Nucleotide-Binding Proteins
(G Proteins)
The adrenergic receptors are not coupled directly
to adenylate cyclase but interact through an inter-
mediary stimulatory protein G
s
, which contains three
subunits, , , and .
179 182
We know that the G
s
protein
associated with adrenergic stimulation is only one of
a very large number of related G proteins, so named
because of their property of binding and hydrolyzing
GTP. In its unactivated state the subunit of a G

heterotrimer carries a molecule of bound GDP. Appar-


ently, the G
s
proteins and the hormone receptors,
which are activated by the binding of hormones, diffuse
within the membrane until they make contact and form
molecules of the G
s
GDP complex (Eq. 11-10, step b).
The complex then undergoes a rapid exchange of the
bound GDP for GTP, after which the hormone and
receptor dissociate. The G

GTP complex may also


dissociate from the complex, perhaps entering the
cytosol as a soluble protein (Eq. 11-10, step c). The
G

GTP complex combines with adenylate cyclase


(step d) and activates it to generate cAMP. However,
the activation is transient. G

also contains GTP-


hydrolyzing (GTPase) activity and within a few min-
utes G

GTP is completely converted to G

GDP and
the adenylate cyclase dissociates (Eq. 11-10, step e).
The G

GDP recombines with the complex which


may serve as a membrane anchor, to complete the
regulatory cycle. The overall effect is for hormone
binding to cause a rapid release of cAMP in a short
burst that may last only about 15 s.
It is not the binding of GTP but a slow subsequent
conformational alteration that activates the G

GTP
adenylate cyclase complex. This was deduced by study
of analogs of GTP, such as guanosine 5'-(, -imido)
triphosphate (GMP-P-(NH)-P or GppNp), which are
558 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Guanosine 5'-(,-imido) triphosphate (GppNp);
in place of the NH, O is present in GTP, S is present is GTPS
N
HN
O
H
2
N
N
N
O
OH OH
CH
2
O
P
O
P
N
P
O

O O

O O HO
O
H

5
Receptor (R)
HR
Hormone (H)
G

GDP
HRG

GDP
GTP
GDP

GTP
Hormone
Activated G protein
AC
Inactive
adenylate cyclase
G

GTPAC
G

GDP
Active adenylate cyclase
P
i
H
2
O
a
b
c
e
d
ATP cAMP
Cyclic AMP
G protein,
GDP complex
(11-10)
not hydrolyzed to GDP. These compounds may require
many minutes to activate the complex, but because
the GppNp is not hydrolyzed, the complex remains
activated and able to catalyze cAMP formation contin-
uously.
Occupied receptors for adrenaline, glucagon,
ACTH, and histamine activate adenylate cyclase via G
s
proteins. Other G
s
proteins, which contain subunits
designated
olf
and which exist as a number of subtypes,
mediate olfactory responses. Subunit
o
is another
specialized polypeptide which is located primarily in
neural tissues. A variety of additional G proteins have
been discovered in organisms ranging from bacteria
to mammals.
179,183 186
All have similar structures with
39 - to 45-kDa subunits, 35- to 36-kDa subunits
and 5 - to 8-kDa subunits. Whereas the subunits
are unique to each G protein, and subunits may be
shared among several G proteins. These proteins appear
to function with many kinds of hormone receptors and
in nerve transmission, secretion, and endocytosis.
187
Mammals can form at least 23 different
s
subunits,
which are coded for by 16 genes. Some of the forms
arise by alternative splicing of mRNA transcribed from
a single gene.
180
There are at least 6 and 12 sub-
units.
179,180,182,188,189
Other G proteins, found in heart
muscle, induce the opening of K
+
or Ca
2+
channels in
cell membranes.
185,190
It has often been assumed that
all of the effects of G proteins are mediated by activated
subunits. However, the complex, which sticks
together very tightly, can also have signaling func-
tions.
191
For example, an auxiliary protein called
phosducin binds to the complex of G
s
, slowing
the reconversion to the trimer.
192
Phosducin was
discovered in rod cells of the retina but has been
found to be distributed broadly in other tissues as
well.
192,193
In yeast the subunit of a G protein linked
to the receptor for the yeast mating factor initiates the
pheromone response of a cell.
193a
There is another large class of receptors whose
occupancy by an agonist leads to inhibition of adenylate
cyclase. These include the
2
adrenergic receptors,
receptors for acetylcholine, adenosine, prostaglandin
E
2
(Chapter 21), somatostatin, and some receptors for
dopamine. Their responses are mediated by inhibitory
proteins G
i
, which closely resemble G
s
in their sizes,
amino acid sequences, and heterotrimeric structures,
but which inhibit adenylate cyclase when activated.
180
A clear distinction between the G
s
and G
i
proteins is
evident in the fact that G
s
is irreversibly activated by
the action of cholera toxin, while G
i
loses its ability
to respond to occupied receptors when modified by
the action of Pertussis toxin (Box 11-A). A specialized
heterotrimeric G
i
protein known as transducin
mediates the light-induced activation of a cyclic GMP
phosphodiesterase in the retina
194,195
(see Chapter 23).
Its subunit is designated
t
. The related gustducin
is found in taste buds.
196
Monomeric G proteins. An entirely different
class of G proteins was discovered when it was found
that the small 189-residue, 21-kDa protein products of
the human oncogenes and proto-oncogenes known as
ras are monomeric G proteins.
186,197 200
There are over
80 related proteins of this group of nine families.
200a
They include the much larger elongation factor EF-Tu,
which functions in protein synthesis (Chapter 29).
Even though there is a large difference in size, the
three-dimensional structures of the GTP-binding
domains of EF-Tu
201 204
and of the 21-kDa Ras p21
proteins
205 209
are very similar (Fig. 11-7). They also
resemble that of adenylate kinase (Fig. 12-30).
205
The
human genome contains four true ras genes: H-ras,
N-ras, K-ras, and K-rasB. The occurrence of mutations
in these genes in human cancer is discussed in Section
H, as are the complex signaling functions of ras genes,
(see also Fig. 11-13).
210
559 D. Hormones and Their Receptors
Other proteins in the Ras superfamily include
some members of the Rho family, which function in
the cytoskeleton.
200a,211,212,212a,b,c,213
These include at
least 14 mammalian proteins, among them RhoA, RhoB,
etc., Rac1, Rac2, etc., and Cdc42. The latter is associated
with the formation of fingerlike extensions of the cell
membrane containing actin bundles.
200a,212d,212e
Genes
rac1 and rac2 encode proteins involved in the initiation
of superoxide radical formation by activated phagocytes
(Chapter 18).
214
The Rab family of proteins are involved
in exocytosis and endocytosis and vesicular transport.
There are 24 or more human rab genes.
212,215,216
Yeast
cells also contain Ras proteins. One RAS gene must be
present for yeast to form spores.
217
ADP-ribosylation
factors (ARFs), which stimulate the action of cholera
toxin A subunit (Box 11-A) are also members of the
Ras superfamily.
120,217a
Like the trimeric G proteins, the monomeric pro-
teins also serve as timed switches that are turned
on by GDPGTP exchange and are turned off by the
hydrolysis of the bound GTP. In the absence of other
regulatory proteins both the exchange of bound GDP
for GTP and the hydrolysis of bound GTP are slow.
While the receptor-associated trimeric G proteins are
activated by the hormone receptors, the monomeric
G proteins are activated toward GDPGTP exchange
by binding to proteins called guanine nucleotide
dissociation stimulators (GDSs).
217b
They speed the
release of GDP from the G protein allowing the active
GTP complex to be formed. The velocity of hydrolysis
of the bound GTP in the activated G protein is greatly
increased by GTPase activating proteins (GAPs).
These auxiliary proteins can be thought of as signaling
molecules that pass their messages to the G protein,
controlling the extent of its activation and therefore
the strength of a signal that it sends on to the processes
that it is controlling.
200,210,218
One group of GAP proteins
are known as regulators of G-protein signaling
(RGS).
218-218b
These proteins, which contain a charac-
teristic 12-residue core, were first recognized as
negative regulators of signaling in yeast cells called
GDP-dissociation inhibitors (GDIs).
219
They can
also be regarded as a family of GAPs.
Acylation and prenylation. The amino terminus
(usually glycine) of the subunit of any G protein is
nearly always converted to an N-myristoyl group.
220223
This modification occurs in a cotranslational process
after removal of the initiating methionine (Chapter
29) and can be described as an acyl transfer from
coenzyme A.
224
The C termini of the subunits of
heterotrimeric G proteins, and also of the monomeric
proteins of the Ras family, also undergo processing.
For example, the C-terminal end of an intact Ras
protein contains 18 residues which probably assume a
largely -helical conformation. A cysteine side chain
near the terminus and having the sequence CAAX is
converted within cells to a thioether with all-trans-
farnesol or longer polyprenyl alcohols.
225
Another
nearby cysteine may form a thioester with palmitic
acid,
221,226,227
for example:
The farnesyl (or longer) polyprenyl and palmitoyl
groups, together with nearby nonpolar side chains,
serve as a membrane anchor. After the prenylation
the C-terminal three amino acids (VVM in N-ras p21)
are removed proteolytically and the new terminal
carboxyl group of the farnesylated Cys 186 is converted
to a methyl ester.
212c,225,228
N-terminal myristoylation
is usually regarded as irreversible. However, the thio-
ester linkages by which palmitoyl groups are attached
to proteins are labile and may be cleaved rapidly by
hydrolases. It follows that palmitoylation can be a
rapid, regulated modification of proteins,
229230a,b
strongly affecting the location and extent of adherance
to membrane surfaces.
Three-dimensional structures. The structure
of the GTP-binding domain of elongation factor EF-Tu
was determined by Jurnak in 1985
201
and that of the
complete three-domain structure later.
202,203
When the
structure of the catalytic domain of the first Ras protein
was determined (Fig. 11-7A) it was clear that it was
similar to that of EF-Tu.
205,207
The same was true for the
transducin
t
,
194,231,232
for the inhibitory G
i1
,
218b,233,234
and for other G proteins.
235
In every case the differ-
ences in structure of the enzyme with GDP or with
analogs of GTP were small and limited to a region
close to the -phospho group of the bound GTP. This
group can be seen clearly in Fig. 11-7A adjacent to
residue 60 of the protein. See also Fig. 12-36.
The strands and helices in the GTP-binding domain
of the G proteins are connected by a series of loops.
The first, second, and fourth loops (residues 10 17,
32 40, and 57 65, respectively) in the Ras structure of
Fig. 11-7A form the catalytic site in which the hydro-
lysis to GDP takes place. The first loop, also called
the P loop, is conserved in all GTP-binding proteins.
In the Ras proteins it has the following sequence:
10 17
GAGGVGKS Ras
GX X X X GKS(T) Consensus
Shaded residues are removed and
C186 is converted to methyl ester
S
O
S
TNGCMGLPCVVM
Palmitoyl group
Farnesyl group
C terminus
186
N-ras p21
. . .
560 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Figure 11-7 (A) Stereoscopic drawing showing -carbon positions in the 166-residue catalytic domain of the human c-H-ras
gene product and the bound GTP analog GppNp (solid bond lines). The intact protein contains an additional 23-residue
C-terminal extension. From Pai et al.
241
(B), (C) Ribbon drawings showing two nearly orthogonal views of a hetero-trimeric
inhibitory G
i
brain protein produced using a cloned bovine gene. (B) The amino termini (N) of the three subunits are seen in
the left-to-right order: , , . A side view of the propeller domain of the subunit is seen at top center. The Ras-like domain
and the additional large helical domain of the subunit are marked. (C) View from the flared end of the propeller looking
toward the subunit. The strands of each propeller blade are labeled A, B, C, and D and the seven blades are numbered
around the periphery of the propeller. From Wall et al.
242
(B) and (C) courtesy of Stephen Sprang.
A
B C
Also shown here is the consensus sequence for all
GTP-binding proteins. Cancer cells frequently have
mutations in Ras at Gly12, which may be substituted
by Asp, Lys, Val, or Arg, and at Gly 13 and Gln 61.
The latter is in loop 4, a part of the sequence that
appears especially mobile but is highly conserved. These
mutations all activate the protein, i.e., they decrease
the GTPase activity, allowing the G protein to remain
in its active conformation longer than normal.
Ras proteins fulfill their functions by interacting
closely with two or more proteins in signaling pathways
as described in Section H. Other G proteins have
additional domains. The 405-residue EF-Tu from
Thermus thermophilus has three domains: the C-terminal
nucleotide-binding domain and two -barrel domains
following it. A major difference in conformation is
observed between forms of the protein with bound
GTP or bound GDP, the GDP form being opened up
by a hinging motion of ~ 90 between domains I and
II.
202,203
The conformational change is triggered by
the GDPGTP exchange. An enhanced rate of GTP
hydrolysis, which results from binding of a transfer
RNA and binding to a ribosome, is apparently accom-
plished by interaction with loop 2 of the catalytic do-
main (corresponding to residues 32 40 in Fig. 11-7A).
The function of this protein is considered further in
Chapter 29.
Transducin has a 113-residue domain inserted into
loop 2 of the catalytic domain. In this case, too, a large
hinging movement is associated with the GDPGTP
exchange. The light-activated receptor rhodopsin
induces the conformational change.
194,231
Again,
structural changes resulting from the presence or absence
of interactions of the protein with the -phospho group
561 D. Hormones and Their Receptors
of GTP are involved. The GTPase activity of hetero-
trimeric G proteins can also be activated by aluminum
fluoride AlF
4
.
232,236
From the X-ray structure of the
transducin GDPAlF
4

complex it is seen that hydrated


AlF
4

is covalently bonded in the position of the -phospho


group and makes hydrogen bonds to active site groups,
mimicking a possible transition state for the GTPase
reaction
232
(see Chapter 12, Section D).
The subunit of the complex of transducin is
a seven-bladed propeller (Fig.11-7C). It is composed
of seven GHWD repeat units: [GH X
n
WD]
4 8

where GH = Gly His, WD = Trp Asp, and X


n
is a
core repeating sequence, usually 32 42 residues in
length. This motif is also found in at least 40 other
eukaryotic proteins.
188,237 240
In the complex (Fig.
11-7B) the subunit assumes an elongated, largely
-helical structure. It is often anchored at its C terminus
by a farnesyl or geranylgeranyl chain, while G

may
be myristoylated or palmitoylated.
195,230,243
The three-dimensional structure of the GDP com-
plex of the intact transducin heterotrimer
195
also shows
a tight interaction between and subunits. The
major interaction is probably disrupted by replacement
of the bound GDP by GTP and the conformational
change that occurs around the - phospho group. This
explains the dissociation of the subunit from upon
activation. An entirely similar picture has been obtained
for the action of the inhibitory G protein, G
i2
, for which
structures of the subunit and of the heterotrimer
(Fig. 11-7,B,C) have been determined.
188,233,234,242
The
structures resemble those of transducin, but differ in
details.
4. Guanylate Cyclase (Guanylyl Cyclase),
Nitric Oxide, and the Sensing of Light
Formation of the less abundant cyclic guanosine
monophosphate (cGMP) is catalyzed by guanylate
cyclases found in both soluble and particulate frac-
tions of tissue homogenates.
244 247
However, its sig-
nificance in metabolism is only now becoming well
understood. There are cGMP-dependent protein
kinases,
247
but, until recently cGMP could not be
regarded as a second messenger for any mammalian
hormone. Now we know that cGMP has several
essential functions. It mediates the effects of the atrial
natriuretic factor, a peptide hormone causing dilation
of blood vessels (Box 23-D),
245,248 250
and also of a pep-
tide called guanylin, which is formed in the intestinal
GMP O
P
O Al
O

O
OH
2
F F
F F
2

epithelium.
245,251
The receptors for both of these peptides
are transmembrane proteins with cytoplasmic domains that
have guanylate cyclase activity. The released cGMP appears
to induce relaxation of smooth muscles of the blood
vessel walls. Another mediator of smooth muscle
relaxation is the endothelial cell-derived relaxing
factor, which is evidently nitric oxide, NO
252
(see
Chapter 18). A soluble guanylate cyclase appears to be
an NO receptor and is activated by binding of the NO
to a heme group in one of the two subunits of the
cyclase.
253 255
The cyclase can also be activated by carbon
monoxide, CO, in a similar fashion.
256,257
Carbon mon-
oxide is produced in the body by degradation of heme
(Fig. 24-24) and is thought to be a neurohormone.
258
Cyclic GMP also plays an important role in vision.
Specific phosphodiesterases hydrolyze cGMP to GMP.
The cGMP phosphodiesterases of the rod and cone
cells of the retina are activated by the G protein trans-
ducin, which has been activated by light absorbed by
the visual pigments.
194
The resulting decrease in the
cGMP concentration is thought to cause the closing of
cation channels and thereby to initiate a nerve impulse
(see Chapter 23). Cyclic nucleotide forms of the
pyrimidine nucleotides are present only in very small
amounts in cells.
5. Bacterial Chemoreceptors
Bacteria are attracted to foods with the aid of
chemoreceptors that bind certain amino acids such as
aspartate. The receptors send signals to the mechanisms
that ensure that the bacterium is swimming toward
the food
113,259, 259a
(more details are given in Chapter
19). The aspartate and serine chemoreceptors found in
membranes of E. coli and Salmonella typhimurium are
among the most carefully studied of all receptors. Like
the seven-helix receptors, they have an extracellular
sensory domain to which the signaling molecule,
aspartate or serine, binds, and a transmembrane helical
bundle. In these receptors the transmembrane part
consists of four helical segments coming from the two
subunits of the dimeric protein. For the aspartate
receptor there is a high-resolution X-ray structure for
the cytoplasmic domain
260,261
including the bound
aspartate
262
as is shown in Fig. 11-8A,B.
260,263
The
structures of the receptors for aspartate, serine, and
many other signaling molecules are evidently very
similar. The structure of the cytosolic domain of the
serine receptor has been determined and a model for
the complete receptor has been proposed.
259a
The
molecular mass of the 188-residue extracellular domain
is ~18 kDa, while that of the larger cytoplasmic do-
mains is ~36 kDa. The latter includes a linker region
as well as a long four-helical bundle domain which
can be divided into a methylation domain and a sig-
naling domain as shown in (Fig. 11-8C).
562 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
26 nm
~4.0 nm
~8.0 nm
380 nm
2.5 nm
Figure 11-8 (A) Stereoscopic view of the sensory domain of the dimer with modeled transmembrane region. Each monomer
contains a four -helix bundle with two helices continuing through the membrane. From Scott et al.
260
(B) Stereoscopic image
of aspartate in the major binding site of the receptor. The atomic model of aspartate (green) has been fitted into the observed
difference electron density map. From Yeh et al.
262
(C) Model of an intact E. coli serine chemoreceptor. Left. Ribbon drawing
viewed perpendicular to the molecular twofold axis. Methylation sites are represented by the dark balls by the cytoplasmic
domain. The bound serine is drawn as a partially hidden green ball at the upper left in the extracellular domain. Right.
Diagram of the receptor. The presumed membrane bilayer is represented by the gray band. Positions of some residues are
marked on the left side. On the right, the numbers of residues in various peptide segments are indicated. From Kim et al.
259a
Courtesy of Sung-Hou Kim.
A
B
C
What happens chemically when aspartate binds
into a deep pocket in the center of the sensory domain?
What kind of signal can be passed from the aspartate
to the signaling domain? Several possibilities were
discussed by Kim,
264
who suggested that the binding
of aspartate causes the rotation of the sensory domain
of one subunit relative to that of the other and that this
rotation is transmitted through the membrane to the
signaling domain. The signaling domain of the receptor
forms a complex with a multimeric protein histidine
kinase called CheA (chemotaxis protein A) together
with an auxiliary protein CheW. A more likely possi-
bility is that some kind of piston action occurs between
the transmembrane helices of the two subunits.
264a
Binding of aspartate to the receptor, and the associated
alterations in the receptorCheACheW complex, causes
a strong decrease in the catalytic activity of CheA.
In its active form CheA undergoes autophos-
phorylation, that is, the phosphorylation of a histidine
imidazole group in one of its subunits by the protein
kinase active site of an adjacent subunit. The phospho
group is then transferred from phospho-CheA to
another protein, CheY. Phospho-CheY interacts with
the flagellar motor proteins (Chapter 19) periodically
causing a reversal of direction of the bacterial flagella.
As a result the bacteria tumble and then usually move
R 73
R 69
R 64
563 E. Calcium, Inositol Polyphosphates, and Diacylglycerols
in a new direction. If the attractant molecule aspartate
is present on the receptor the signal is weakened and
the bacteria continue to swim in the same direction
toward food. Phospho-CheY is spontaneously hydro-
lyzed to remove the phospho group, but the dephos-
phorylation is promoted by an additional protein CheZ.
At least two other auxiliary proteins also function in
this control system. CheR is a methyltransferase
that methylates the glutamate side chain carboxylates
in the methylation domain of the coiled coil region and
CheB is a methylesterase that removes the methyl
groups. These two proteins control the methylation
level in the coiled coil region of the receptor (Fig. 11-8A).
Increased methylation increases the strength of the
signal sent from the receptor, perhaps because methyl-
ation removes negative carboxylate charges that repell
each other and weaken the structure, interfering with
signaling.
259
See also Fig. 19-3.
E. Calcium, Inositol Polyphosphates, and
Diacylglycerols
Calcium ions entering cells from the outside or
released from internal stores trigger many biological
responses (see Box 6-D). Within cells Ca
2+
often accu-
mulates in mitochondria, in the ER, or in vesicles called
calciosomes.
265
Release of the stored Ca
2+
is induced
by hormones or by nerve impulses. For example,
impulses flow from the nerve endings into the muscle
fibers and along the invaginations of the plasma mem-
brane called transverse tubules (Chapter 19). There
they induce release of Ca
2+
from the ER. The released
ions activate enzymes
266
and induce contraction of the
muscle fibers. In many cells, Ca
2+
causes release of
secreted materials, for example, neurotransmitters in
the brain.
267,268
1. Alpha Adrenergic Receptors
The release of stored Ca
2+
is often triggered by
adrenergic receptors.
269
Like adrenergic receptors,
the receptors are activated by adrenaline. Specific
inhibitors distinguish them. For example, the recep-
tors are inhibited by propranolol, while the receptors
are blocked by phenoxybenzamine. The synthetic
agonist phenylephrine activates only receptors and
no increases in cAMP or in protein kinase activity are
observed.
There are two major adrenergic receptor sub-
types.
151,269
Activation of the
2
receptors, which are
present in various tissues including blood platelets,
causes inhibition of adenylate cyclase. This inhibition
is evidently mediated by the G
i
protein considered in
Section D,3. The nucleotide sequences of cloned
2
receptor genes and other properties suggest close
structural similarity to the receptors.
270 273
Subtle
differences in hydrogen bonding to the serine side
chains shown in Fig. 11-6C may distinguish
2
from

2
receptors.
273
A characteristic of
2
receptors is that
pertussis toxin (Box 11-A) abolishes the inhibition of
adenylate cyclase, which they mediate. In contrast,
the action of
1
adrenergic receptors is not affected by
pertussis toxin.
The
1
receptors are activated not only by catecho-
lamines but also by the hormones vasopressin and
angiotensin II. Binding of these hormones to
1
receptors induces a complex response that involves
rapid hydrolysis of phosphatidylinositol derivatives
and release of Ca
2+
into the cytoplasm, and of diacylgly-
cerols into the lipid bilayer of the membrane. The response
is mediated by another G protein called G
q
.
274,275
When
this G protein is activated it induces the hydrolysis of
phosphatidylinositol 4,5-bisphosphate (PtdInsP
2
),
a normal minor component of the lipid bilayer, by
phospholipase C (phosphoinositidase C).
265,276 281
The products, inositol 1,4,5-trisphosphate [abbrevi-
ated InsP
3
or Ins(1,4,5)P
3
], and diacylglycerol (DAG)
are both regarded as second messengers for the catecho-
lamines acting on
1
receptors and for about 20 other
hormones, neurotransmitters, and growth factors upon
binding to their specific receptors.
269,282
In addition to
vasopressin, the gonadotropin-releasing hormones,
histamine, thrombin (upon binding to platelet surfaces),
and acetylcholine (upon binding to its muscarinic
receptors; Chapter 30) all stimulate inositol phosphate
release.
2. Phosphatidylinositol and the Release of
Calcium Ions
The phosphoinositides constitute ~2 8% of the
lipid of eukaryotic cell membranes but are metabolized
more rapidly than are other lipids.
265,278,279,283285
A
simplified picture of this metabolism is presented in
Fig. 11-9. Phosphatidylinositol is converted by the
consecutive action of two kinases into phosphatidyli-
nositol 4,5-bisphosphate.
286,287
The InsP
3
released from
this precursor molecule by receptor-stimulated phos-
pholipase C is thought to mobilize calcium ions by
HO
C
H
OH
CH
2
N H
CH
3
H
Phenylephrine
+
Receptor G
q
phospholipase C
inositol phosphates
+ diacylglycerol (DAG) increased [Ca
2+
] (11-11)
564 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
opening gates of calcium channels in membranes of
the ER or of calciosomes.
282,288289a
It diffuses across
the peripheral cytoplasm to InsP
3
receptors which are
embedded in the membranes of the ER.
282,290
One of
the several isotypes of InsP
3
receptors is a 2749-residue
protein thought to contain a calcium ion channel.
290
Similar receptors are also found in inner membranes
of the nucleus.
291,292
Several uncertainties have complicated our under-
standing of the role of Ca
2+
in signaling. What is the
source of Ca
2+
? How much of it enters cells from the
outside and how much is released from internal stores?
Where are the internal stores? What other kinds of
ion channels are present and what second messengers
regulate them? The sarcoplasmic reticulum of skeletal
muscle and also membranes in many other cells contain
ryanodine receptors as well as InsP
3
receptors.
282,293
Both of these receptors have similar structures and
contain Ca
2+
channels. However, the ryanodine receptors
are activated by cyclic ADP ribose (cADPR),
294,295
which was first discovered as a compound inducing
the release of Ca
2+
in sea urchin eggs.
296
The 2-phospho
derivative of cADPR may also have a similar function.
297
Cyclic ADP-ribose (cADPR)
Phospholipase C, which initiates the release of
phosphatidylinositol derivatives, also requires Ca
2+
for activity. It is difficult to determine whether release
of Ca
2+
is a primary or secondary response. There are
three isoenzyme types of phospholipase C, , and
and several subforms of each with a variety of
regulatory mechanisms.
298 300a
For example, the iso-
enzymes are activated by binding to the tyrosine kinase
domain of receptors such as that for epidermal growth
factor (see Fig. 11-13). In contrast, the forms are
often activated by inhibitory G
i
proteins and also by
G
q
, which is specific for inositol phosphate release.
Calcium ions are usually released in distinct pulses
or quanta. The kinetic characteristics of the system
of receptors, diffusing InsP
3
, calcium buffers, and cal-
cium pumps in the cell membrane, and the membrane
potential may account for this behavior.
174,301 306
Since
O
OH OH
H
2
C
N
H H
N
N
N
O
OH OH
H
CH
O
P
O
O O

P
O

O
O
H
NH
2
+
2'-phospho in 2'-P-CADPR
the phosphoinositols are chelators of Ca
2+
, the equilibria
involved in this control system are complex.
307
In addition to InsP
3
, several other inositol deriva-
tives are released by adrenergic stimulation. Inositol
1,3,4,5-tetrakisphosphate (InsP
4
) is formed from
InsP
3
by action of a soluble kinase.
308,309,309a,b
A contro-
versial suggestion is that InsP
4
may induce opening
of Ca
2+
channels through the outer membrane of the
cell
310 312
and may also function to promote storage of
Ca
2+
. It also acts as a transcriptional regulator. Other
possible second messengers are inositol 1,2-cyclic-3,4-
trisphosphate and related metabolites that arise by the
action of phospholipase C on PtdIns 4-P followed by
additional actions of kinases and phosphatases.
278,309,313315
A different inositol tetrakisphosphate, Ins(3,4,5,6)P
4
,
may control chloride ion channels.
316,317
Both inositol
1,3,4,5,6-pentakisphosphate (InsP
5
) and inositol hexa-
kisphosphate (InsP
6
) are also found in plants
318
and
animals.
319
InsP
5
serves as an allosteric effector regu-
lating hemoglobin in avian erythrocytes (Chapter 7).
InsP
6
, also known as phytate, is present in large
amounts in cereals and recently has been found to
play a role in regulating the export of mRNA from the
nucleus.
319a
Additional phospho groups can be added
to InsP
6
to form pyrophosphates
320,320a
and other com-
plex polyphosphates.
321
Both InsP
5
and InsP
6
can also
serve as precursors to Ins(1,3,4,5)P
4
and Ins(1,4,5)P
3
by
hydrolytic dephosphorylation.
322
Following stimulation of a cell the induced meta-
bolism of phosphoinositides decays rapidly. The diacyl-
glycerols are converted into phosphatidic acid and
resynthesized into phospholipids (Chapter 21). The
InsP
4
, InsP
3
, and other inositol metabolites are hydro-
lyzed by phosphatases.
278,309,323 327
Two of these
phosphatases are inhibited by Li
+
as indicated in
Fig. 11-9. They may represent one site of action of
lithium ions in the brain.
324,327,328
Lithium salts are
one of the most important drugs for treatment of
bipolar (manic-depressive) illness. By blocking
the release of free inositol, which can be resynthesized
into PtdInsP
2
, Li
+
may prevent neuronal receptors
from becoming too active. However, the basis for the
therapeutic effect of lithium ions remains uncertain.
The diacylglycerols released by phospholipase C
diffuse laterally through the bilayer and, together
with the incoming Ca
2+
, activate protein kinases C.
These kinases also require phosphatidylserine for
their activity and phosphorylate serine and threonine
side chains in a variety of proteins.
329330b
They are
stimulated by the released unsaturated diacylglycerols.
In addition protein kinases C can be activated by
phorbol esters, which are the best known tumor
promoters (Box 11-D). The diacylglycerol requirement
favors a function for these protein kinases in membranes.
They also appear to cooperate with calmodulin to
activate the Ca
2+
-dependent contraction of smooth
muscle.
330
565
Figure 11-9 Scheme showing synthesis and release of diacylglycerol and inositol phosphates and their regulation of calcium
concentration in response to hormonal stimulation.
Hormone
Receptor
G protein
O
P
Often
arachidonoyl
Phosphatidyl-Ins (3,4) P
2
Phosphatidyl-Ins 4-P
Phosphatidyl-Ins (4,5) P
2
Phospholipase C
(phosphoinositidase C)
Eicosanoids
Arachidonate
Monoacylglycerol
Resynthesis
of PI
CH
2
H
2
C
R
1
R
2
C
O
H
C
C
O
O
O
HO
HO
OH
OH
OH
HO
HO
OP
OP
OH
OP
+ Ca
2+
Ins (1,4,5) P
3
Ins (1,4) P
2
Phosphatidylinositol (PtdIns)
Li
+
Ins 1-P
Ins (1,3,4) P
3
Ins (1,3,4,5) P
4
Ins (1,3,4,5,6) P
5
Ins P
6
(Phytate; present in large amounts in cereal grains)
(Allosteric effector of hemoglobin in birds)
Ca
2+
channels in
plasma membrane
+
Receptors and Ca
2+
channels in ER and
in plasma membrane
ATP
Ins (1,3,4,6) P
4
Ins (Free
myo-inositol)
Ins (3,4) P
2
Ins 4-P
1
4
5
1
4
5
+
+
Diacylglycerol
CH
2
CH
2
OH
R
1
R
2
C
O
H
C
C
O
O
O
Protein kinase C
+ +
Ca
2+

See Fig. 21-5


Pyrophosphate
derivatives
Ins P
5
P
Ins P
6
P
O
O
O

Phosphatidyl-Ins (3,4,5) P
3
Phosphatidyl-Ins 3-P
5-Kinase
3-Kinase
5-Kinase
5-Kinase
3-Kinase
H
2
O
PTEN
E. Calcium, Inositol Polyphosphates, and Diacylglycerols
Diacylglycerols released by phospholipase C
usually contain arachidonic acid in the 2 position.
Hydrolytic cleavage of this linkage
284,331
is a major
source of arachidonate for synthesis of eicosanoids
such as the prostaglandins whose functions are
discussed in Chapter 21. Arachidonate can also be
formed directly by the action of phospholipase A
2
on
membrane phospholipids
332,332a
(see the structure on
p. 566). It has been suggested that protein G
q
, which
activates the hydrolysis of phosphoinositides, may
also regulate phospholipase A
2
directly. Released
diacylglycerols may not only activate protein kinases
C but also have a direct role in promoting the mem-
brane fusion required in exocytosis and endocytosis.
332
Other breakdown pathways of phosphatidylinositols
are also indicated in the following structure.
298,333
Initially most attention was paid to the water-
soluble inositol phosphates that are released from
phosphoinositides. However, phosphoinositol deriva-
tives that retain the diacylglycerol part of the molecule
have regulatory functions while remaining in mem-
branes. A phosphatidylinositol 3-kinase phosphoryl-
ates the 3-OH of inositol in PtdIns, PtdIns 4-P and
PtdIns (4,5)P
2
to give PtdIns 3-P, PtdIns(3,4)P
2
, and
PtdIns(3,4,5)P
2
, respectively.
335 337d
However, they are
ideally suited to function as spatially restricted mem-
brane signals.
337a
They affect protein kinase C as well
as the Ser / Thr protein kinase Akt (discussed in Section
F,3),
326b
and have several functions in vesicular membrane
transport and in regulation of the cytoskeleton.
337e,f
566 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
C
O
O
C
O
C
H
2
C
H
2
C
H
O
P
O

O
O
Arachidonoyl group
(P)HO
O
OH(P)
OH
OH
HO
2
1
6
5
4
3
Phospholipase A
1
Phospholipase C
Phospholipase D
Phospholipase A
2
Phosphatidylinositol
1. Phospholipase A
1
Fatty acid + lysophosphatidylinositol
2. Phospholipase A
2
Arachidonate + lysophosphatidylinositol
3. Phospholipase C Ins 1-P, Ins(1,3)P
2
, Ins(1,4)P
2
, Ins(1,4,5)P
3
+ cyclic forms + diacylglycerol
4. Phospholipase D Ins(4,5)P
2
+ phosphatidic acid
323,334
Characteristic of many of the phosphoinositide-
regulated proteins is a proline-rich PH domain (Table
7-3) which can transmit regulatory signals to additional
proteins in a cascade. Their importance is emphasized
by the finding that a 405-residue phosphatase PTEN
(named after its gene symbol) which catalyses hydro-
lytic removal of one phospho group from PtdIns(3,4,5)
P
3
to form PtdIns(3,4) P
2
, (Fig. 11-9) is a major human
tumor suppressor (see Box 11-D).
338,338a,b
These lipids
are also important in insulin action (Section G,3). The
steroid-like fungal metabolite wortmannin is a specific
inhibitor of the 3-kinase.
339,339a
F. Regulatory Cascades
The effect of a regulated change in the activity of
an enzyme is often amplified through a cascade mech-
anism. The first enzyme acts on a second enzyme, the
second on a third, etc. The effect is to rapidly create a
large amount of the active form of the last enzyme in
the series.
We have already considered regulatory cascades
initiated respectively by the and
2
adrenergic recep-
tors. The effect of these cascades on glycogen phos-
phorylase is outlined on the left side of Fig. 11-4. One
branch of the cascade sequence begins with release of
adrenaline under control of the autonomic nervous
system. In muscle, binding of this hormone to the
receptors on the cell membrane releases cAMP which
activates a protein kinase. The kinase then phosphoryl-
ates phosphorylase kinase. At this point the muscles
are prepared for the rapid breakdown of glycogen.
However, an additional initiating signal is the release
of Ca
2+
into the cytoplasm in response to impulses to
specific muscles via the motor neurons. Calcium ions
can also be released in the liver by adrenergic stimu-
lation. Phosphorylase kinase is activated by the calcium
ions, and in their presence it converts inactive phos-
phorylase b to the active phosphorylase a. Both protein
kinases and phosphorylase kinase also act on glycogen
synthase, phosphorylating it and converting it to an
inactive form. This turns off the biosynthetic pathway
at the same time that glycogenolysis (glycogen break-
down) is turned on. Spontaneous reversion of the
enzymes to their resting states occurs through the action
of phosphatases that cut off the phospho groups placed
on the protein by the kinases. Also essential are phos-
phodiesterases, which destroy the cAMP, and the cal-
cium ion pump, which reduces the concentration of
the activating calcium ion to a low level.
Elaborate cascades initiate the clotting of blood
(Chapter 12) and the action of the protective comple-
ment system (Chapter 31). Cascades considered later
in the book are involved in controlling transcription
(Fig. 11-13) and in the regulation of mammalian pyru-
vate dehydrogenase (Eq. 17-9), 3-hydroxy-3-methyl-
glutaryl-CoA reductase and eicosanoids (Chapter 21),
and glutamine synthetase (Chapter 24).
1. Advantages of Regulatory Cascades
Computer simulations as well as studies of experi-
mental models have led to the following conclusions.
340,341
Even simple cascade mechanisms, such as the one
shown in Fig. 11-10, can provide a more flexible response
O
O
O
O O
C
CH
3
CH
3
CH
3
O
Wortmannin
H
3
C O
567 G. Insulin and Related Growth-Regulating Hormones
to allosteric effectors (such as e
1
in the figure) than if
the effector acted directly on the enzyme rather than
on the protein kinase. The cascade also provides
amplification. This is especially true if additional
cycles are added. A response can result from the bind-
ing of only a small number of hormone molecules to a
receptor in a cell membrane or from activation of only
a few molecules of a protease in the initiation of blood
clotting. A striking amplification occurs in visual
responses. Under appropriate conditions a single
quantum of light falling on a receptor cell in the retina
of an eye can initiate a nerve impulse (Chapter 23).
The latter requires the flow of a large number of Na
+
ions across the plasma membrane. It would be hard to
imagine how absorption of one quantum could initiate
a photochemical reaction leading to that much sodium
transport without intermediate amplification stages.
Another advantage of cascades is that they may provide
ultrasensitive responses. Not only can a response
be sensitive to a higher power than the first of the
concentration of a signaling molecule but also the
amplification provided by the cascade confers a high
sensitivity to the response.
342
Cascade systems also provide for response to more
than one allosteric stimulus in a single pathway. Thus,
as shown in Fig. 11-4, glycogen catabolism can be initiated
in more than one way. Two pathways are known for
initiation of both blood clotting and activation of the
complement system. Many pathways activate the
MAP kinase pathway shown in Fig. 11-13.
2. Substrate Cycles
Although cascade systems offer advantage to cells,
there is a distinct energy expenditure associated with
the controls.
343
As can be seen in Fig. 11-10, in addition
to providing for turning on and turning off the regulated
enzyme, the kinase and the phosphatase together
catalyze hydrolysis of ATP to ADP and inorganic
phosphate. Thus, the regulated enzyme is continually
cycling between active and inactive forms. The relative
amounts of each are determined by the amounts of the
effectors e
1
and e
2
, the concentrations of modifying
enzymes, and the kinetic constants. Such cycles have
sometimes been called futile cycles because they
seemingly waste ATP. This is particularly true of cycles
that involve major metabolites, sometimes called
substrate cycles. An example is the conversion of
fructose 6-P to fructose 1,6-bisphosphate by phospho-
fructokinase and hydrolysis of the bisphosphate back
to fructose 6-P by a phosphatase (Fig. 11-2). However,
the cycles are not futile because the hydrolysis of the
ATP provides the energy required to maintain the
concentrations of modified (phosphorylated) enzyme
or enzymes at steady-state levels that are required for
efficient catalysis.
341,344
The energy utilized in this way
in regulation of enzymes is often small.
345
Substrate
cycles are not only an unavoidable consequence of the
need to regulate enzymes but also provide for improved
sensitivity in metabolic control. Consider Fig. 11-2
again. If the enzymes are set for conversion of fructose
1,6-P
2
to glycogen (backwards direction), the flux of
materials flowing in the forward direction (glycolysis)
will be efficiently curtailed because any fructose 1,6-P
2
formed will be hydrolyzed rapidly by the active phos-
phatase. Under these circumstances the flux in the
forward direction will be ultrasensitive to the activa-
tion of phosphofructokinase and to the inhibition of
fructose 1,6-bisphosphatase.
346,347
G. Insulin and Related Growth-Regulating
Hormones
Since its isolation in 1921, insulin has been the
object of an enormous amount of experimentation
aimed at clarifying its mode of action. It is produced
by the cells of the pancreatic islets of Langerhans
and released into the bloodstream in response to ele-
vated glucose levels. The absence of insulin or of a
normal response to insulin results in the condition
of diabetes mellitus, which is the most prevalent
human metabolic disorder (see Box 17-G).
348
Enzyme-P Enzyme
P
i
H
2
O
e
1
Kinase (active)
Effector e
1
K
1
Kinase (inactive)
e
2
Phosphatase (active)
Effector e
2
K
2
Phosphatase (inactive)
ATP ADP
Figure 11-10 A monocyclic regulatory cascade involving
phosphorylation and dephosphorylation of an enzyme.
After Stadtman and Chock.
341
568 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Insulin or insulin-like material is also produced in
ciliated protozoa, vertebrate and invertebrate animals,
fungi, green plants,
349,350
and even in E. coli.
351
1. Metabolic Effects of Insulin
Insulin has many effects on metabolism.
348,352,353
(Some of these are listed in Table 17-3). They can be
summarized by saying that: (1) In most tissues insulin
stimulates synthesis of proteins, glycogen, and lipids.
(2) It affects the permeability of membranes, promoting
the uptake and utilization of glucose and amino acids
and of various ions from the blood. (3) It promotes
both synthesis of glycogen and the breakdown of
glucose by glycolysis. At the same time, it inhibits
synthesis of glucose from amino acids by the gluco-
neogenesis pathway. The effects are not uniformly
the same in all tissues. Some can be observed within
a few minutes after administration, presumably as a
result of regulation of enzymes. Effects on mRNA
metabolism, protein synthesis, and cell growth are
seen at later times.
The uptake of glucose by brain, liver, kidneys,
erythrocytes, and the islets of Langerhans is unaffected
by insulin. However, in muscle and adipose tissues
insulin stimulates glucose uptake. Part of this effect
results from insulin-induced translocation of molecules
of the 509-residue glucose transport protein GLUT4
(Chapter 8) from the cytosol into the plasma membrane
where it can function.
354 356a
Insulin apparently also
increases the rate of synthesis of the transporters.
Insulin stimulates the phosphorylation of serine
side chains of many proteins, including ATP citratelyase,
acetyl-CoA carboxylase, and ribosomal subunit S6.
At the same time it stimulates the dephosphorylation
of other proteins, including acetyl-CoA carboxylase,
glycogen synthase in skeletal muscle (Fig. 11-4),
49
pyruvate dehydrogenase, and hormone-sensitive lipase
in adipose tissue. Yet another effect of insulin is to alter
the amounts of specific messenger RNA molecules. For
example, the transcription of the gene for phosphoenol-
pyruvate carboxykinase (PEPCK; Eq. 13-46) a key enzyme
in gluconeogenesis, is inhibited by insulin within
seconds after binding. The hexokinase isoenzyme
called glucokinase phosphorylates glucose to glucose-
6-P in liver and in pancreatic cells. Its synthesis in
liver is induced by insulin. (However, in the cells
the synthesis of glucokinase is induced by glucose.)
2. Insulin Receptors
All of the effects of insulin appear to result from
its binding to insulin receptors, of which ~10
2
to 10
5
are present in the plasma membranes of most animal
cells. First isolated in 1972,
357
insulin receptors and
their cloned genes have been studied intensively. The
receptors are
2

2
disulfide crosslinked oligomers
composed of pairs of identical 120 - to 135-kDa sub-
units and 95-kDa subunits. An and a subunit are
cut from a single precursor chain. The human insulin
receptor precursor exists as two isoforms, A and B,
which arise as a result of a difference in splicing of the
mRNA.
358
Twenty-one introns are removed by splicing
to form the mRNA for the 1355-residue B precursor.
348,358a
The mRNA for the A form lacks a 36-nucleotide segment
(exon 11) that is discarded during splicing. The chains
come from the N-terminal part of the precursor and
the chains from the C-terminal part. The receptor
sequence is numbered as in the longer B form precursor.
(However, many authors number the chains of the
A form receptor as in its precursor, 12 less than the
numbers given here for residues 719 or higher
359
).
The B form receptor has 731-residue chains while
the A form has 719-residue chains as a result of the
missing sequence from exon 11 (see Fig. 11-11A). Four
residues (732735 in the B form) are cut out and dis-
carded, leaving 620-residue chains for both isoforms.
The chains become linked by three or more disulfide
crossbridges.
360
The two subunits, which contain the insulin
binding sites, are apparently present entirely on the
outer surface of the plasma membrane. The subunits
pass through the membrane with their C termini in the
cytoplasm.
361363
Both and subunits are glycoproteins.
Study of the amino acid sequences suggests that only
one 23-residue segment (residues 930 952 of the B iso-
form) of hydrophobic amino acid residues in each
subunit is likely to exist as an helix that spans the
membrane. This raises several questions. How can a
signal be sent from the cell surface into the cytoplasm
through a single pair of helices? Are there additional
parts of the receptor that span the bilayer of the mem-
brane? To make it easier to visualize these questions
refer to Fig. 11-11B, which is a more realistic, although
fanciful, drawing than that in Fig.11-11A. Electron
microscopy and crystallography are now providing
the first direct images of the receptor.
363a, b, c
One possible way in which a signal could be sent
through the membrane is for binding of insulin to
promote aggregation of two or more receptors.
364
If
insulin induces the receptors to stick together on the
outside of the cell, the parts protruding into the cyto-
plasm would also tend to aggregate. This could induce
a response. A second possibility is that a conforma-
tional change in the subunits pulls or pushes on the
subunit allowing the latter to be exposed less or more
on the cytoplasmic side. This difference might be
sufficient to cause a conformational change in the
cytoplasmic domain of the subunit. A third possibility
involves activation by a twisting mechanism as proposed
for the aspartate receptor (Fig. 11-8).
A large part of the cytoplasmic domain of the insulin
569
119
312
428
L2
717
731
736
930
952
981
1282
1355
C
Tyrosine
kinase
domain
Cytosol
ATP binding
Plasma
membrane
Arg-Lys-Arg
Tyr 972
1003 GXGXXG
Ser 1090
Tyr 1162, 1158, 1163
Tyr 1328, 1334
C
N

Cysteine-rich
domain
N N

1
Phe 34
Leu 87
L1
155
S S
S S
S
S S
S
Asp
707
C C
Exon 11
N
G. Insulin and Related Growth-Regulating Hormones
Figure 11-11 Schematic drawings of the insulin receptor.
(A) Typical textbook drawing. The bars represent the extended
peptide chains with positions of domain boundaries and chain
ends mapped (left side). Locations of some residues of special
interest are marked on the right side. (B) A fanciful but more
realistic picture of the receptor. See Fabry et al.
367
C C
S S
S S
Cysteine
rich

L
2
L
1
Cysteine
rich
L
1
L
2

Insulin

Tyrosine
kinase

S
S S
S
A B
receptor consists of a tyrosine-specific protein kinase
whose three-dimensional structure is depicted in Fig.
11-12.
365,365a
Not only can it phosphorylate OH groups
on tyrosine side chains of other proteins
362
but also it
catalyzes ATP-dependent autophosphorylation of
several residues in the C-terminal region. The most
readily phosphorylated residue is Tyr 1158.
366
Some
diabetic individuals have receptors with impaired
tyrosine kinase activity.
362
Using directed mutation of the cloned receptor
gene, Lys 1018 in the ATP-binding part of the tyrosine
kinase domain was replaced by alanine. This caused a
loss both of kinase activity and of biologic response to
insulin.
362
Thus, both the tyrosine kinase activity and
autophosphorylation appear essential. If so, aggrega-
tion of two or more receptors may increase the extent
of autophosphorylation and initiate a response.
Studies of many mutant proteins indicate that
insulin binds to the chains of the receptor between
the two domains labeled L1 and L2 in Fig. 11-11.
368
Among essential residues is Phe 39 (marked).
369
Insulin
contains three disulfide linkages (Fig. 7-16) and might
undergo a thiol disulfide exchange reaction (Eq. 11-7)
with SH groups present in a cytoplasmic cysteine-rich
domain of the subunit of the receptor or with an external
thiol compound.
369a,b
Such an exchange may also be
essential for activation of the receptor.
370
3. A Second Messenger for Insulin?
The known regulatory effects of insulin (Table 17-3)
often involve phosphorylation of serine or threonine
side chains on specific proteins. The tyrosine kinase of
the activated insulin receptors does not catalyze such
phosphorylation. Therefore, it seems likely that one
or more second messengers or mediator substances
are needed. Much effort has gone into searching for
570 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Figure 11-12 (A) Stereoscopic view of an -carbon trace of the insulin receptor kinase domain. Every tenth residue is marked
with a filled circle and every twentieth residue is labeled. (B) Locations of missense mutations in noninsulin-dependent
diabetes mellitus patients mapped onto the receptor kinase structure. The mutations are R993Q, G1008V, A1048D, R1164Q,
R1174Q, P1178L, W1193L, and W1200S. Here, R993Q is the mutant in which arginine 993 is replaced with glutamine, etc.
From Hubbard et al.
365
A B
substrates for the tyrosine kinase,
353,362,371,372
whose
activity on external protein substrates reaches a maxi-
mum after receptor tyrosines 1158, 1162, and 1163 have
been autophosphorylated. Phosphorylation of such
insulin receptor substrates (IRSs) as well as of
some smaller adapter proteins is thought to initiate
a series of complex cascades that serve to pass the
insulin signal on to a variety of sites of action within
a cell.
373 376
The large 185-kDA insulin receptor
substrate-1 (IRS-1), which is present in most cells,
is phosphorylated on several tyrosines, usually within
the sequences YXXM or YMXM. Phosphotyrosine
within such sequences is known to be a ligand for the
recognition domain SH2, which is present in many
proteins. Binding of SH2 domains of other proteins to
IRS-1 allows the insulin signal to be passed to several
different proteins in the branched signaling pathways.
Other insulin receptor substrates include IRS-2,
376a,b
protein Gab-1
376c
, and the smaller protein called Shc,
which occurs as 46-, 52-, and 66-kDa isoforms and is
discussed further on p. 568.
376,377
Shc is an adapter
protein which forms a complex that triggers the activa-
tion of the G protein Ras and the MAP kinase cascade
shown in Fig. 11-13.
377,378
This pathway (see Section
H,2) is thought to mediate the mitogenic (growth pro-
moting) effects of insulin and also to promote phospho-
rylation of serine and threonine side chains of many
proteins in the cytoplasm, the cytoskeleton, ribosomes,
membranes, and the nucleus.
379,380
One serine kinase
specifically phosphorylates the insulin receptor on
serine 1078.
381
In addition to IRS-1, IRS-2, and Shc, there are
additional adapter proteins that interact with the
phosphorylated tyrosine kinase domain of the insulin
receptor.
382
Furthermore, the receptor tyrosine kinase
may catalyze direct phosphorylation of some proteins,
e.g. a cytoplasmic loop of the
2
adrenergic receptor
(Fig. 11-6), without intervention of an adapter.
383
Phosphorylated IRS-1 activates a second signaling
pathway by interacting with an 85-kDa SH2-containing
protein that is a subunit of phophatidylinositol 3-
kinase.
384 386
This activates the 110-kDa catalytic
subunit of the 3-kinase, which catalyzes formation of
phosphatidylinositol 3-phosphate as well as PtdIns
(3,4)P
2
and PtdIns (3,4,5)P
3
.
387,387a
These compounds,
which remain within membranes, activate other
branches of the signaling cascade, some of which
may converge with those of the MAP kinase cascade.
However, there appears to be specific activation of a
ribosomal Ser/Thr kinase that, among other activities,
phosphorylates ribosomal protein S6, a component of
the small ribosomal subunit.
388
It also phosphorylates
some isoforms of protein kinase C and other enzymes.
PtdIns 3-kinase may also activate 6-phosphofructo-2-
kinase (Fig. 11-2, step d).
384,388
One of the most important effects of insulin is to
increase glucose uptake by cells.
373,389
The mechanism
is thought to depend upon the transporter protein
GLUT4, which is stored within the membranes of
small cytoplasmic vesicles. Binding of insulin to its
receptors induces movement of these vesicles to the
plasma membrane where fusion with the plasma
membrane makes the GLUT4 molecules available for
glucose transport.
390
Phosphatidylinositol 3-kinase
also plays an important role. The PtdIns(3,4)P
2
and
PtdIns(3,4,5)P
3
generated by this enzyme (Fig. 11-9)
571 H. Growth Factors, Oncogenes, and the Cell Cycle
remain in the membrane and provide sites to which
various proteins, e.g., those containing PH domains
(Chapter 7) may bind. Among the proteins recruited
to the inner membrane surface in this way is a Ser/Thr
protein kinase known as PKB/Akt. The Akt abbre-
viation refers to its relationship to a particular viral
oncogene.
390a 390e
and the phosphatidyl-inositol deriv-
atives generated by the 3-kinase may have a direct
effect upon the exocytosis of the GLUT-4 containing
vesicles as may Gq-coupled receptors.
391
Although insulin unquestionably stimulates phos-
phorylation of many proteins, the major metabolic effect
appears to result from dephosphorylation by phosphatases
of phosphorylated forms of several enzymes.
379,392
These include glycogen synthase and pyruvate
dehydrogenase, which are both activated by dephos-
phorylation, and glycogen phosphorylase
393
and
hormone-sensitive lipase, which are deactivated by
dephosphorylation. These changes result in increased
synthesis and storage of both glycogen and triacyglyc-
erols. Insulin also stimulates a membrane-bound cAMP
phosphodiesterase causing a reduction in cAMP con-
centrations.
394
Insulin injected into the human body has a half-
life of only 5 10 minutes. Much of the hormone is
destroyed by hydrolytic cleavage of the peptide chain
by a non-lysosomal protease.
395 397
The receptors
together with the hormone are taken into cells by endo-
cytosis from clathrin-coated pits and both may be
degraded in the lysosomes.
396,398
Some signaling may
arise from insulin receptors in endosomes.
399
Some of
the hormone enters the nucleus, perhaps still bound
to the receptors,
400
and may have a direct effect upon
gene transcription. Some receptors are recycled to the
cell surface.
398
This latter process, together with that
of new receptor synthesis, controls the number of
receptors on the surface and, in turn, the sensitivity of
the cell to insulin. This is only one mechanism used for
desensitization or downregulation of receptor sensi-
tivity and numbers, something that happens normally
for any hormone when its concentration is high.
401,402
H. Growth Factors, Oncogenes, and the Cell
Cycle
Insulin is just one of a large number of proteins
that are secreted by cells and which influence the
growth of nearby cells.
403 405
For example, blood
platelets, which aggregate at the site of an injury to
a blood vessel, contain granules ( granules) which,
when the platelet is activated (see Box 8-A) release
platelet-derived growth factor (PDGF), a 28- to 31-
kDa glycosylated peptide which stimulates growth and
tissue repair in the injured region.
406,407
There are many
other protein growth factors, a few of which are con-
sidered here. Others are discussed in Chapters 3032.
1. Oncogenes and Proto-oncogenes
Studies of cancer-causing cellular oncogenes and
proto-oncogenes (c-onc) identified in the human
genome
408411
have contributed greatly to our under-
standing of the action of growth factors. Proto-onco-
genes are segments of DNA that code for proteins that
have a normal function but which may become acti-
vated by a mutation or by chromosomal rearrange-
ment to become cancer-causing oncogenes (Box 11-D).
Oncogenes were discovered first in oncogenic (cancer-
causing) retroviruses,
412
where they are designated
v-onc. The v-onc genes are usually very similar but
not identical to the corresponding c-onc genes and are
thought to have arisen from them. The v-onc genes are
often incomplete or have become fused to other genes
by genetic recombination. Considerable excitement
has attended the discovery that many solid human
tumors contain activated oncogenes,
413
several of which
are listed in Table 11-3.
In 1983 it was found that the oncogene sis, which is
carried by the simian sarcoma virus, has a 104-residue
sequence nearly identical to that of human PDGF.
404,414
The PDGF receptor has a large extracellular domain
consisting of five immunoglobulin-like domains. Like
the insulin receptor, it has tyrosine kinase activity which
resides in a C-terminal cytoplasmic domain.
406,407,415
This suggests that the malignant transformation of
cells by the viral v-sis gene leads to an excessive pro-
duction of normal PDGF and, consequently, to exces-
sive growth. The sequences related to v-sis are often
found in human tumors and are located on chromo-
some 22.
416
Rearrangements by which part of this
chromosome is moved to another location are well
known and sometimes lead to conversion of the proto-
oncogene to an active cancer-inducing oncogene.
The avian v-erbB oncogene and oncogene neu
417- 419
both have sequences homologous to that of the gene
for the receptor for the 53-residue epidermal growth
factor EGF.
420423a
The corresponding cellular c-erbB
is the gene for the EGF receptor, a large 170-kDa 1186-
residue protein. It also resembles the insulin receptor
in having an N-terminal domain outside the cell, a
single hydrophobic helix that spans the membrane,
and a cytoplasmic domain with tyrosine-specific pro-
tein kinase activity.
403,419,424,425
It differs from the
insulin receptor in being a single peptide chain. How-
ever, when EGF binds, the receptor dimerizes and the
protein kinase is activated.
426
It phosphorylates its
own Tyr 1173 near the C terminus, as well as tyrosines
in the lipocortins, 36- to 38-kDa calcium-binding proteins
located on the cytosolic face of plasma membranes.
427
The activated receptor also stimulates phospholipase
C with a resultant increase in concentrations of inositol
trisphosphate and Ca
2+
(see Fig. 11-13).
428
The active
oncogene v-erbB encodes a major fragment of the EGF
receptor. However, most of the N-terminal domain
572 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
TABLE 11-3
A Few Oncogenes That Have Interested Biochemists
Oncogene
Symbol Source Properties
v-sis Simian sarcoma virus Gene product is closely related to the B-chain of platelet-derived
growth factor
v-erbB Avian erythroblastosis virus Gene product is shortened version of the EGF receptor, a
tyrosine kinase
neu (Her-2) Rat neuroblastomas Homologous to EGF receptor; has tyrosine
A similar gene is found in kinase activity; may control
human breast cancers and phosphotidylinositol 3-kinase
adenocarcinomas
v-src Rous avian sarcoma virus Gene product is another tyrosine-specific
protein kinase
abl Chronic myelogenous leukemia Similar to src; characteristic chromosomal
translocations yield cancer
ras, has, Human bladder, colon, Homology with G
s
and G
i
regulatory
v-H-ras lung carcinomas proteins; several human proto-oncogenes
v-K-ras exist; a single mutation in H-ras may lead
N-ras to cancer
bas, kis Murine sarcoma viruses
v-myc Burkitts lymphoma, Nuclear location for gene product; which
mouse plasmacytomas, forms complex with protein Jun
avian retrovirus MC29
mos Burkitts lymphoma,
mouse plasmacytomas
v-fos Osteosarcoma virus (mouse) Nuclear location for gene product, which forms complex with
protein Jun
v-jun Protein product is subunit of transcription factor AP-1
bcl1 Cyclin D1
including the EGF binding site is missing and the C-
terminal end has been shortened. The tyrosine kinase
domain is intact but Tyr 1173 is missing.
The rat neu oncogene (also called erbB-2 and HER-2),
which apparently is derived from the gene for another
growth factor receptor, differs from normal neu by a
single nucleotide. This change causes valine to be
substituted for glutamic acid at position 664 in the
membrane-spanning domain of the 185-kDa protein.
418
This evidently gives an overactive and perhaps uncon-
trolled tyrosine-specific protein kinase. The c-erbA
proto-oncogene appears to be the nuclear receptor for
the thyroid hromone triiodothyronine (Chapter 25).
The Rous sarcoma virus oncogene v-src and a
family of related oncogenes are derived from protein
tyrosine kinases that are attached with the aid of a
myristoyl anchor to the inner, cytoplasmic surfaces
of membranes.
429431
They may be activated by inter-
action with an occupied surface receptor. It has been
difficult to understand the functioning of the normal
Src protein. However, inactivation of c-src in mice
caused the serious bone disease osteopetrosis in which
the osteoclasts fail to function properly in resorbing
the bone matrix, thereby allowing excessive accumula-
tion of calcium phosphate.
432,433
A c-src deficiency also
decreases formation of the bone adhesion protein
osteopontin, an RGD protein.
434
There is actually a
family of src proteins, some of which have important
functions in lymphocytes.
435438
The gene for one of
these is mutated in the cell disorder
agammaglobulinemia.
437
Another oncogene derived from a tyrosine kinase
573
BOX 11-D CANCER
Although cancer occurs in about 200 clinically
distinct types, most cancers can be classified into four
categories. In leukemias, which account for 3% of the
~700,000 cases of cancer diagnosed per year in the
United States, an abnormal number of leukocytes are
produced by the bone marrow. Lymphomas, such as
Hodgkins disease and Burkitts lymphoma, arise from
lymphocytes. They account for ~5% of human cancers.
In these diseases malignant cells are produced in the
spleen and lymph nodes and sometimes aggregate in
lymphoid tissues. Sarcomas, solid tumors of bone or
other connective tissue, contribute ~2% to the total of
human cancers, while carcinomas, cancers of epithelial
tissue, account for 85%. Carcinomas may develop from
either the external or the internal epithelia, including
the glands, lungs, and nerves.
a d
More than one-third
of all cancers in the United States are nonmelanoma
carcinomas of the skin;
e
for these the mortality rate is
low. Lung, colorectal, breast, and prostate tumors
account for 55% of cancer deaths.
c
Carcinomas pre-
dominate in humans, but lymphomas, leukemias,
and sarcomas are much more prevalent in laboratory
animals and fowl.
An important characteristic of cancer cells is their
uncontrolled proliferation. They dont respond to the
normal signals from adjacent cells that indicate that
cell division should stop. Cancer cells also differ
dramatically from those present in warts and other
benign tumors and in psoriasis. These conditions also
result in excessive proliferation of cells and partial
derangement of normal regulatory processes.
A second characteristic of cancer cells is that they
usually appear less differentiated than the tissues from
which they arise and are more like embryonic cells.
Many cancers produce ectopic proteins, proteins
inappropriate to the tissue involved and often identical
to proteins synthesized by embryonic or fetal cells. A
well-known example is -fetoprotein, a 72-kDa
glycoprotein normally present in serum in almost
undetectable amounts but present in large amounts when
some types of cancer are present.
f
A third property of
cancers is the tendency toward metastasis, the detach-
ment of cells from the cancer and their development in
distant parts of the body.
g i
Cancer cells dont grow any faster than normal cells
but they continue to divide when normal cells would
not. For this reason, a cancer can grow rapidly and its
demands for nutrients can literally starve the host.
Cancer tends to weaken the immune system, making
the host more susceptible to infections. In addition,
cancers often interfere directly with the functioning of
various organs and may cause death in this way.
What initiates a cancer? We know that cancers can
arise from only one or a very small number of cells and
that cancer can be induced by carcinogenic chemical
compounds, by certain viruses, and by radiation. Use
of tobacco appears to be responsible for about 30% of
all human tumors.
c,j
Diet also affects the likelihood of
developing cancer. For example, diets containing less
animal fat and more fruits and vegetables are associated
with lower levels of colon cancer.
c,k
Many carcinogenic
compounds are naturally present in foods. Genetic
factors help to determine susceptibility to cancer
b,l
and
characteristic chromosomal aberrations are usually
associated with cancer.
m o
The incidence of cancer
increases markedly with age.
A common feature of all the agents that induce
cancer is the production of mutations and cancer
probably always involves some alteration in the cells
DNA. The long lag between exposure to carcinogenic
materials and development of cancers, often 20 years
or more, suggested that more than one mutation or
chromosomal rearrangement is required for produc-
tion of a cancer. Recent evidence confirms that several
mutations are required.
b,p
Relevant to this conclusion
is the fact that carcinogenic compounds can be applied
to the skin in amounts sufficient to cause a number
of mutations in the epithelial cells but insufficient to
actually induce cancer. Then, even many years later,
irritant compounds known as cancer promoters can
be applied and cancer will develop promptly. The
promoters apparently induce cell proliferation which
leads to more errors in DNA replication, converting
an initially mutated cell to a cancerous cell. The most
studied promoters are the phorbol esters, which are
known to activate the protein kinase C isoenzymes
(Section E,2). Any factors that increase rates of cell
division such as some hormones, excess calories, or
chronic inflammation cause increased cancer.
c
Cell
divisions are accompanied by errors in replication of
DNA and sometimes by translocation or deletion of
parts of chromosomes. Chronic infection by bacteria,
viruses, or other organisms may cause cancer as a
result of continuing inflammation. Other cancers arise
from integration of viral DNA into the hosts DNA.
Virally induced cancers are often epidemic among
poultry and rodents. When infected with cancer-
causing viruses from these animals, cells in culture
often become transformed. Whereas normal cells
tend to respond to contact inhibition and grow as a
monolayer, transformed cells continue to divide after
the monolayer is complete. In laboratory studies,
transformation of cells is often taken as the equivalent
to an early step in cancer production in an animal.
Studies of transformation led to the identification and
characterization of several viral oncogenes which
cause the transformation. These are designated by
abbreviations such as v-src (the oncogene of Rous
sarcoma virus, which induces cancer in chickens) and
v-sis (the oncogene of simian sarcoma virus, which
causes cancers in monkeys). Some other oncogenes are
described in the main text. Naturally occurring gene
H. Growth Factors, Oncogenes, and the Cell Cycle
574 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
BOX 11-D CANCER (continued)
sequences closely homologous to those of the viral
oncogenes have been found in many solid human tumors.
Study of oncogenes has shown that they are related to and
derived from proto-oncogenes, normal cellular genes
that are involved in control of growth and differentiation.
Oncogenes are often amplified in tumor cells
so that their copy number is greater than that of the
corresponding genes in normal cells. For example an
oncogene related to the viral oncogene neu is amplified
in many human breast and ovarian cancers
q
and onco-
gene src in many colon cancers.
r
Mutated ras genes
have been found in over one-third of human colorectal
cancers.
s
Amplified oncogenes ras, myc, and myb have
been observed in other cancers.
t
Oncogenes are often
overexpressed or are responsible for over-expression
of other genes. One idea that developed from these
observations is that cancer cells may secrete new or
mutated growth factors that stimulate their own recep-
tors (autocrine stimulation) in a way that promotes
uncontrolled growth.
u
Cancer develops in stages and in many cases defec-
tive proto-oncogenes appear before truly malignant cells
appear. The latter must result from additional mutations
that often involve loss of parts of chromosomes. A major
breakthrough in our understanding of cancer and how
it is induced by loss of genes has come from studies of
some rare cancers that are inherited in a Medelian fashion.
One of these is retinoblastoma, an intraocular tumor
which affects 1 child in 20,000. Homozygotes always
develop the disease between the ages of 1 and 5. The
hereditary defect has been traced to the absence of a
functional retinoblastoma gene RB1, which is found in
band q14 of chromosome 13.
v
Additional mutational
events are required to induce cancer. The RB1 gene
was the first tumor-suppressor gene (anti-oncogene)
identified. These suppressor genes encode proteins
that inhibit growth.
w z
The retinoblastoma gene
encodes a 105-kDa DNA-binding phosphoprotein
(Rb-P).
aa bb
The Rb protein is phosphorylated and
dephosphorylated in a cyclic fashion that is synchro-
nized with the cell replication cycle (Fig. 11-15). It
forms a complex with a transcription factor E2F that
functions in transcription of the adenovirus genes and
is also involved in control of the cell replication cycle.
cc
Deletion of the Rb gene from mice leads to death of
embryos homozygous for the mutation.
dd
Study of other rare hereditary cancers has led to
the location of 20 or more additional probable tumor-
suppressor genes. One of these, p53, is inactive in over
50% of all human cancers and over 90% of sqamous
cell carcinomas of the skin.
ee
In small-cell lung cancers
and osteosarcoma both RB and p53 are inactive.
z
Protein
p53 is a stronger tumor suppressor than protein Rb.
Results of a variety of experiments have suggested
that p53, a DNA-binding protein of known structure,
ff
plays a key role in checking DNA for damage at the G
1
to S-phase checkpoint in the cell cycle. If the DNA has
too many defects the cycle is stopped in the G
1
stage
and the cell may be killed by the process known as
apoptosis.
gg jj
Protein p53 has been called the
guardian of the genome. The mechanisms by which
it functions are complex and poorly understood. It
may act with the assistance of Rb and many other
proteins. Mutations in DNA and their repair are dis-
cussed in Chapter 27 and the cell cycle is discussed in
this chapter and further in Chapters 26 and 32.
Many other cancer susceptibility genes also encode
suppressors. Mutation in genes BRACA1
kk
and BRACA2
ll
are responsible for early onset ovarian and breast
cancer. Gene DPC4 may be a suppressor of pancreatic
cancer.
mm
The gene ptc (patched), first studied as a
developmental gene in Drosophila, may encode a sup-
pressor of basal cell carcinoma, the commonest form
of human skin cancer.
nn
Gene p16 (also called CDKN2)
may be a major suppressor that is mutated in many
cancers including the dangerous skin melanoma.
oo
Mutations in the NF gene, which may be a cytoskeletal
protein, are associated with neurofibromatosis,
pp
a
relatively common hereditary disease causing tumors
of the nervous system. The tumors are usually not
malignant but are numerous and disfiguring. Several
cancer susceptibility genes are associated with faulty
mismatch repair of DNA. Among these is the APC
gene, whose malfunction is associated with human
familial adenomatous polyposis which causes
thousands of benign tumors in the lining of the large
intestine and often colorectal cancer.
qq,rr,ss
In the much
more common nonpolyposis colon cancer a complex
pattern of instability in several genes is associated with
DNA repair,
tt
a topic dealt with further in Chapter 27.
The ATM gene defective in ataxia telangiectasia
(Chapter 27) may encode a phosphatidylinositol kinase
that is in some way involved in repair of DNA.
ss,uu,vv
A transcription factor gene nm23 may be a suppressor
gene for metastasis
ww
and the cell cell adhesion
molecule E-cadherin may suppress tumor invasion in
some kinds of breast cancer.
xx
The VHL (van Hippel-
Lindau cancer syndrome) suppressor protein is defec-
tive in the majority of kidney cancers. It normally
binds to the elongin complex, a DNA-binding com-
plex that functions in control of transcription.
zz
In addition to treatment by surgery there are
numerous chemical approaches to combating cancer.
xy
They usually exploit the tendency of cancers to grow
continuously. For example, a toxic analog of a metabo-
lite needed for growth, such as methotrexate (Chapter
16) and 5-fluorouridine (Box 28-C) may be taken up
more rapidly by tumor cells than by normal cells. A
variety of DNA-binding compounds are useful in chemo-
therapy (Box 5-B). Alkylating agents such as the nitrogen
mustards (Eq. 5-20) and certain antibiotic compounds
are also used widely, as are intercalating compounds
575
BOX 11-D (continued)
a
Cairns, J. (1978) Cancer: Science and Society, Freeman, San
Francisco, California
b
Cavenee, W. K., and White, R. L. (1995) Sci. Am. 272(Mar),
7279
c
Ames, B. N., Gold, L. S., and Willett, W. C. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 52585265
d
Ruddon, R. W. (1987) Cancer Biology, 2nd ed., Oxford Univ.
Press, London
e
Preston, D. S., and Stern, R. S. (1992) N. Engl. J. Med. 327,
16491662
f
Zhang, D., Hoyt, P. R., and Papaconstantinou, J. (1990) J. Biol.
Chem. 265, 33823391
g
Packard, B. (1986) Trends Biochem. Sci. 11, 490491
h
Feldman, M., and Eisenbach, L. (1988) Sci. Am. 259(Nov), 6085
i
Marx, J. (1993) Science 259, 626629
j
zur Hausen, H. (1991) Science 254, 11671173
k
Willett, W. (1989) Nature (London) 338, 389394
l
Dragani, T. A., Canzian, F., and Pierotti, M. A. (1996) FASEB J.
10, 865870
m
Solomon, E., Borrow, J., and Goddard, A. D. (1991) Science 254,
11531160
n
Nowell, P. C. (1994) FASEB J. 8, 408413
o
Pennisi, E. (1996) Science 272, 649
p
Marx, J. (1989) Science 246, 13861388
q
Slamon, D. J., Godolphin, W., Jones, L. A., Holt, J. A., Wong, S.
G., Keith, D. E., Levin, W. J., Stuart, S. G., Udove, J., Ullrich, A.,
and Press, M. F. (1989) Science 244, 707712
r
Cartwright, C. A., Meisler, A. I., and Eckhart, W. (1990) Proc.
Natl. Acad. Sci. U.S.A. 87, 558562
s
Bos, J. L., Fearon, E. R., Hamilton, S. R., Verlaan-de Vries, M.,
van Boom, J. H., van der Eb, A. J., and Vogelstein, B. (1987)
Nature (London) 327, 293297
t
Yokota, J., Tsunetsugu-Yokota, Y., Battifora, H., Le Fevre, C.,
and Cline, M. J. (1986) Science 231, 261265
u
Sporn, M. B., and Roberts, A. B. (1985) Nature (London) 313,
745747
v
Weinberg, R. A. (1990) Trends Biochem. Sci. 15, 199202
w
Stanbridge, E. J. (1990) Science 247, 1213
x
Weinberg, R. A. (1991) Science 254, 11381146
y
Knudson, A. G. (1993) Proc. Natl. Acad. Sci. U.S.A. 90, 10914
10921
z
Yokota, J., and Sugimura, T. (1993) FASEB J. 7, 920925
aa
Wiman, K. G. (1993) FASEB J. 7, 841845
bb
Cobrinik, D., Dowdy, S. F., Hinds, P. W., Mittnacht, S., and
Weinberg, R. A. (1992) Trends Biochem. Sci. 17, 312315
cc
Nevins, J. R. (1992) Science 258, 424429
dd
Jacks, T., Fazeli, A., Schmitt, E. M., Bronson, R. T., Goodell, M.
A., and Weinberg, R. A. (1992) Nature (London) 359, 295300
ee
Ziegler, A., Jonason, A. S., Leffell, D. J., Simon, J. A., Sharma,
H. W., Kimmelman, J., Remington, L., Jacks, T., and Brash, D.
E. (1994) Nature (London) 372, 773776
ff
Cho, Y., Gorina, S., Jeffrey, P. D., and Pavletich, N. P. (1994)
Science 265, 346355
gg
Marx, J. (1993) Science 262, 16441645
hh
Enoch, T., and Norbury, C. (1995) Trends Biochem. Sci. 20,
426430
ii
Kaufmann, W. K., and Paules, R. S. (1996) FASEB J. 10,
238247
jj
Hartwell, L. H., and Kastan, M. B. (1994) Science 266, 18211828
kk
Futreal, P. A., and 26 other authors (1994) Science 266, 120122
ll
Wooster, R., and 30 other authors (1994) Science 265, 20882090
mm
Hahn, S. A., Schutte, M., Hoque, A. T. M. S., Moskaluk, C. A.,
da Costa, L. T., Rozenblum, E., Weinstein, C. L., Fischer, A.,
Yeo, C. J., Hruban, R. H., and Kern, S. (1996) Science 271,
350353
nn
Pennisi, E. (1996) Science 272, 15831584
oo
Marx, J. (1994) Science 265, 13641365
pp
Rouleau, G. A., and 20 other authors (1993) Nature (London)
363, 515521
qq
Peltomki, P., Aaltonen, L. A., Sistonen, P., Pylkknen, L.,
Mecklin, J.-P., Jrvinen, H., Green, J. S., Jass, J. R., Weber, J. L.,
Leach, F. S., Petersen, G. M., Hamilton, S. R., de la Chapelle,A.,
and Vogelstein, B. (1993) Science 260, 810819
rr
Huang, J., Papadopoulos, N., McKinley, A. J., Farrington, S.
M., Curtis, L. J., Wyllie, A. H., Zheng, S., Willson, J. K. V.,
Markowitz, S. D., Morin, P., Kinzler, K. W., Vogelstein, B., and
Dunlop, M. G. (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 9049
9054
ss
Kolodner, R. D. (1995) Trends Biochem. Sci. 20, 397401
tt
Papadopoulos, N., and 19 other authors (1994) Science 18,
16251629
uu
Keith, C. T., and Schreiber, S. L. (1995) Science 270, 5051
vv
Sanchez, Y., Desany, B. A., Jones, W. J., Liu, Q., Wang, B., and
Elledge, S. J. (1996) Science 271, 357360
ww
Marx, J. (1993) Science 261, 428429
xx
Berx, G., Cleton-Jansen, A.-M., Nollet, F., de Leeuw, W. J. F.,
van de Vijver, M. J., Cornelisse, C., and van Roy, F. (1995)
EMBO J. 14, 61076115
xy
Hoffman, E. J. (1999) Cancer and the Search for Selective
Biochemical Inhibitors, CRC Press, Boca Raton, Florida
yy
Henderson, B. E., Ross, R. K., and Pike, M. C. (1993) Science
259, 633638
yz
Young, R. C. (2000) Nature (London) 408, 141
zz
Stebbins, C. E., Kaelin, W. G., Jr., and Pavletich, N. P. (1999)
Science 284, 455461
such as adriamycin (14-hydroxydaunomycin; Fig. 5-22)
and cisplatin (Box 5-B). A disadvantage to most present-
day chemotherapy is that normal proliferation of cells,
especially of glandular tissues, intestinal epithelium,
hair, etc., is severely damaged. A possibility for circum-
venting this problem is to put normal growth on hold
temporarily while cancer growth is being inhibited.
Some inhibitors of topoisomerase I (see Chapter 27)
have low toxicity and could be useful. Another thera-
peutic approach is based on the fact that one function
of the immune system is to destroy cancerous or pre-
cancerous cells. The immune system tends to weaken
with age, which may be one reason that the incidence
of cancers rises rapidly in older age Are there ways of
stimulating the immune system into increased activity
against cancer cells? Another approach is to find ways
of increasing the activity of tumor-suppressor genes.
Can drugs be developed to prevent cancer? Such
chemoprevention
yz
may be appropriate for persons
carrying genes that make them highly susceptible to
cancer. The antiestrogenic drug tamoxifen (Chapter
22) is currently being tested on women with a high risk
for breast cancer. Use of oral contraceptives appears to
have cut the risk of endometrial cancer substantially.
yy
Newer approaches to contraception may help prevent
breast cancer and chemoprevention may also be possi-
ble for prostate cancer.
yy
H. Growth Factors, Oncogenes, and the Cell Cycle
576 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
gene is the Abelson murine leukemia virus v-abl.
438a
In the mouse the c-abl gene is split into at least ten
exons. However, v-abl contains all of these as a correctly
spliced sequence suggesting that v-abl was derived
from a c-abl messenger RNA. Part of one exon of the
cellular gene is missing in v-abl and there is at least one
base substitution mutation as well. Human c-abl is
located on the long arm q of chromosome 9. This arm
has long been known to be translocated to chromosome
22 (the Philadelphia translocation) in patients with
chronic myelogenous leukemia. There the c-abl gene
is fused with another gene.
439
The human c-sis gene
is also located in the region of chromosome 22 that is
translocated to chromosome 9 in the same patients.
A group of human pituitary tumors have been
shown to contain an oncogene that is apparently a
mutated gene for the stimulatory protein G
s
.
440
This
is the protein that activates adenylate cyclase in response
to hormonal activation. The oncogenic mutations inhibit
the GTPase activity that normally turns off this activa-
tion. These tumors secrete growth hormone which
binds to receptors on the tumor cells activating the
defective G
s
proteins and causing excessive synthesis
of cAMP. This in turn promotes growth of the tumor.
The ras oncogenes. Activated ras-oncogenes
have been found in at least 25% of all human tumors.
The proto-oncogenes, which are designated c-H-ras,
c-K-ras, and c-N-ras, are found on the short arms of
chromosomes 11, 12, and 1, respectively. A single base
substitution (G T) at position 35 of any of the genes,
resulting in a Ras protein containing valine instead of
glycine at position 12 of the 21 kDa protein product
(usually designated p21) produces an active oncogene.
Substitutions at position 13 or at positions 59 and 61,
which are adjacent to Gly 12 in the three-dimensional
structure, can also activate the oncogenes.
441
From the
drawing in Fig. 11-7A the locations of glycines 12 and
13 and of residues 59/61 are seen to be close to the
phospho group of bound GTP. Proteins encoded by
activated ras oncogenes are less active in catalyzing
GTP hydrolysis than are the corresponding normal
proteins. In addition, the GTPase-activating protein
(GAP) that binds to normal ras proteins and stimulates
their GTPase activity does not affect the mutant onco-
genic proteins.
A single-base alteration is capable of activating a
ras gene with respect to cell transformation. However,
initiation of a malignant tumor requires the additional
presence of at least a second activated oncogene such
as myc
442,443
or fos
444
or previous transformation of a
fibroblast into a nonmalignant but immortalized
form by treatment with carcinogens. The c-myc gene,
which is found in active form in plasmacytomas (tumors
of B lymphocytes) of mice as well as in the human
Burkitts lymphoma, is normally located on human
chromosome 8. In most Burkitts lymphomas a trans-
location has brought the c-myc gene into the locus of
the immunoglobulin heavy chains on chromosome 14.
There its transcription may be subject to different
controls than in its original location.
445,446
During the
translocation process the c-myc gene is often broken
within the first intron. Thus, the activated gene lacks
the first exon and is placed after a new controlling
sequence that may drastically alter its transcription
rate. Viral myc genes are found in at least one human
virus, cytomegalovirus, which has been associated
with carcinomas.
Transcription factors. The proto-oncogenes
c-myc
447451a
, c-myb,
452454
c-fos, c-jun and c-ets
455
all
encode nuclear proteins involved in regulation of
transcription. The 39 kDa protein Jun, which is encoded
by c-jun, is a major component of the transcriptional
activator called AP-1.
456459
It binds to palindromic
enhancer sites (Chapter 28) in DNA promoters to
increase the transcription rate for a group of genes.
Jun is actually a multigene family whose encoded
proteins bind to DNA as complexes formed with the
62 kDa phosphoprotein Fos, the product of the c-fos
gene.
460
The heterodimeric Fos/Jun complex is held
together, at least in part, by interactions between leucine
side chains lying along a pair of parallel -helices in a
leucine zipper (Fig. 5-36; Fig. 2-21).
461,462
Regulation of the synthesis of Jun is complex, but
growth factors such as Neu, EGF,
463
and PDGF stimu-
late transcription of c-jun in cultured cells. Messenger
RNA for synthesis of Fos appears within a few minutes
of stimulation of PDGF receptors.
464,465
This is one of
the earliest known nuclear reactions to a mitogenic
stimulus and suggested that a Ras p21 protein is
involved in stimulating transcription of c-fos in the
signaling pathway from PDGF.
466
Synthesis of Fos is
also induced by a variety of other stimuli.
456
Upon
translocation into the nucleus Fos combines with pre-
existing Jun to form AP-1, which binds to sites on
DNA and induces the transcription of a large number
of proteins (Chapter 28). Deletion of the c-fos gene in
mice leads to defects in developing bone, teeth, and
blood cells,
467
while excessive synthesis of Fos has
been associated with the human bone disease fibrous
dysplasia.
468
2. The MAP Kinase Cascade
Insulin, platelet-derived growth factor (PDGF),
epidermal growth factor (EGF), and many other pro-
teins have mitogenic activity, that is, they induce
cells to transcribe genes, to grow, and to divide. How
is this accomplished? Study of such oncogenes as src
and ras suggested that the proteins that they encode
also participate in the process, as do transcription
factors, including those encoded by the proto-oncogenes
577
AP-1 A major transcriptional activator
protein
ARK Beta adrenergic receptor kinase
EGF Epidermal growth factor
EGFR Epidermal growth factor receptor
ERK-1, ERK-2 Extracellular signal-regulated protein
kinases (proline-directed protein
kinases)
Fos Protein encoded by proto-oncogene fos
(Table 11-3)
Grb2 Adapter protein containing one SH2
and two SH3 domains
IGF-1 Insulin-like growth factor 1
InsRTK Insulin receptor tyrosine kinase
InsP
3
or IP
3
Inositol (1,3,5)-trisphosphate
Jun Protein encoded by proto-oncogene
jun, an AP-1 gene
MAPK Mitogen-activated protein kinase
(also designated ERK)
MAPKK A kinase acting on MAPK
MEK-1,MEK-2 Mitogen-activated ERK-activating
kinases; dual function Ser/Thr and
Tyr protein kinases; also called
MAP kinase kinases
c-myc A cellular proto-oncogene
PDGF Platelet-derived growth factor
PtdIns (or PI) Phosphatidylinositol 3-kinase
3-kinase
PKA (cAPK) cyclic AMP-dependent protein kinase
Table 11-4
A Few Abbreviations Used in Discussions of Cell Signaling
H. Growth Factors, Oncogenes, and the Cell Cycle
PKC, PKC, , Protein kinase C,, , subforms
PKR dsRNA-activated protein kinase
PLA Phospholipase A
PLC Phospholipase C
PP2A Protein phosphatase, type 2A
PTK Protein tyrosine kinase
PTP (PTPase, Protein tyrosine phosphatases
PTP 1B, etc.)
pY Phosphotyrosine residue
Raf-1 A cytoplasmic serine/threonine
protein kinase; also called MAP
kinase kinase kinase (MAP3K)
Ras A monomeric G protein encoded by
the proto-oncogene ras
RSK Ribosomal protein S6 kinase
RTK Receptor tyrosine kinase
RTK-P Phosphorylated receptor tyrosine
kinase
SH2, SH3 Src homology domains 2 and 3
Sos son of sevenless, a GDP GTP
exchange factor named for a
similarity to the protein encoded
by the Drosophila sevenless gene
Src Protein tyrosine kinase encoded by
oncogene src (Table 11-3); contains
recognition domains SH2 and SH3
V2R Type 2 vasopressin receptor
Shc A proline-rich adapter protein
containing SH2 and PH domains
myc, fos, and jun. The Src and Ras proteins are an-
chored on the inner surfaces of cytoplasmic mem-
branes. Although the exact functional relationships
of all of the components, one to another, has not been
completely established, a general picture, usually
described as the mitogen-activated protein kinase
(MAP kinase) cascade, has emerged.
69,380,469- 470b
This is sketched in simplified form in Fig. 11-13. The
many amplification steps provide for ultrasensitive
responses.
342
The binding of a hormone or growth factor (a
ligand) to a dimeric receptor activates the protein
kinase domain of the receptor which phosphorylates
a number of tyrosine hydroxyl groups of the receptor
itself. This autophosphorylation is followed by a
variety of events, which include phosphorylation of
tyrosine side chains of various other proteins.
426
An-
other major event is the binding of a variety of different
protein molecules containing recognition domains to the
phosphotyrosyl groups of the activated receptors.
471
The
major recognition motif is the SH2 domain. See Figs.
7-30 and 11-14.
472 475
Proteins containing SH2 domains
can bind to the phosphotyrosyl groups of the activated
receptors and while bound become phosphorylated by
the receptor tyrosine kinase action and/or be activated
allosterically.
Other proteins interact with the receptors indirectly
through adapter molecules which have no catalytic
activity. Two well-known adapter proteins are Grb2
476
and Shc.
477,478
The 25-kDa protein Grb2 consists entirely
of recognition domains, one SH2 and two SH3 domains
(Fig. 11-14). The larger Shc, which is found in all mam-
malian tissues, contains a 200-residue phosphotyrosyl-
binding PH domain (Chapter 7) at the N terminus, a
578 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
collagen-like domain that binds to Grb2 and an SH2
domain at the C terminus.
376,377,477
Adapter Grb2 binds to a phosphotyrosine side chain
of an activated receptor, such as that for EGF, and
simultaneously binds to the GDP-GTP exchange protein
called Sos (Table 11-4). This signals Sos to activate the
membrane-bound Ras protein by converting it into the
GTP form. The second adapter Shc may also participate
in formation of the receptor kinase - Grb2Sos com-
plex,
479
perhaps permitting formation of a more robust
complex that may receive signals from more than one
kind of receptor. Functions of other members of the
Grb adapter family are being discovered.
479a
Activated Ras binds to and activates the cytoplasmic
serine/threonine protein kinase called Raf-1.
380,480,481
This kinase becomes transiently activated within 2 3
min of the binding of a mitogen to a receptor. Raf-1
initiates a cascade of other protein kinases by acting on
the dual-function Ser/Thr and tyrosine protein kinases
called MEK-1 and MEK-2. The phosphorylated, active
MEK proteins phosphorylate the mitogen-activated
protein kinases MAPK which act on a variety of other
proteins. Two of the best known MAPK proteins are
designated ERK-1 and ERK-2. These are proline-
directed kinases which phosphorylate serines and
threonines that are neighbors to prolines, e.g. in the
sequence PLS/TP.
380
The activated ERKs are able to
phosphorylate a large number of different proteins
including nuclear proteins that control the transcription
of such protein transcription factors as AP-1 and Myc.
A protein known as the serum response factor binds
to nucleotide sequences CC(A/T)
6
GC in the DNA to
locate initiation sites for transcription. Other proteins
that have been phosphorylated by the MAP kinase
cascade then induce transcription.
482
The induction of
c-fos mRNA is one of the earliest identified responses
to growth factors.
375,456,483
Protein Jun, whose synthesis
is induced independently by almost all growth factors,
is usually present in excess.
The Fos/Jun complex is transcription factor AP-1,
which induces transcription of many genes needed
for cell growth. However, transcription of specific
genes often depends upon additional nucleotide
sequences. For example, the sequence CGGAAA is
present in an insulin response element found in the
promoter sequences of genes encoding such proteins
as phosphoenolpyruvate carboxykinase, glyceraldehyde
phosphate dehydrogenase, and prolactinproteins
whose synthesis is induced by insulin.
484
The MAPK cascade also has direct effects upon
protein synthesis, i.e., on the translation of mRNA
messages. For example, insulin stimulates phospho-
rylation of proteins that regulate a translation initia-
tion factor, a protein called eIF-4E (see Chapter 29).
Phosphorylation of inhibitory proteins allows them
to dissociate from the initiation factor so that protein
synthesis can proceed.
485,486
The scheme in Fig. 11-13 is complex, but in reality
it is much more complex than is shown. Each protein
kinase (receptor kinase, Raf-1, MEK, and MAPK) will
phosphorylate not only the proteins indicated in this
scheme but also any others that meet the specificity
requirements of the kinases. Thus, there will be
branches diverging from the pathways shown.
486a,b,c
There are isoenzymes that provide further divergence
and interaction.
487
Not only do pathways diverge but
also others converge. Thus, binding of many different
ligands to their receptors activates the same MAPK
cascade. For example, seven-helix G protein-coupled
receptors release their subunits which may also
activate Ras as indicated on the right edge of Fig. 11-13A.
At the same time the subunits of the heterotrimer
G proteins can affect not only adenylate cyclase but
also phospholipases C which can, in some cases, also
activate the MAP kinase pathway.
488,489
Sphingosine
1-phosphate may be released from membrane spingo-
lipids and activate the same cascade.
490
However,
hormones do not all affect cells in the same way. In
view of all the converging pathways, how is this possi-
ble? Part of the answer lies in the proximity or spatial
separation of components of the pathway. The kinases
exist in complexes with other signaling proteins and
may phosphorylate them, sending a signal back, as
well as forward via other protein substrates. There
are also unknown kinetic considerations. Hormones,
neurotransmitters, and calcium ions are often released
in pulses. The signaling system must integrate effects
of all the stimuli that arise from different parts of the
cell, at different times, and from differing receptors.
At the same time all of the phosphorylated proteins
are acted upon by phosphatases that may either acti-
vate or deactivate the proteins and by proteases that
process newly formed peptides and modify or destroy
mature proteins. The various modifying enzymes act
on cytosolic proteins, proteins of membranes, of the
cytoskeleton, and of the nucleus. The regulatory
processes that we discuss in such minute detail involve
the very substance of living cytoplasm which is ever-
changing and responding to its surroundings. The
flow of energy, provided by synthesis of ATP and by
the use of ATP by kinases, phosphatases, and the
protein synthetic machinery, goes along with the flow
of information and drives the signaling network.
Evolution has shaped this system to allow it to respond
appropriately for every species.
3. The Cell Cycle and Control of Growth
When a cell divides it is of utmost importance that
the DNA be replicated reliably. This requires that the
dividing cell be large enough and contain enough
biosynthetic precursor materials to complete the elabo-
rate process of DNA synthesis and of mitosis. The cell
579
Figure 11-13 (A) A simplified version of the mitogen-activated kinase (MAPK) signaling cascade. At left is shown a hormone
receptor, e.g., that for the epidermal growth factor (EGF). The receptor tyrosine kinase undergoes autophosphorylation on
numerous tyrosines. The resulting phosphotyrosyl (Y-P) groups bind to SH2 domains of adapters such as Grb2 and Shc.
Two pathways from the activated receptor are shown. At the left is activation of phospholipase C and formation, at a
membrane-bound site, of inositol trisphosphate and diacylglycerol (DAG). The main pathway, in the center, activates Ras
with the aid of the G protein Sos. Activated Ras, in turn, activates Raf and successive components of the MAPK cascade. At
the right a seven-helix receptor activates both phospholipase C and Ras via interaction with a subunit. (B) A generalized
scheme for the MAP kinase pathway. See Seger and Krebs.
380
c-fos
c-myc
GAP
Pi
H
2
O Ras GTP
Ras GDP
MEK-1,2-P
Ligand
Receptor
Plasma
membrane
Activated
receptor
tyrosine
kinase
(RTK)
Basic pattern
Stimulus
Initiating
events
MAPKKK
MAPKK
MAPK
Substrates
Y
Y
Y
Y
P
P
P
P
Y
Y
Y
Y
Y
P
P
P
P
P
Shc
Y
P
Raf-1,2
MEK-1,2 (MAPKK)
ERK-P ERK-1,2 (MAPK)
7-Helix
receptor
Hormone
G
q
or G
i

q
Phospholipase
C

RSK
c-jun
+
Protein
Kinase C
Nucleus
SH
2
SH
3
SH
3
S
o
s
Other signals
SH
2
Phospho-
lipase C

Ca
2+
+
DAG
PtdIns (4,5) P
2
Ins (1,4,5) P
3
Endoplasmic
reticulum
Genes for
transcription factors
Grb2
A B
replication cycle (or simply cell cycle) is commonly
shown as a circle in which the time from one cell division
to the next, in a rapidly growing organism or tissue, is
represented by the circumference. The time required
for DNA synthesis is the S-phase and the time required
for mitosis the mitotic or M-phase (Fig. 11-15). After
metaphase there is a gap in time denoted G
1
. A second
gap G
2
separates the synthetic S-phase and the M-phase.
The total time required for one cycle varies with condi-
tions. It may be as short as 860 min in an early embryo
but is usually two hours or more. A slowly growing
cell may pause before the G
1
phase in a nongrowing
G
0
phase.
491,492
What controls the cell replication cycle? Signals
H. Growth Factors, Oncogenes, and the Cell Cycle
580 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
Figure 11-14 Ribbon drawing of the three-dimensional
structure of adapter protein Grb2. The two SH3 domains at
the N and C termini are labeled, as is the central SH2 domain.
Produced with programs MolScript and Raster3D. From
Maignan et al.
476
Courtesy of Arnaud Ducruix.
from growth factors can stimulate a cell to leave G
0
and enter G
1
. However, to go further the cell depends
upon a large number of proteins of which two types are
prominent. The cyclins are labile 45- to 60-kDa proteins
which are degraded by the ubiquitin system as part of
the cycle. The cyclins associate with a series of protein
Ser/Thr kinases known as cyclin-dependent kinases
(CDKs). It is the cyclinCDK complexes that signal
the start of the next step in the cycle.
493 495
The number of different cyclins and CDK enzymes
needed varies with the organism, often being greater
for more complex species. As a rule, there are at least
two types of cyclins. The G
1
or start cyclins initiate
the passage through the start (G
1
checkpoint) into the
S-phase, while mitotic cyclins initiate the passage
from the S-phase into the M-phase. Both vertebrates
and Drosophila utilize at least four different types of
cyclin (A, B, D, and E) and also four or more CDKs
(CDK1, CDK2, CDK4, and CDK6).
492
A somewhat
different set of these proteins are found in yeast.
Control of cell growth is directly related to the cell
cycle. Mitogenic signals from growth factors act to
initiate progression through G
1
, apparently by stimu-
lating transcription of D-type cyclins.
496
They may
affect other steps as well. Oncogenic signals can arise
from such oncogenes as ras and abl.
497,498
As pointed
out in Box 11-D, the tumor-suppressor protein Rb
becomes phosphorylated in synchrony with the cell
Figure 11-15 The cell cycle,
which depicts the relative lengths
of time for each stage of cell
growth and division. After
mitosis (M) the cell grows during
the G
1
(gap) period. At the G
1
checkpoint the cell prepares to
divide, with DNA synthesis
occurring during the S-phase.
After a second gap (G
2
) mitosis
takes place to complete the cycle.
G
2
(DNA damage) checkpoint
Mitotic cyclin B
+ CDK1
G
2
Cyclin
degradation
Metaphase (spindle assembly) checkpoint
Cyclin D
(G
1
cyclin)
G
1
CDK4 or 6
Start 1 (G
1
damage
checkpoint or
restriction point)
CDK-cyclin D complex
Cyclin
degradation
M
i
t
o
s
i
s
,
M
Cyclin E + CDK2
Growth
factors
Cyclin A
+
Rb-EF2 etc.
Rb-P
n
EF2
etc.
Transcription
+
+

p53
Apoptosis
+

p53
Phosphorylation
of Rb protein
D
N
A
s
y
n
t
h
e
s
i
s
,
S
581 H. Growth Factors, Oncogenes, and the Cell Cycle
1. Krebs, H. A. (1957) Endeavour 16, 125132
2. Newsholme, E. A., and Start, C. (1973)
Regulation in Metabolism, Wiley, New York
3. Heinrich, R., Rapoport, S. M., and Rapoport,
T. A. (1977) Prog. Biophys. and Mol. Biol. 32,
182
4. Martin, B. R. (1987) Metabolic Regulation,
Blackwell Scientific Publ., Oxford
5. Chance, B., and Williams, G. R. (1955) J. Biol.
Chem. 217, 477488
6. Heinrich, R., and Rapoport, T. A. (1974) Eur. J.
Biochem. 42, 8995; 97105
7. Mendes, P. (1997) Trends Biochem. Sci. 22, 361
363
8. Wright, B. E. (1986) Trends Biochem. Sci. 11,
164165
9. Savageau, M. A. (1987) Trends Biochem. Sci. 12,
219220
10. Voit, E. O., and Savageau, M. A. (1987)
Biochemistry 26, 68696880
11. Hlavacek, W. S., and Savageau, M. A. (1996) J.
Mol. Biol. 255, 121139
12. Voit, E. O. (1987) Trends Biochem. Sci. 12, 221
13. Kacser, H., and Burns, J. A. (1979) Biochem.
Soc. Trans. 7, 11491166
14. Crabtree, B., and Newsholme, E. A. (1987)
Trends Biochem. Sci. 12, 412
15. Kholodenko, B. N., and Westerhoff, H. V.
(1995) Trends Biochem. Sci. 20, 5254
16. Fell, D. (1997) Understanding the Control of
Metabolism, Portland Press, London
17. Quant, P. A. (1993) Trends Biochem. Sci. 18,
2630
18. Salter, M., Knowles, R. G., and Pogson, C. I.
(1994) in Essays in Biochemistry, Vol. 28
(Tipton, K. F., ed), Portland Press, London
19. Schulze, E.-D. (1994) Flux Control in Biological
Systems, Academic Press, San Diego,
California
20. Kacser, H., and Burns, J. A. (1973) Symp. Soc.
Exp. Biol XXVII, 65104
21. Stark, G. R., and Wahl, G. M. (1984) Ann. Rev.
Biochem. 53, 447491
22. Wilkinson, J. H. (1970) Isoenzymes, 2nd ed.,
Lippincott, Philadelphia, Pennsylvania
23. Markert, C. L., ed. (1975) Isozymes, Vol. 4,
Academic Press, New York
24. Christen, P., and Metzler, D. E., eds. (1985)
Transaminases, Wiley, New York
25. Salerno, C., Giartosio, A., and Fasella, P.
(1986) in Vitamin B
6
Pyridoxal Phosphate, Vol.
1b (Dolphin, D., Poulson, R., and Avramovic,
O., eds), Wiley, New York
26. McPhalen, C. A., Vincent, M. G., and Jansonius,
J. N. (1992) J. Mol. Biol. 225, 495517
References
27. Mehta, P. K., Hale, T. I., and Christen, P. (1989)
Eur. J. Biochem. 186, 249253
28. Kamitori, S., Okamoto, A., Hirotsu, K.,
Higuchi, T., Kuramitsu, S., Kagamiyama, H.,
Matsuura, Y., and Katsube, Y. (1990)
J. Biochem. 108, 175184
29. Hogrefe, H. H., Griffith, J. P., Rossman, M. G.,
and Goldberg, E. (1987) J. Biol. Chem. 262,
1315513162
30. Purich, D. L., and Fromm, H. J. (1972) Curr.
Top. Cell. Regul. 6, 131167
31. Huijing, F. (1979) Trends Biochem. Sci. 4, N132
N134
31a. Aleshin, A. E., Kirby, C., Liu, X., Bourenkov,
G. P., Bartunik, H. D., Fromm, H. J., and
Honzatko, R. B. (2000) J. Mol. Biol. 296, 1001
1015
32. Purich, D. L., Fromm, H. J., and Rudolph, F.
R. (1973) Adv. Enzymol. 39, 249326
33. Breitbar, R. E., Andreadis, A., and Nadal -
Ginard, B. (1987) Ann. Rev. Biochem. 56, 467
495
34. Joh, K., Arai, Y., Mukai, T., and Hori, K. (1986)
J. Mol. Biol. 190, 401410
35. Haldane, J. B. S. (1930) Enzymes, Longmans,
Green, New York
36. Hayashi, S.-i, Murakami, Y., and Matsufuji, S.
(1996) Trends Biochem. Sci. 21, 2730
37. Li, X., Stebbins, B., Hoffman, L., Pratt, G.,
Rechsteiner, M., and Coffino, P. (1996) J. Biol.
Chem. 271, 44414446
38. Sprang, S. R., Acharya, K. R., Goldsmith, E. J.,
Stuart, D. I., Varvill, K., Fletterick, R. J.,
Madsen, N. B., and Johnson, L. N. (1988)
Nature (London) 336, 215221
39. Cohen, P. (1983) Control of Enzyme Activity,
2nd ed., Chapman and Hall, London
40. Kantrowitz, E. R., and Lipscomb, W. N. (1990)
Trends Biochem. Sci. 15, 5359
41. Zhou, B.-B., and Schachman, H. K. (1993)
Protein Sci. 2, 103112
42. Xi, X. G., De Staercke, C., Van Vliet, F.,
Triniolles, F., Jacobs, A., Stas, P. P., Ladjimi, M.
M., Simon, V., Cunin, R., and Herv, G. (1994)
J. Mol. Biol. 242, 139149
43. Uyeda, K., Furuya, E., and Sherry, A. D.
(1981) J. Biol. Chem. 256, 86798684
44. Hers, H., Hue, L., and Schaftingen, E. (1982)
Trends Biochem. Sci. 7, 329331
45. Lively, M. O., El-Maghrabi, M. R., Pilkis, J.,
DAngelo, G., Colosia, A. D., Ciavola, J.-A.,
Fraser, B. A., and Pilkis, S. J. (1988) J. Biol.
Chem. 263, 839849
46. Kitamura, K., and Uyeda, K. (1988) J. Biol.
Chem. 263, 90279033
46a. Choe, J.-Y., Fromm, H. J., and Honzatko, R. B.
(2000) Biochemistry 39, 85658574
47. Graves, D. J., Martin, B. L., and Wang, J. H.
(1994) Co and Post Translational Modification
of Proteins, Oxford Univ. Press, New York
47a. Skamnaki, V. T., Owen, D. J., Noble, M. E. M.,
Lowe, E. D., Lowe, G., Oikonomakos, N. G.,
and Johnson, L. N. (1999) Biochemistry 38,
1471814730
48. Johnson, L. N. (1992) FASEB J. 6, 22742282
48a. Cheng, A., Fitzgerald, T. J., Bhatnagar, D.,
Roskoski, R., Jr., and Carlson, G. M. (1988) J.
Biol. Chem. 263, 55345542
49. Dent, P., Lavoinne, A., Nakielny, S., Caudwell,
F. B., Watt, P., and Cohen, P. (1990) Nature
(London) 348, 302308
49a. Brady, M. J., Bourbonais, F. J., and Saltiel, A.
R. (1998) J. Biol. Chem. 273, 1406314066
50. Krebs, E. G., and Fischer, E. H. (1956) Biochim.
Biophys. Acta. 20, 150157
51. Edelman, A. M., Blumenthal, D. K., and Krebs,
E. G. (1987) Ann. Rev. Biochem. 56, 567613
52. Allen, J. F. (1992) Trends Biochem. Sci. 17, 1217
53. Hershey, J. W. B. (1989) J. Biol. Chem. 264,
2082320826
54. Kennelly, P. J., and Krebs, E. G. (1991) J. Biol.
Chem. 266, 1555515558
55. Hanks, S. K., and Hunter, T. (1995) FASEB J. 9,
576596
56. Woodgett, J. R., ed. (1994) Protein Kinases, IRL
Press, Oxford
57. Hunter, T. (1994) Sem. Cell Biol. 5, 367376
58. Kemp, B. E., Parker, M. W., Hu, S., Tiganis, T.,
and House, C. (1994) Trends Biochem. Sci. 19,
440444
59. Imazu, M., Stricklund, W. G., Chrisman, T. D.,
and Exton, J. H. (1984) J. Biol. Chem. 259,
18131821
60. He, X., Saint-Jeannet, J.-P., Woodgett, J. R.,
Varmus, H. E., and David, I. B. (1995) Nature
(London) 374, 617622
61. Hartl, F. T., and Roskoski, R., Jr. (1983) J. Biol.
Chem. 258, 39503955
62. Zheng, J., Knighton, D. R., Xuong, N.-H.,
Taylor, S. S., Sowadski, J. M., and Eyck, L. F. T.
(1993) Protein Sci. 2, 15591573
63. Beebe, S. J., Reimann, E. M., and Schlender, K.
K. (1984) J. Biol. Chem. 259, 14151422
64. Taylor, S. S., Knighton, D. R., Zheng, J.,
Sowadski, J. M., Gibbs, C. S., and Zoller, M. J.
(1993) Trends Biochem. Sci. 18, 8489
65. Narayana, N., Cox, S., Shaltiel, S., Taylor, S. S.,
and Xuong, N.-h. (1997) Biochemistry 36,
44384448
cycle. This seems to be an essential process for passage
through the G
1
checkpoint. The checkpoints in the
cycle should be viewed not as points but as essential
interlocking processes that must be accomplished
before transcription of genes essential for the next step
in the cycle can take place.
499
A possibility is that the
cyclin-dependent kinase in the cyclin DCDK4 (or
CDK6) complex phosphorylates the Rb protein which
in its unphosphorylated state forms a complex with
transcriptional regulators of the E2F family. Phospho-
rylation of Rb allows this complex to dissociate and
permits E2F to induce transcription of genes encoding
essential proteins for the next step in the cycle.
499a,b,c
Each CDK may also phosphorylate its associated
cyclin, a modification that may be essential to the
proteolysis of the cyclin and progression to the next
stage of the cycle.
The powerful cancer suppressor, p53, which is also
described in Box 11-D, in some way senses DNA dam-
age. It prevents passage through the G
1
checkpoint
and also through the G
2
checkpoint if the DNA has not
been adequately repaired.
496,497
Protein p53, whose
three-dimensional structure is known,
500,501
binds to
DNA and also induces transcription of genes that
cause arrest of the cell cycle. It may also induce cell
death (apoptosis), a process that may also require the
protein product of protooncogene c-myc.
502-505
A
variety of protein kinases and phosphatases act on p53
and influence its activity.
506
582 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
126. Sakakibara, Y., Takami, Y., Zwieb, C., Nakayama,
T., Suiko, M., Nakajima, H., and Liu, M.-C.
(1995) J. Biol. Chem. 270, 3047030478
127. Rosenquist, G. L., and Nicholas, H. B., Jr.
(1993) Protein Sci. 2, 215222
128. Sundaram, K. S., and Lev, M. (1992) J. Biol.
Chem. 267, 2404124044
129. Hooper, L. V., Manzella, S. M., and Baenziger,
J. U. (1996) FASEB J. 10, 11371146
130. Terwilliger, T. C., and Koshland, D. E., Jr.
(1984) J. Biol. Chem. 259, 77197725
131. Favre, B., Zolnierowicz, S., Turowski, P., and
Hemmings, B. A. (1994) J. Biol. Chem. 269,
1631116317
132. Edmondson, D. G., and Roth, S. Y. (1996)
FASEB J. 10, 11731182
133. Kuo, M.-H., Brownell, J. E., Sobel, R. E.,
Ranalli, T. A., Cook, R. G., Edmondson, D. G.,
Roth, S. Y., and Allis, C. D. (1996) Nature
(London) 383, 269272
134. Brandes, H. K., Larimer, F. W., and Hartman,
F. C. (1996) J. Biol. Chem. 271, 33333335
135. Brandes, H. K., Hartman, F. C., Lu, T. -Y. S.,
and Larimer, F. W. (1996) J. Biol. Chem. 271,
64906496
136. Swerdlow, R. D., Green, C. L., Setlow, B., and
Setlow, D. (1979) J. Biol. Chem. 254, 68356837
137. Hentze, M. W., Rouault, T. A., Harford, J. B.,
and Klausner, R. D. (1989) Science 244, 357359
138. Wadsworth, W. G., and Riddle, D. L. (1988)
Proc. Natl. Acad. Sci. U.S.A. 85, 84358438
139. Boron, W. F. (1984) Nature (London) 312, 312
140. Siffert, W., and Akkerman, J. W. N. (1988)
Trends Biochem. Sci. 13, 148151
141. Reinertsen, K. V., Tonnessen, T. I., Jacobsen, J.,
Sandvig, K., and Olsnes, S. (1988) J. Biol.
Chem. 263, 1111711125
142. Schramm, V. L. (1982) Trends Biochem. Sci. 7,
369371
143. Srere, P. A. (1987) Ann. Rev. Biochem. 56, 89124
144. Srivastava, D. K., and Bernhard, S. A. (1987)
Ann. Rev. Biophys. Biophys. Chem. 16, 175204
145. Ovdi, J. (1988) Trends Biochem. Sci. 13, 486490
146. Kaprelyants, A. S. (1988) Trends Biochem. Sci.
13, 4346
146a. Miles, E. W., Rhee, S., and Davies, D. R. (1999)
J. Biol. Chem. 274, 1219312196
146b. Serre, V., Guy, H., Penverne, B., Lux, M.,
Rotgeri, A., Evans, D., and Herv, G. (1999) J.
Biol. Chem. 274, 2379423801
146c. Geck, M. K., and Kirsch, J. F. (1999)
Biochemistry 38, 80328037
146d. DellAcqua, M. L., and Scott, J. D. (1997) J.
Biol. Chem. 272, 1288112884
147. Ovdi, J., and Srere, P. A. (1992) Trends
Biochem. Sci. 17, 445447
148. Inagaki, N., Ito, M., Nakano, T., and Inagaki,
M. (1994) Trends Biochem. Sci. 19, 448452
149. Faux, M. C., and Scott, J. D. (1996) Trends
Biochem. Sci. 21, 312315
150. Fraser, C. M. (1989) J. Biol. Chem. 264, 92669270
151. Strosberg, A. D. (1993) Protein Sci. 2, 11981209
152. Scheel, A. A., and Pelham, H. R. B. (1996)
Biochemistry 35, 1020310209
153. Levitzki, A. (1988) Science 241, 800806
153a. Stacey, M., Lin, H.-H., Gordon, S., and
McKnight, A. J. (2000) Trends Biochem. Sci. 25,
284289
153b. Leurs, R., Smit, M. J., Alewijnse, A. E., and
Timmerman, H. (1998) Trends Biochem. Sci. 23,
418422
153c. Bockaert, J., and Pin, J. P. (1999) EMBO J. 18,
17231729
154. Insel, P. A., and Wasserman, S. I. (1990) FASEB
J. 4, 27322736
155. Turki, J., Lorenz, J. N., Green, S. A., Donnelly,
E. T., Jacinto, M., and Liggett, S. B. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1048310488
66. Su, Y., Dostmann, W. R. G., Herberg, F. W.,
Durick, K., Xuong, N.-h, Eyck, L. T., Taylor, S.
S., and Varughese, K. I. (1995) Science 269,
807813
67. Lincoln, T. M., Thompson, M., and Cornwell,
T. L. (1988) J. Biol. Chem. 263, 1763217637
68. Weber, I. T., Shabb, J. B., and Corbin, J. D.
(1989) Biochemistry 28, 61226127
69. Fry, M. J., Panayotou, G., Booker, G. W., and
Waterfield, M. D. (1993) Protein Sci. 2, 1785
1797
70. Xu, R.-M., Carmel, G., Sweet, R. M., Kuret, J.,
and Cheng, X. (1995) EMBO J. 14, 10151023
71. Allende, J. E., and Allende, C. C. (1995) FASEB
J. 9, 313323
72. Seldin, D. C., and Leder, P. (1995) Science 267,
894897
72a. Niefind, K., Guerra, B., Pinna, L. A., Issinger,
O.-G., and Schomburg, D. (1998) EMBO J. 17,
24512462
73. Chan, K.- F., and Graves, D. J. (1984) in
Calcium and Cell Function, Vol. 5 (Cheung, W.
Y., ed), pp. 132, Academic Press, New York
74. Heilmeyer, L. M. G., Jr. (1991) Biochim.
Biophys. Acta. 1094, 168174
75. Norcum, M. T., Wilkinson, D. A., Carlson, M.
C., Hainfeld, J. F., and Carlson, G. M. (1994) J.
Mol. Biol. 241, 94102
76. Kee, S. M., and Graves, D. J. (1987) J. Biol.
Chem. 262, 94489453
77. Paudel, H. K., and Carlson, G. M. (1987) J.
Biol. Chem. 262, 1191211915
78. Cheng, A., Fitzgerald, T. J., Bhatnagar, D.,
Roskoski, R., Jr., and Carlson, G. M. (1988) J.
Biol. Chem. 263, 55345542
79. Owen, D. J., Papageorgiou, A. C., Garman, E.
F., Noble, M. E. M., and Johnson, L. N. (1995)
J. Mol. Biol. 246, 374381
80. Ohmstede, C.-A., Bland, M. M., Merrill, B. M.,
and Sahyoun, N. (1991) Proc. Natl. Acad. Sci.
U.S.A. 88, 57845788
81. Hanson, P. I., and Schulman, H. (1992) Ann.
Rev. Biochem. 61, 559601
82. Harper, J. F., Sussman, M. R., Schaller, G. E.,
Putnam-Evans, C., Charbonneau, H., and
Harmon, A. C. (1991) Science 252, 951954
83. Ullrich, A., Bell, J. R., Chen, E. Y., Herrera, R.,
Petruzzelli, L. M., Dull, T. J., Gray, A.,
Coussens, L., Liao, Y.-C., Tsubokawa, M.,
Mason, A., Seeburg, P. H., Grunfield, C.,
Roden, O. M., and Ramachandran, J. (1985)
Nature (London) 313, 756761
84. Wang, J. Y. J. (1994) Trends Biochem. Sci. 19,
373376
85. Clark, E. A., Shattil, S. J., and Brugge, J. S.
(1994) Trends Biochem. Sci. 19, 464469
86. Sun, H., and Tonks, N. K. (1994) Trends
Biochem. Sci. 19, 480485
87. Ingebritsen, T. S., and Cohen, P. (1983) Science
221, 331338
88. Cohen, P. (1989) Ann. Rev. Biochem. 58, 453508
89. Goldberg, J., Huang, H., Kwon, Y., Greengard,
P., Nairn, A. C., and Kuriyan, J. (1995) Nature
(London) 376, 745753
90. Fischer, E. H., Charbonneau, H., and Tonks,
N. K. (1991) Science 253, 401406
91. Yuvaniyama, J., Denu, J. M., Dixon, J. E., and
Saper, M. A. (1996) Science 272, 13281331
92. MacKintosh, C., and MacKintosh, R. W. (1994)
Trends Biochem. Sci. 19, 444448
93. Hidaka, H., and Kobayashi, R. (1994) in Essays
in Biochemistry, Vol. 28 (Tipton, K. F., ed), pp.
7398, Portland Press, London
94. Cohen, P., Holmes, C. F. B., and Tsukitani, Y.
(1990) Trends Biochem. Sci. 15, 98102
95. Price, N. E., and Mumby, M. C. (2000)
Biochemistry 39, 1131211318
96. Zhang, M., Van Etten, R. L., and Stauffacher,
C. V. (1994) Biochemistry 33, 1109711105
References
97. Barford, D., Flint, A. J., and Tonks, N. K.
(1994) Science 263, 13971403
98. Stone, R. L., and Dixon, J. E. (1994) J. Biol.
Chem. 269, 3132331326
99. Chen, W., Wilborn, M., and Rudolph, J. (2000)
Biochemistry 39, 1078110789
100. Kissinger, C. R., Parge, H. E., Knighton, D. R.,
Lewis, C. T., Pelletier, L. A., Tempczyk, A.,
Kalish, V. J., Tucker, K. D., Showalter, R. E.,
Moomaw, E. W., Gastinel, L. N., Habuka, N.,
Chen, X., Maldonado, F., Barker, J. E.,
Bacquet, R., and Villafranca, J. E. (1995)
Nature (London) 378, 641644
101. LaPorte, D. C., and Chung, T. (1985) J. Biol.
Chem. 260, 1529115297
102. Pilkis, S. J., Claus, T. H., Kurland, I. J., and
Lange, A. J. (1995) Ann. Rev. Biochem. 64, 799
835
103. Istvan, E. S., Hasemann, C. A., Kurumbail, R.
G., Uyeda, K., and Deisenhofer, J. (1995)
Protein Sci. 4, 24392441
104. Vertommen, D., Bertrand, L., Sontag, B., Di
Pietro, A., Louckx, M. P., Vidal, H., Hue, L.,
and Rider, M. H. (1996) J. Biol. Chem. 271,
1787517880
105. Abe, Y., Minami, Y., Li, Y., Nguyen, C., and
Uyeda, K. (1995) Biochemistry 34, 25532559
106. Ogushi, S., Lawson, J. W. R., Dobson, G. P.,
Veech, R. L., and Uyeda, K. (1990) J. Biol.
Chem. 265, 1094310949
107. Nimmo, H. G. (1984) Trends Biochem. Sci. 9,
475478
108. Dean, A. M., and Koshland, D. E., Jr. (1993)
Biochemistry 32, 93029309
109. Dean, A. M., and Koshland, D. E., Jr. (1990)
Science 249, 10441046
110. Saier, M. H., Jr., Wu, L.-F., and Reizer, J. (1990)
Trends Biochem. Sci. 15, 391395
110a. Song, H. K., Lee, J. Y., Lee, M. G., Moon, J.,
Min, K., Yang, J. K., and Suh, S. W. (1999) J.
Mol. Biol. 293, 753761
111. Swanson, R. V., Alex, L. A., and Simon, M. I.
(1994) Trends Biochem. Sci. 19, 485490
112. Saier, M. H., Jr. (1989) Microbiol. Rev. 53, 109120
113. Stock, J. B., Stock, A. M., and Mottonen, J. M.
(1990) Nature (London) 344, 395400
114. Boon, P., Chock, P. B., Rhee, S. G., and
Stadtman, E. R. (1980) Ann. Rev. Biochem. 49,
813843
115. Mura, U., Chock, P. B., and Stadtman, E. R.
(1981) J. Biol. Chem. 256, 1302213029
116. Almassy, R. J., Janson, C. A., Hamlin, R.,
Xuong, N.-H., and Eisenberg, D. (1986) Nature
(London) 323, 304309
117. Garcia, E., and Rhee, S. G. (1983) J. Biol. Chem.
258, 22462253
118. Moss, J., and Vaughan, M. (1995) J. Biol. Chem.
270, 1232712330
119. Amor, J. C., Harrison, D. H., Kahn, R. A., and
Ringe, D. (1994) Nature (London) 372, 704708
120. Boman, A. L., and Kahn, R. A. (1995) Trends
Biochem. Sci. 20, 147150
121. Lee, F.-J. S., Stevens, L. A., Hall, L. M.,
Murtagh, J. J., Jr., Kao, Y. L., Moss, J., and
Vaughan, M. (1994) J. Biol. Chem. 269, 21555
21560
122. Moss, J., Stanley, S. J., Nightingale, M. S.,
Murtagh, J. J., Jr., Monaco, L., Mishima, K.,
Chen, H.-C., Williamson, K.-C., and Tsai, S.-C.
(1992) J. Biol. Chem. 267, 1048110488
123. Guse, A. H., da Silva, C. P., Weber, K.,
Ashamu, G. A., Potter, B. V. L., and Mayr, G.
W. (1996) J. Biol. Chem. 271, 2394623953
124. Gaal, J. C., Smith, K. R., and Pearson, C. K.
(1987) Trends Biochem. Sci. 12, 129130
125. Huttner, W. B. (1987) Trends Biochem. Sci. 12,
361363
583
References
155a. Rohrer, D. K., Chruscinski, A., Schauble, E.
H., Bernstein, D., and Kobilka, B. K. (1999) J.
Biol. Chem. 274, 1670116708
155b. Limbird, L. E., and Vaughan, D. E. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 71257127
156. Premont, R. T., Inglese, J., and Lefkowitz, R. J.
(1995) FASEB J. 9, 175182
156a. Pitcher, J. A., Freedman, N. J., and Lefkowitz,
R. J. (1998) Ann. Rev. Biochem. 67, 653692
157. Mochly-Rosen, D. (1995) Science 268, 247251
158. Benovic, J. L., DeBlasi, A., Stone, W. C., Caron,
M. G., and Lefkowitz, R. J. (1989) Science 246,
235240
158a. Danner, S., Frank, M., and Lohse, M. J. (1998)
J. Biol. Chem. 273, 32233229
158b. Gabilondo, A. M., Hegler, J., Krasel, C.,
Boivin-Jahns, V., Hein, L., and Lohse, M. J.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 12285
12290
159. Rodbell, M., Birnbaumer, L., Pohl, S. L., and
Krans, H. M. J. (1971) J. Biol. Chem. 246, 1877
1882
160. Unson, C. G., Cypess, A. M., Wu, C.-R.,
Goldsmith, P. K., Merrifield, R. B., and
Sakmar, T. P. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 310315
161. Rosenthal, W., Antaramian, A., Gilbert, S.,
and Birnbaumer, M. (1993) J. Biol. Chem. 268,
1303013033
162. Sharif, M., and Hanley, M. R. (1992) Nature
(London) 357, 279280
163. McFarland, K. C., Sprengel, R., Phillips, H. S.,
Khler, M., Rosemblit, N., Nikolics, K.,
Segaloff, D. L., and Seeburg, P. H. (1989)
Science 245, 494499
164. Kudo, M., Osuga, Y., Kobilka, B. K., and
Hsueh, A. J. W. (1996) J. Biol. Chem. 271,
2247022478
165. Perlman, J. H., Laakkonen, L., Osman, R., and
Gershengorn, M. C. (1994) J. Biol. Chem. 269,
2338323386
166. Conti, M., Iona, S., Cuomo, M., Swinnen, J. V.,
Odeh, J., and Svoboda, M. E. (1995)
Biochemistry 34, 79797987
167. Smith, K. J., Scotland, G., Beattie, J., Trayer, I.
P., and Houslay, M. D. (1996) J. Biol. Chem.
271, 1670316711
168. Sutherland, E. W. (1972) Science 177, 401408
169. Pastan, I. (1972) Sci. Am. 227(Aug), 97105
170. Lalli, E., and Sassone-Corsi, P. (1994) J. Biol.
Chem. 269, 17359 17362
171. Bolwell, G. P. (1995) Trends Biochem. Sci. 20,
492495
172. Taussig, R., and Gilman, A. G. (1995) J. Biol.
Chem. 270, 14
173. Dessauer, C. W., Tesmer, J. J. G., Sprang, S. R.,
and Gilman, A. G. (1998) J. Biol. Chem. 273,
2583125839
174. Cooper, D. M. F., Mons, N., and Karpen, J. W.
(1995) Nature (London) 374, 421424
175. Dsaubry, L., Shoshani, I., and Johnson, R. A.
(1996) J. Biol. Chem. 271, 1402814034
176. Hurley, J. H. (1999) J. Biol. Chem. 274, 7599
7602
177. Hellevuo, K., Yoshimura, M., Mons, N.,
Hoffman, P. L., Cooper, D. M. F., and Tabakoff,
B. (1995) J. Biol. Chem. 270, 1158111589
178. Walseth, T. F., Gander, J. E., Eide, S. J., Krick,
T. P., and Goldberg, N. D. (1983) J. Biol. Chem.
258, 15441558
178a. Chen, Y., Cann, M. J., Litvin, T. N.,
Lourgenko, V., Sinclair, M. L., Levin, L. R.,
and Buck, J. (2000) Science 289, 625628
178b. Kaupp, U. B., and Weyand, I. (2000) Science
289, 559560
179. Hamm, H. E. (1998) J. Biol. Chem. 273, 669672
180. Neer, E. J. (1994) Protein Sci. 3, 314
181. Collins, S., Caron, M. G., and Lefkowitz, R. J.
(1992) Trends Biochem. Sci. 17, 3739
182. Hepler, J. R., and Gilman, A. G. (1992) Trends
Biochem. Sci. 17, 383387
183. Levitzki, A. (1988) Trends Biochem. Sci. 13,
298301
184. Rawls, R. L. (1987) Chem. Eng. News 65
December 21, 2639
185. Wang, N., Yan, K., and Rasenick, M. M. (1990)
J. Biol. Chem. 265, 12391242
186. Wang, Q., Mullah, B. K., and Robishaw, J. D.
(1999) J. Biol. Chem. 274, 1736517371
187. Mayorga, L. S., Diaz, R., and Stahl, P. D. (1989)
Science 244, 14751477
188. Coleman, D. E., and Sprang, S. R. (1996)
Trends Biochem. Sci. 21, 4144
189. Ray, K., Kunsch, C., Bonner, L. M., and
Robishaw, J. D. (1995) J. Biol. Chem. 270,
2176521771
190. Brown, A. M. (1991) FASEB J. 5, 21752179
191. Chen, J., DeVivo, M., Dingus, J., Harry, A., Li,
J., Sui, J., Carty, D. J., Blank, J. L., Exton, J. H.,
Stoffel, R. H., Inglese, J., Lefkowitz, R. J.,
Logothetis, D. E., Hildebrandt, J. D., and
Iyengar, R. (1995) Science 268, 11661169
192. Schulz, K., Danner, S., Bauer, P., Schrder, S.,
and Lohse, M. J. (1996) J. Biol. Chem. 271,
2254622551
193. Yoshida, T., Willardson, B. M., Wilkins, J. F.,
Jensen, G. J., Thornton, B. D., and Bitensky, M.
W. (1994) J. Biol. Chem. 269, 2405024057
193a. Whiteway, M., Hougan, L., Dignard, D., Tho-
mas, D. Y., Bell, L., Saari, G. C., Grant, F. J.,
O'Hara, P., and MacKay, V. L. (1989) Cell 56,
467477
194. Noel, J. P., Hamm, H. E., and Sigler, P. B.
(1993) Nature (London) 366, 654663
195. Lambright, D. G., Sondek, J., Bohm, A., Skiba,
N. P., Hamm, H. E., and Sigler, P. B. (1996)
Nature (London) 379, 311319
196. McLaughlin, S. K., McKinnon, P. J., and
Margolskee, R. F. (1992) Nature (London) 357,
563569
197. Hall, A. (1990) Science 249, 635640
198. Valencia, A., Chardin, P., Wittinghofer, A., and
Sander, C. (1991) Biochemistry 30, 46374648
199. Macara, I. G., Lounsbury, K. M., Richards, S.
A., McKiernan, C., and Bar-Sagi, D. (1996)
FASEB J. 10, 625630
200. Boguski, M. S., and McCormick, F. (1993)
Nature (London) 366, 643654
200a. Scita, G., Tenca, P., Frittoli, E., Tocchetti, A.,
Innocenti, M., Giardina, G., and Di Fiore, P. P.
(2000) EMBO J. 19, 23932398
201. Jurnak, F. (1985) Science 230, 3236
202. Sprinzl, M. (1994) Trends Biochem. Sci. 19,
245250
203. Berchtold, H., Reshetnikova, L., Reiser, C. O.
A., Schirmer, N. K., Sprinzl, M., and Hilgenfeld,
R. (1993) Nature (London) 365, 126132
204. Kawashima, T., Berthet-Colominas, C., Wulff,
M., Cusack, S., and Leberman, R. (1996)
Nature (London) 379, 511518
205. Jurnak, F. (1988) Trends Biochem. Sci. 13, 195198
206. Downward, J. (1990) Trends Biochem. Sci. 15,
469472
207. Wittinghofer, A., and Pai, E. F. (1991) Trends
Biochem. Sci. 16, 382387
208. Schlichting, I., Almo, S. C., Rapp, G., Wilson,
K., Petratos, K., Lentfer, A., Wittinghofer, A.,
Kabsch, W., Pai, E. F., Petsko, G. A., and Goody,
R. S. (1990) Nature (London) 345, 309315
209. Kraulis, P. J., Domaille, P. J., Campbell-Burk,
S. L., Van Aken, T., and Laue, E. D. (1994)
Biochemistry 33, 35153531
210. Bokoch, G. M., and Der, C. J. (1993) FASEB J.
7, 750759
211. Takai, Y., Sasaki, T., Tanaka, K., and Nakanishi,
H. (1995) Trends Biochem. Sci. 20, 227231
212. Fischer von Mollard, G., Stahl, B., Li, C.,
Sdhof, T. C., and Jahn, R. (1994) Trends
Biochem. Sci. 19, 164168
212a. Burridge, K. (1999) Science 283, 20282029
212b. Lin, R., Cerione, R. A., and Manor, D. (1999) J.
Biol. Chem. 274, 2363323641
212c. Desrosiers, R. R., Gauthier, F., Lanthier, J., and
Bliveau, R. (2000) J. Biol. Chem. 275, 14949
14957
212d. Schwartz, M. A., and Shattil, S. J. (2000) Trends
Biochem. Sci. 25, 388391
212e. Zhang, B., Zhang, Y., Wang, Z.-x, and Zheng,
Y. (2000) J. Biol. Chem. 275, 2529925307
213. Symons, M. (1996) Trends Biochem. Sci. 21,
178181
214. Ando, S., Kaibuchi, K., Sasaki, T., Hiraoka, K.,
Nishiyama, T., Mizuno, T., Asada, M., Nunoi,
H., Matsuda, I., Matsuura, Y., Polakis, P.,
McCormick, F., and Takai, Y. (1992) J. Biol.
Chem. 267, 2570925713
215. Nuoffer, C., and Balch, W. E. (1994) Ann. Rev.
Biochem. 63, 949990
216. Rybin, V., Ullrich, O., Rubino, M., Alexandrov,
K., Simon, I., Seabra, M. C., Goody, R., and
Zerial, M. (1996) Nature (London) 383, 266269
217. Garrett, M. D., Self, A. J., van Oers, C., and
Hall, A. (1989) J. Biol. Chem. 264, 1013
217a. Moss, J., and Vaughan, M. (1998) J. Biol. Chem.
273, 2143121434
217b. Hutchinson, J. P., and Eccleston, J. F. (2000)
Biochemistry 39, 1134811359
218. Scheffzek, K., Ahmadian, M. R., and
Wittinghofer, A. (1998) Trends Biochem. Sci. 23,
257262
218a. Berman, D. M., and Gilman, A. G. (1998) J.
Biol. Chem. 273, 12691272
218b. Coleman, D. E., and Sprang, S. R. (1999) J.
Biol. Chem. 274, 1666916672
219. Schalk, I., Zeng, K., Wu, S.-K., Stura, E. A.,
Matteson, J., Huang, M., Tandon, A., Wilson,
I. A., and Balch, W. E. (1996) Nature (London)
381, 4248
220. Buss, J. E., Mumby, S. M., Casey, P. J., Gilman,
A. G., and Sefton, B. M. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 74937497
221. Wedegaertner, P. B., Wilson, P. T., and Bourne,
H. R. (1995) J. Biol. Chem. 270, 503506
222. Song, J., Hirschman, J., Gunn, K., and Dohlman,
H. G. (1996) J. Biol. Chem. 271, 2027320283
223. Hepler, J. R., Biddlecome, G. H., Kleuss, C.,
Camp, L. A., Hofmann, S. L., Ross, E. M., and
Gilman, A. G. (1996) J. Biol. Chem. 271, 496504
224. Peseckis, S. M., and Resh, M. D. (1994) J. Biol.
Chem. 269, 3088830892
225. Giner, J.-L., and Rando, R. R. (1994) Biochemistry
33, 1511615123
226. Liu, L., Dudler, T., and Gelb, M. H. (1996) J.
Biol. Chem. 271, 2326923276
227. Casey, P. J. (1995) Science 268, 221225
228. Maltese, W. A., Sheridan, K. M., Repko, E. M.,
and Erdman, R. A. (1990) J. Biol. Chem. 265,
21482155
229. Milligan, G., Parenti, M., and Magee, A. I.
(1995) Trends Biochem. Sci. 20, 181186
230. Duncan, J. A., and Gilman, A. G. (1996) J. Biol.
Chem. 271, 2359423600
230a. Iiri, T., Backlund, P. S., Jr., Jones, T. L. Z.,
Wedegaertner, P. B., and Bourne, H. R. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1459214597
230b. Lee, T. W., Seifert, R., Guan, X., and Kobilka,
B. K. (1999) Biochemistry 38, 1380113809
231. Lambright, D. G., Noel, J. P., Hamm, H. E.,
and Sigler, P. B. (1994) Nature (London) 369,
621628
232. Sondek, J., Lambright, D. G., Noel, J. P.,
Hamm, H. E., and Sigler, P. B. (1994) Nature
(London) 372, 276279
584 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
293. Taylor, C. W., and Marshall, I. C. B. (1992)
Trends Biochem. Sci. 17, 403407
294. Gu, Q.-M., and Sih, C. J. (1994) J. Am. Chem.
Soc. 116, 74817486
295. Berridge, M. J. (1993) Nature (London) 365,
388389
296. Lee, H. C., Walseth, T. F., Bratt, G. T., Hayes,
R. N., and Clapper, D. L. (1989) J. Biol. Chem.
264, 16081615
297. Vu, C. Q., Lu, P.-J., Chen, C.-S., and Jacobson,
M. K. (1996) J. Biol. Chem. 271, 47474754
298. Dennis, E. A., Rhee, S. G., Billah, M. M., and
Hannun, Y. A. (1991) FASEB J. 5, 20682077
299. Sternweis, P. C., and Smrcka, A. V. (1992)
Trends Biochem. Sci. 17, 502506
300. Rhee, S. G., and Choi, K. D. (1992) J. Biol.
Chem. 267, 1239312396
300a. Kim, M. J., Chang, J.-S., Park, S. K., Hwang, J.-
I., Ryu, S. H., and Suh, P.-G. (2000) Biochemis-
try 39, 86748682
301. Jafri, M. S., and Keizer, J. (1995) Biophys. J. 69,
21392153
302. Combettes, L., Cheek, T. R., and Taylor, C. W.
(1996) EMBO J. 15, 20862093
303. van de Put, F. H. M. M., De Pont, J. J. H. H.
M., and Willems, P. H. G. M. (1994) J. Biol.
Chem. 269, 1243812443
304. Kukuljan, M., Rojas, E., Catt, K. J., and Stojilkovic,
S. S. (1994) J. Biol. Chem. 269, 48604865
305. Berridge, M. J., and Galione, A. (1988) FASEB
J. 2, 30743082
306. Meyer, T., Wensel, T., and Stryer, L. (1990)
Biochemistry 29, 3237
307. Luttrell, B. M. (1993) J. Biol. Chem. 268, 1521
1524
308. Wilson, M. P., and Majerus, P. W. (1996) J. Biol.
Chem. 271, 1190411910
309. Shears, S. B. (1989) J. Biol. Chem. 264, 19879
19886
309a. Zhu, D.-M., Tekle, E., Huang, C. Y., and Chock,
P. B. (2000) J. Biol. Chem. 275, 6063 6066
309b. Odom, A. R., Stahlberg, A., Wente, S. R., and
York, J. D. (2000) Science 287, 20262029
310. Ryu, S. H., Lee, S. Y., Lee, K.-Y., and Rhee, S.
G. (1987) FASEB J. 1, 388393
311. Fukuda, M., and Mikoshiba, K. (1996) J. Biol.
Chem. 271, 1883818842
312. Bird, G. St. J., and Putney, J. W., Jr. (1996) J.
Biol. Chem. 271, 67666770
313. Dixon, J. F., and Hokin, L. E. (1987) J. Biol.
Chem. 262, 1389213895
314. Heinz, D. W., Ryan, M., Bullock, T. L., and
Griffith, O. H. (1995) EMBO J. 14, 38553863
315. Ali, N., Craxton, A., and Shears, S. B. (1993) J.
Biol. Chem. 268, 61616167
316. Xie, W., Kaetzel, M. A., Bruzik, K. S., Dedman,
J. R., Shears, S. B., and Nelson, D. J. (1996) J.
Biol. Chem. 271, 1409214097
317. Ismailov, I. I., Fuller, C. M., Berdiev, B. K.,
Shlyonsky, V. G., Benos, D. J., and Barrett, K.
E. (1996) Proc. Natl. Acad. Sci. U.S.A. 93,
1050510509
318. Dasgupta, S., Dasgupta, D., Sen, M., Biswas,
S., and Biswas, B. B. (1996) Biochemistry 35,
49945001
319. Voglmaier, S. M., Bembenek, M. E., Kaplin, A.
I., Dormn, G., Olszewski, J. D., Prestwich, G.
D., and Snyder, S. H. (1996) Proc. Natl. Acad.
Sci. U.S.A. 93, 43054310
319a. Chi, T. H., and Crabtree, G. R. (2000) Science
287, 19371939
320. Menniti, F. S., Oliver, K. G., Putney, J. W., Jr.,
and Shears, S. B. (1993) Trends Biochem. Sci. 18,
5356
320a. Yang, X., Safrany, S. T., and Shears, S. B. (1999)
J. Biol. Chem. 274, 3543435440
233. Coleman, D. E., Berghuis, A. M., Lee, E.,
Linder, M. E., Gilman, A. G., and Sprang, S. R.
(1994) Science 265, 14051412
234. Mixon, M. B., Lee, E., Coleman, D. E.,
Berghuis, A. M., Gilman, A. G., and Sprang, S.
R. (1995) Science 270, 954960
235. Kjeldgaard, M., Nyborg, J., and Clark, B. F. C.
(1996) FASEB J. 10, 13471368
236. Higashijima, T., Graziano, M. P., Suga, H.,
Kainosho, M., and Gilman, A. G. (1991) J. Biol.
Chem. 266, 33963401
237. Sondek, J., Bohm, A., Lambright, D. G.,
Hamm, H. E., and Sigler, P. B. (1996) Nature
(London) 379, 369374
238. Clapham, D. E. (1996) Nature (London) 379,
297299
239. Neer, E. J., Schmidt, C. J., Nambudripad, R.,
and Smith, T. F. (1994) Nature (London) 371,
297300
240. Faber, X. X. (1994) Structure 3, 551559
241. Pai, E. F., Kabsch, W., Krengel, U., Holmes, K.
C., John, J., and Wittinghofer, A. (1989) Nature
(London) 341, 209214
242. Wall, M. A., Posner, B. A., and Sprang, S. R.
(1998) Structure 6, 11691183
243. Wilson, P. T., and Bourne, H. R. (1995) J. Biol.
Chem. 270, 96679675
244. Zhao, Y., Brandish, P. E., DiValentin, M.,
Schelvis, J. P. M., Babcock, G. T., and Marletta,
M. A. (2000) Biochemistry 39, 1084810854
245. Garbers, D. L., and Lowe, D. G. (1994) J. Biol.
Chem. 269, 3074130744
246. Subbaraya, I., Ruiz, C. C., Helekar, B. S., Zhao,
X., Gorczyca, W. A., Pettenati, M. J., Rao, P.
N., Palczewski, K., and Baehr, W. (1994) J.
Biol. Chem. 269, 3108031089
247. Lincoln, T. M., and Cornwell, T. L. (1993)
FASEB J. 7, 328338
248. Houslay, M. D. (1985) Trends Biochem. Sci. 10,
465466
249. Garbers, D. L. (1989) J. Biol. Chem. 264, 9103
9106
250. Lowe, D. G., Chang, M.-S., Hellmiss, R.,
Chen, E., Singh, S., Garbers, D. L., and
Goeddel, D. V. (1989) EMBO J. 8, 13771384
251. Hamra, F. K., Forte, L. R., Eber, S. L.,
Pidhorodeckyj, N. V., Krause, W. J., Freeman,
R. H., Chin, D. T., Tompkins, J. A., Fok, K. F.,
Smith, C. E., Duffin, K. L., Siegel, N. R., and
Currie, M. G. (1993) Proc. Natl. Acad. Sci.
U.S.A. 90, 1046410468
252. Stamler, J. S., Singel, D. J., and Loscalzo, J.
(1992) Science 258, 18981902
253. Yuen, P. S. T., Doolittle, L. K., and Garbers, D.
L. (1994) J. Biol. Chem. 269, 791793
254. Stone, J. R., and Marletta, M. A. (1996)
Biochemistry 35, 10931099
255. Mayer, B., Schrammel, A., Klatt, P., Koesling,
D., and Schmidt, K. (1995) J. Biol. Chem. 270,
1735517360
256. Stone, J. R., and Marletta, M. A. (1995)
Biochemistry 34, 1639716403
257. Deinum, G., Stone, J. R., Babcock, G. T., and
Marletta, M. A. (1996) Biochemistry 35, 1540
1547
258. Verma, A., Hirsch, D. J., Glatt, C. E., Ronnett,
G. V., and Snyder, S. H. (1993) Science 259,
381384
259. Stock, J. B., Surette, M. G., McCleary, W. R.,
and Stock, A. M. (1992) J. Biol. Chem. 267,
1975319756
259a. Kim, K. K., Yokota, H., and Kim, S.-H. (1999)
Nature (London) 400, 787792
260. Scott, W. G., Milligan, D. L., Milburn, M. V.,
Priv, G. G., Yeh, J., Koshland, D. E., Jr., and
Kim, S.-H. (1993) J. Mol. Biol. 232, 555573
261. Danielson, M. A., Biemann, H.-P., Koshland,
D. E., Jr., and Falke, J. J. (1994) Biochemistry 33,
61006109
References
262. Yeh, J. I., Biemann, H.-P., Priv, G. G., Pandit,
J., Koshland, D. E., Jr., and Kim, S.-H. (1996) J.
Mol. Biol. 262, 186201
263. Danielson, M. A., Bass, R. B., and Falke, J. J.
(1997) J. Biol. Chem. 272, 3287832888
264. Kim, S.-H. (1994) Protein Sci. 3, 159165
264a. Ottemann, K. M., Xiao, W., Shin, Y.-K., and
Koshland, D. E., Jr. (1999) Science 285, 1751
1754
265. Berridge, M. J., and Irvine, R. F. (1989) Nature
(London) 341, 197205
266. Schulman, H., and Lou, L. L. (1989) Trends
Biochem. Sci. 14, 6266
267. Carafoli, E. (1987) Ann. Rev. Biochem. 56, 395
433
268. Rasmussen, H. (1989) Sci. Am. 261(Oct), 6673
269. Kobilka, B. K., Matsui, H., Kobilka, T. S.,
Yang-Feng, T. L., Francke, U., Caron, M. G.,
Lefkowitz, R. J., and Regan, J. W. (1987)
Science 238, 650656
270. Cotecchia, S., Kobilka, B. K., Daniel, K. W.,
Nolan, R. D., Lapetina, E. Y., Caron, M. G.,
Lefkowitz, R. J., and Regan, J. W. (1990) J. Biol.
Chem. 265, 6369
271. Ceresa, B. P., and Limbird, L. E. (1994) J. Biol.
Chem. 269, 2955729564
272. Scheer, A., Fanelli, F., Costa, T., De Benedetti,
P. G., and Cotecchia, S. (1996) EMBO J. 15,
35663578
273. Hwa, J., and Perez, D. M. (1996) J. Biol. Chem.
271, 63226327
274. Boyer, J. L., Waldo, G. L., Evans, T., Northup,
J. K., Downes, C. P., and Harden, T. K. (1989)
J. Biol. Chem. 264, 1391713922
275. Wu, D., Jiang, H., and Simon, M. I. (1995) J.
Biol. Chem. 270, 98289832
276. Nishizuka, Y. (1995) FASEB J. 9, 484 496
277. Majerus, P. W. (1992) Ann. Rev. Biochem. 61,
225250
278. Majerus, P. W., Conolly, T. M., Bansal, V. S.,
Inhorn, R. C., Ross, T. S., and Lips, D. L.
(1988) J. Biol. Chem. 263, 30513054
279. Rhee, S. G., Suh, P.-G., Ryu, S.-H., and Lee, S.
Y. (1989) Science 244, 546550
280. Hough, E., Hansen, L. K., Birknes, B., Jynge,
K., Hansen, S., Hordvik, A., Little, C.,
Dodson, E., and Derewenda, Z. (1989) Nature
(London) 338, 357360
281. Essen, L.-O., Perisic, O., Cheung, R., Katan,
M., and Williams, R. L. (1996) Nature (London)
380, 595602
282. Berridge, M. J. (1993) Nature (London) 361,
315325
283. Parthasarathy, R., and Eisenberg, F. J. (1986)
Biochem. J. 235, 313322
284. Majerus, P. W., Connolly, T. M., Deckmyn, H.,
Ross, T. S., Bross, T. E., Ishii, H., Bansal, V. S.,
and Wilson, D. B. (1986) Science 234, 15191526
285. Houslay, M. D. (1987) Trends Biochem. Sci. 12, 12
286. Nickels, J. T., Jr., Buxeda, R. J., and Carman,
G. M. (1994) J. Biol. Chem. 269, 1101811024
287. Boronenkov, I. V., and Anderson, R. A. (1995)
J. Biol. Chem. 270, 28812884
288. Hughes, A. R., Takemura, H., and Putney, J.
W., Jr. (1988) J. Biol. Chem. 263, 1031410319
289. Furuichi, T., Yoshikawa, S., Miyawaki, A.,
Wada, K., Maeda, N., and Mikoshiba, K.
(1989) Nature (London) 342, 3238
289a. Zimmermann, B., and Walz, B. (1999) EMBO
J. 18, 32223231
290. Yoshikawa, F., Morita, M., Monkawa, T.,
Michikawa, T., Furuichi, T., and Mikoshiba, K.
(1996) J. Biol. Chem. 271, 1827718284
291. Humbert, J.-P., Matter, N., Artault, J.-C.,
Kppler, P., and Malviya, A. N. (1996) J. Biol.
Chem. 271, 478485
292. Hennager, D. J., Welsh, M. J., and DeLisle, S.
(1995) J. Biol. Chem. 270, 49594962
585
References
321. Stephens, L., Radenberg, T., Thiel.U, Vogel,
G., Khoo, K.-H., Dell, A., Jackson, T. R.,
Hawkins, P. T., and Mayr, G. W. (1993) J. Biol.
Chem. 268, 40094015
322. Van Dijken, P., de Haas, J.-R., Craxton, A.,
Erneux, C., Shears, S. B., and Van Haastert, P.
J. M. (1995) J. Biol. Chem. 270, 2972429731
323. Bansal, V. S., Caldwell, K. K., and Majerus, P.
W. (1990) J. Biol. Chem. 265, 18061811
324. York, J. D., Ponder, J. W., Chen, Z.-w,
Mathews, F. S., and Majerus, P. W. (1994)
Biochemistry 33, 1316413171
325. Norris, F. A., Auethavekiat, V., and Majerus, P.
W. (1995) J. Biol. Chem. 270, 1612816133
326. Jefferson, A. B., and Majerus, P. W. (1995) J.
Biol. Chem. 270, 93709377
326a. Norris, F. A., Wilson, M. P., Wallis, T. S.,
Galyov, E. E., and Majerus, P. W. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 1405714059
326b. Munday, A. D., Norris, F. A., Caldwell, K. K.,
Brown, S., Majerus, P. W., and Mitchell, C. A.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 3640
3645
327. Bone, R., Springer, J. P., and Atack, J. R. (1992)
Proc. Natl. Acad. Sci. U.S.A. 89, 1003110035
328. Ganzhorn, A. J., Lepage, P., Pelton, P. D.,
Strasser, F., Vincendon, P., and Rondeau, J.-M.
(1996) Biochemistry 35, 1095710966
329. Burgoyne, R. D. (1989) Trends Biochem. Sci. 14,
8788
330. Huang, K.-P., Huang, F. L., Nakabayashi, H.,
and Yoshida, Y. (1988) J. Biol. Chem. 263,
1483914845
330a. Ron, D., and Kazanietz, M. G. (1999) FASEB J.
13, 16581676
330b. Ward, N. E., Stewart, J. R., Ionnides, C. G.,
and OBrian, C. A. (2000) Biochemistry 39,
1031910329
331. Pessin, M. S., and Raben, D. M. (1989) J. Biol.
Chem. 264, 87298738
332. Exton, J. H. (1990) J. Biol. Chem. 265, 14
332a. Perisic, O., Paterson, H. F., Mosedale, G.,
Lara-Gonzlez, S., and Williams, R. L. (1999)
J. Biol. Chem. 274, 1497914987
333. Roberts, M. F. (1996) FASEB J. 10, 11591172
334. Liscovitch, M., Chalifa, V., Pertile, P., Chen,
C.-S., and Cantley, L. C. (1994) J. Biol. Chem.
269, 2140321406
335. Stein, R. L., Melandri, F., and Dick, L. (1935)
Biochemistry 35, 38993908
336. Ward, S. G., Mills, S. J., Liu, C., Westwick, J.,
and Potter, B. V. L. (1995) J. Biol. Chem. 270,
1207512084
337. Woscholski, R., Kodaki, T., Palmer, R. H.,
Waterfield, M. D., and Parker, P. J. (1995)
Biochemistry 34, 1148911493
337a. Odorizzi, G., Babst, M., and Emr, S. D. (2000)
Trends Biochem. Sci. 25, 229235
337b. Walker, E. H., Perisic, O., Ried, C., Stephens,
L., and Williams, R. L. (1999) Nature (London)
402, 313320
337c. Ching, T.-T., Wang, D.-S., Hsu, A.-L., Lu, P.-J.,
and Chen, C.-S. (1999) J. Biol. Chem. 274, 8611
8617
337d. Bertsch, U., Deschermeier, C., Fanick, W.,
Girkontaite, I., Hillemeier, K., Johnen, H.,
Weglhner, W., Emmrich, F., and Mayr, G. W.
(2000) J. Biol. Chem. 275, 15571564
337e. Jackson, T. R., Kearns, B. G., and Theibert, A.
B. (2000) Trends Biochem. Sci. 25, 489495
337f. Gillooly, D. J., Morrow, I. C., Lindsay, M.,
Gould, R., Bryant, N. J., Gaullier, J.-M., Par-
ton, R. G., and Stenmark, H. (2000) EMBO J.
19, 45774588
338. Pennisi, E. (1997) Science 275, 18761878
338a. Stambolic, V., Suzuki, A., de la Pompa, J. L.,
Brothers, G. M., Mirtsos, C., Sasaki, T.,
Ruland, J., Penninger, J. M., Siderovski, D. P.,
and Mak, T. W. (1998) Cell 95, 2939
338b. Toker, A., and Cantley, L. C. (1997) Nature
(London) 387, 673675
339. Ui, M., Okada, T., Hazeki, K., and Hazeki, O.
(1995) Trends Biochem. Sci. 20, 303307
339a. Takasuga, S., Katada, T., Ui, M., and Hazeki,
O. (1999) J. Biol. Chem. 274, 1954519550
340. Chock, P. B., Rhee, S. G., and Stadtman, E. R.
(1980) Ann. Rev. Biochem. 49, 813843
341. Stadtman, E. R., and Chock, P. B. (1977) Proc.
Natl. Acad. Sci. U.S.A. 74, 27612765
342. Huang, C.-Y. F., and Ferrell, J. E., Jr. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1007810083
343. Goldbeter, A., and Koshland, D. E., Jr. (1987) J.
Biol. Chem. 262, 44604471
344. Schacter-Noiman, E., Chock, P. B., and
Stadtman, E. R. (1983) Philos. Trans. R. Soc.
London B 302, 157166
345. Schacter, E., Chock, P. B., and Stadtman, E. R.
(1984) J. Biol. Chem. 259, 1226012264
346. Newsholme, E. A., Challiss, R. A. J., and Crabtree,
B. (1984) Trends Biochem. Sci. 9, 277280
347. Koshland, D. E., Jr. (1987) Trends Biochem. Sci.
12, 225228
348. Taylor, S. I. (1995) in The Metabolic and Molecular
Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver,
C. R., Beaudet, A. L., Sly, W. S., and Valle, D.,
eds), pp. 843896, McGraw-Hill, New York
349. Mller, G., Rouveyre, N., Crecelius, A., and
Bandlow, W. (1998) Biochemistry 37, 86838695
350. Smit, A. B., Vreugdenhil, E., Ebberink, R. H.
M., Geraerts, W. P. M., Klootwijk, J., and
Joosse, J. (1988) Nature (London) 331, 535538
351. LeRoith, D., Shiloach, J., Roth, J., and Lesniak,
M. A. (1983) J. Biol. Chem. 256, 65336536
352. Rosen, O. M. (1987) Science 237, 14521458
353. White, M. F., and Kahn, C. R. (1994) J. Biol.
Chem. 269, 14
354. Barnard, R. J., and Youngren, J. F. (1992)
FASEB J. 6, 32383244
355. Bell, G. I., Burant, C. F., Takeda, J., and Gould,
G. W. (1993) J. Biol. Chem. 268, 1916119164
356. Mueckler, M. (1994) Eur. J. Biochem. 219, 713
725
356a. Li, J., Houseknecht, K. L., Stenbit, A. E., Katz,
E. B., and Charron, M. J. (2000) FASEB J. 14,
11171125
357. Cuatrecasas, P. (1972) Proc. Natl. Acad. Sci.
U.S.A. 69, 12771281
358. Pashmforoush, M., Yoshimasa, Y., and Steiner,
D. F. (1994) J. Biol. Chem. 269, 3263932648
359. Ullrich, A., Bell, J. R., Chen, E. Y., Herrera, R.,
Petruzzelli, L. M., Dull, T. J., Gray, A.,
Coussens, L., Liao, Y.-C., Tsubokawa, M.,
Mason, A., Seeburg, P. H., Grunfeld, C.,
Rosen, O. M., and Ramachandran, J. (1985)
Nature (London) 313, 756761
360. Xu, Q.-Y., Paxton, R. J., and Fujita-Yamaguchi,
Y. (1990) J. Biol. Chem. 265, 1867318681
361. Herrera, R., Petruzzelli, L., Thomas, N.,
Bramson, H. N., Kaiser, E. T., and Rosen, O.
M. (1985) Proc. Natl. Acad. Sci. U.S.A. 82,
78997903
362. McClain, D. A., Maegawa, H., Lee, J., Dull, T.
J., Ullrich, A., and Olefsky, J. M. (1987) J. Biol.
Chem. 262, 1466314671
363. Perlman, R., Bottaro, D. P., White, M. F., and
Kahn, C. R. (1989) J. Biol. Chem. 264, 89468950
363a. Rouard, M., Bass, J., Grigorescu, F., Garrett, T.
P. J., Ward, C. W., Lipkind, G., Jaffiole, C.,
Steiner, D. F., and Bell, G. I. (1999) J. Biol.
Chem. 274, 1848718491
363b. Luo, R. Z.-T., Beniac, D. R., Fernandes, A., Yip,
C. C., and Ottensmeyer, F. P. (1999) Science
285, 10771080
363c. Woldin, C. N., Hing, F. S., Lee, J., Pilch, P. F.,
and Shipley, G. G. (1999) J. Biol. Chem. 274,
3498134992
364. Lemmon, M. A., and Schlessinger, J. (1994)
Trends Biochem. Sci. 19, 459563
365. Hubbard, S. R., Wei, L., Ellis, L., and
Hendrickson, W. A. (1994) Nature (London)
372, 746754
365a. Hubbard, S. R. (1997) EMBO J. 16, 55725581
366. White, M. F., Shoelson, S. E., Keutmann, H.,
and Kahn, C. R. (1988) J. Biol. Chem. 263,
29692980
367. Fabry, M., Schaefer, E., Ellis, L., Kojro, E.,
Fahrenholz, F., and Brandenburg, D. (1992)
J. Biol. Chem. 267, 89508956
368. Williams, P. F., Mynarcik, D. C., Yu, G. Q., and
Whittaker, J. (1995) J. Biol. Chem. 270, 30123016
369. Kjeldsen, T., Wiberg, F. C., and Andersen, A.
S. (1994) J. Biol. Chem. 269, 3294232946
369a. Sparrow, L. G., McKern, N. M., Gorman, J. J.,
Strike, P. M., Robinson, C. P., Bentley, J. D.,
and Ward, C. W. (1997) J. Biol. Chem. 272,
2946029467
369b. Garant, M. J., Kole, S., Maksimova, E. M., and
Bernier, M. (1999) Biochemistry 38, 58965904
370. Clark, S., and Harrison, L. C. (1983) J. Biol.
Chem. 258, 1143411437
371. Saltiel, A. R. (1994) FASEB J. 8, 10341040
372. Carter, W. G., Asamoah, K. A., and Sale, G. J.
(1995) Biochemistry 34, 94889499
373. Myers, M. G., Jr., Sun, X. J., and White, M. F.
(1994) Trends Biochem. Sci. 19, 289293
374. Yenush, L., Makati, K. J., Smith-Hall, J.,
Ishibashi, O., Myers, M. G. J., and White, M. F.
(1996) J. Biol. Chem. 271, 2430024306
375. Kowalski-Chauvel, A., Pradayrol, L., Vaysse,
N., and Seva, C. (1996) J. Biol. Chem. 271,
2635626361
376. Paz, K., Voliovitch, H., Hadari, Y. R., Roberts,
C. T., Jr., LeRoith, D., and Zick, Y. (1996) J.
Biol. Chem. 271, 69987003
376a. Rother, K. I., Imai, Y., Caruso, M., Beguinot, F.,
Formisano, P., and Accili, D. (1998) J. Biol.
Chem. 273, 1749117497
376b. Clark, S. F., Molero, J.-C., and James, D. E.
(2000) J. Biol. Chem. 275, 38193826
376c. Lehr, S., Kotzka, J., Herkner, A., Sikmann, A.,
Meyer, H. E., Krone, W., and Mller-Wieland,
D. (2000) Biochemistry 39, 1089810907
377. Ricketts, W. A., Rose, D. W., Shoelson, S., and
Olefsky, J. M. (1996) J. Biol. Chem. 271, 26165
26169
378. Okada, S., and Pessin, J. E. (1996) J. Biol. Chem.
271, 2553325538
379. Czech, M. P., Klarlund, J. K., Yagaloff, K. A.,
Bradford, A. P., and Lewis, R. E. (1988) J. Biol.
Chem. 263, 1101711020
380. Seger, R., and Krebs, E. G. (1995) FASEB J. 9,
726735
381. Carter, W. G., Sullivan, A. C., Asamoah, K. A.,
and Sale, G. J. (1996) Biochemistry 35, 14340
14351
382. Kasus-Jacobi, A., Perdereau, D., Auzan, C.,
Clauser, E., Van Obberghen, E., Mauvais-
Jarvis, F., Girard, J., and Burnol, A.-F. (1998) J.
Biol. Chem. 273, 2602626035
383. Baltensperger, K., Karoor, V., Paul, H., Ruoho,
A., Czech, M. P., and Malbon, C. C. (1996) J.
Biol. Chem. 271, 10611064
384. Hara, K., Yonezawa, K., Sakaue, H., Ando, A.,
Kotani, K., Kitamura, T., Kitamura, Y., Ueda,
H., Stephens, L., Jackson, T. R., Hawkins, P. T.,
Dhand, R., Clark, A. E., Holman, G. D.,
Waterfield, M. D., and Kasuga, M. (1994) Proc.
Natl. Acad. Sci. U.S.A. 91, 74157419
385. Lam, K., Carpenter, C. L., Ruderman, N. B.,
Friel, J. C., and Kelly, K. L. (1994) J. Biol. Chem.
269, 2064820652
386. Sutherland, C., OBrien, R. M., and Granner,
D. K. (1995) J. Biol. Chem. 270, 1550115506
387. Domin, J., Dhand, R., and Waterfield, M. D.
(1996) J. Biol. Chem. 271, 2161421621
387a. Kosaki, A., Yamada, K., Suga, J., Otaka, A., and
Kuzuya, H. (1998) J. Biol. Chem. 273, 940944
586 Chapter 11. The Regulation of Enzymatic Activity and Metabolism
444. Greenhalgh, D. A., Welty, D. J., Player, A., and
Yuspa, S. H. (1990) Proc. Natl. Acad. Sci. U.S.A.
87, 643647
445. Rabbitts, T. H., Forster, A., Hamlyn, P., and
Baer, R. (1984) Nature (London) 309, 592597
446. Shima, E. A., Le Beau, M. M., McKeithan, T.
W., Minowada, J., Showe, L. C., Mak, T. W.,
Minden, M. D., Rowley, J. D., and Diaz, M. O.
(1986) Proc. Natl. Acad. Sci. U.S.A. 83, 34393443
447. Cramer, C. J., and Truhlar, D. G. (1993) J. Am.
Chem. Soc. 115, 57455753
448. Perrin, C. L., Armstrong, K. B., and Fabian, M.
A. (1994) J. Am. Chem. Soc. 116, 715722
449. Gabb, H. A., and Harvey, S. C. (1993) J. Am.
Chem. Soc. 115, 42184227
450. Plavec, J., Tong, W., and Chattopadhyaya, J.
(1993) J. Am. Chem. Soc. 115, 97349746
451. Ellervik, U., and Magnusson, G. (1994) J. Am.
Chem. Soc. 116, 23402347
451a. Hermeking, H., Rago, C., Schuhmacher, M.,
Li, Q., Barrett, J. F., Obaya, A. J., OConnell, B.
C., Mateyak, M. K., Tam, W., Kohlhuber, F.,
Dang, C. V., Sedivy, J. M., Eick, D., Vogelstein,
B., and Kinzler, K. W. (2000) Proc. Natl. Acad.
Sci. U.S.A. 97, 22292234
452. Eden, A., Simchen, G., and Benvenisty, N.
(1996) J. Biol. Chem. 271, 2024220245
453. Postel, E. H., Berberich, S. J., Flint, S. J., and
Ferrone, C. A. (1993) Science 261, 478480
454. Ying, G.-G., Proost, P., van Damme, J.,
Bruschi, M., Introna, M., and Golay, J. (2000) J.
Biol. Chem. 275, 41524158
455. Macleod, K., Leprince, D., and Stehelin, D.
(1992) Trends Biochem. Sci. 17, 251256
456. Karin, M. (1995) J. Biol. Chem. 270, 1648316486
457. Turner, R., and Tjian, R. (1989) Science 243,
16891694
458. Vogt, P. K., and Bos, T. J. (1989) Trends
Biochem. Sci. 14, 172175
459. Neuberg, M., Schuermann, M., Hunter, J. B.,
and Mller, R. (1989) Nature (London) 338,
589590
460. Treisman, R. (1995) EMBO J. 14, 49054913
461. Abel, T., and Maniatis, T. (1989) Nature
(London) 341, 2425
462. Glover, J. N. M., and Harrison, S. C. (1995)
Nature (London) 373, 257261
463. Quantin, B., and Breathnach, R. (1988) Nature
(London) 334, 538539
464. Greenberg, M. E., and Ziff, E. B. (1984) Nature
(London) 311, 433438
465. Mller, R., Bravo, R., Burckhardt, J., and Curran,
T. (1984) Nature (London) 312, 716720
466. Fukumoto, Y., Kaibuchi, K., Oku, N., Hori, Y.,
and Takai, Y. (1990) J. Biol. Chem. 265, 774780
467. Wang, Z.-Q., Ovitt, C., Grigoriadis, A. E.,
Mhle-Steinlein, U., Rther, U., and Wagner,
E. F. (1992) Nature (London) 360, 741745
468. Candeliere, G. A., Glorieux, F. H.,
Prudhomme, J., and St.-Arnaud, R. (1995) N.
Engl. J. Med. 332, 15461551
469. Egan, S. E., and Weinberg, R. A. (1993) Nature
(London) 365, 781783
470. Davis, R. J. (1994) Trends Biochem. Sci. 19, 470
473
470a. Gutkind, J. S. (1998) J. Biol. Chem. 273, 18391842
470b. Whitmarsh, A. J., and Davis, R. J. (1998)
Trends Biochem. Sci. 23, 481485
471. Pawson, T. (1995) Nature (London) 373, 573580
472. Daly, R. J., Sanderson, G. M., Janes, P. W., and
Sutherland, R. L. (1996) J. Biol. Chem. 271,
1250212510
473. Feller, S. M., Ren, R., Hanafusa, H., and
Baltimore, D. (1994) Trends Biochem. Sci. 19,
453458
474. Pei, D., Lorenz, U., Klingmller, U., Neel, B.
G., and Walsh, C. T. (1994) Biochemistry 33,
1548315493
References
388. Lefebvre, V., Mchin, M.-C., Louckx, M. P.,
Rider, M. H., and Hue, L. (1996) J. Biol. Chem.
271, 2228922292
389. Quon, M. J., Butte, A. J., Zarnowski, M. J.,
Sesti, G., Cushman, S. W., and Taylor, S. I.
(1994) J. Biol. Chem. 269, 2792027924
390. Kandror, K. V., and Pilch, P. F. (1996) J. Biol.
Chem. 271, 2170321708
390a. Marte, B. M., and Downward, J. (1997) Trends
Biochem. Sci. 22, 355358
390b. Hemmings, B. A. (1997) Science 275, 628630
390c. Chan, T. O., Rittenhouse, S. E., and Tsichlis, P.
N. (1999) Ann. Rev. Biochem. 68, 9651014
390d. Aguirre, V., Uchida, T., Yenush, L., Davis, R.,
and White, M. F. (2000) J. Biol. Chem. 275,
90479054
390e. Nakae, J., Park, B.-C., and Accili, D. (1999) J.
Biol. Chem. 274, 1598215985
391. Kishi, K., Hayashi, H., Wang, L., Kamohara,
S., Tamaoka, K., Shimizu, T., Ushikubi, F.,
Narumiya, S., and Ebina, Y. (1996) J. Biol.
Chem. 271, 2656126568
392. Yamauchi, K., Ribon, V., Saltiel, A. R., and Pessin,
J. E. (1995) J. Biol. Chem. 270, 1771617722
393. Zhang, J., Hiken, J., Davis, A. E., and
Lawrence, J. C., Jr. (1989) J. Biol. Chem. 264,
1751317523
394. Robinson, F. W., Smith, C. J., Flanagan, J. E.,
Shibata, H., and Kono, T. (1989) J. Biol. Chem.
264, 1645816464
395. Najjar, S. M., Choice, C. V., Soni, P., Whitman,
C. M., and Poy, M. N. (1998) J. Biol. Chem. 273,
1292312928
396. Haft, C. R., Klausner, R. D., and Taylor, S. I.
(1994) J. Biol. Chem. 269, 2628626294
397. Safavi, A., Miller, B. C., Cottam, L., and Hersh,
L. B. (1996) Biochemistry 35, 1431814325
398. Kublaoui, B., Lee, J., and Pilch, P. F. (1995) J.
Biol. Chem. 270, 5965
399. Bevan, A. P., Burgess, J. W., Drake, P. G.,
Shaver, A., Bergeron, J. J. M., and Posner, B. I.
(1995) J. Biol. Chem. 270, 1078410791
400. Podlecki, D. A., Smith, R. M., Kao, M., Tsai, P.,
Huecksteadt, T., Brandenburg, D., Lasher, R.
S., Jarett, L., and Olefsky, J. M. (1987) J. Biol.
Chem. 262, 33623368
401. Cherniack, A. D., Klarlund, J. K., Conway, B.
R., and Czech, M. P. (1995) J. Biol. Chem. 270,
14851488
402. Unoue, G., Cheatham, B., and Kahn, C. R.
(1996) J. Biol. Chem. 271, 2820628211
403. Yarden, Y., and Ullrich, A. (1988) Biochemistry
27, 31133119
404. James, R. (1984) Ann. Rev. Biochem. 53, 259292
405. Nilsen-Hamilton, M., ed. (1994) Growth
Factors and Signal Transduction in Development,
Wiley-Liss, New York
406. Claesson-Welsh, L. (1994) J. Biol. Chem. 269,
3202332026
407. Williams, L. T. (1989) Science 243, 15641570
408. Hesketh, R. (1995) The Oncogene Facts Book,
Academic Press, San Diego, California
409. Hunter, T. (1984) Sci. Am. 251(Aug), 7079
410. Marx, J. (1994) Science 266, 19421944
411. Cooper, G. M., ed. (1990) Oncogenes, Jones &
Bartlett, Boston, Massachusetts
412. Bishop, J. M. (1996) FASEB J. 10, 362364
413. Weinberg, R. A. (1983) Sci. Am. 249(Nov),
126142
414. Hickman, C. P. (1973) Biology of the Inverte-
brates, Mosby, St. Louis, Missouri
415. Yarden, Y., Escobedo, J. A., Kuang, W. J.,
Yang-Feng, T. L., Daniel, T. O., Tremble, P. M.,
Chen, E. Y., Ando, M. E., Harkins, R. N.,
Francke, V., Fried, V. A., Ullrich, A., and
Williams, L. T. (1986) Nature (London) 323,
226232
416. Robbins, K. C., Antoniades, H. N., Devare, S.
G., Hunkapillar, M. W., and Aaronson, S. A.
(1983) Nature (London) 305, 605608
417. Yarden, Y., and Peles, E. (1991) Biochemistry
30, 35433550
418. Peles, E., Lamprecht, R., Ben-Levy, R., Tzahar,
E., and Yarden, Y. (1992) J. Biol. Chem. 267,
1226612274
419. Carpenter, G., and Cohen, S. (1990) J. Biol.
Chem. 265, 77097712
420. Suen, T.-C., and Goss, P. E. (2000) J. Biol. Chem.
275, 66006607
421. Stover, D. R., Becker, M., Liebetanz, J., and Lydon,
N. B. (1995) J. Biol. Chem. 270, 1559115597
422. Kauffmann-Zeh, A., Thomas, G. M. H., Ball,
A., Prosser, S., Cunningham, E., Cockcroft, S.,
and Hsuan, J. J. (1995) Science 268, 11881190
422a. Jones, J. T., Ballinger, M. D., Pisacane, P. I.,
Lofgren, J. A., Fitzpatrick, V. D., Fairbrother,
W. J., Wells, J. A., and Sliwkowski, M. X.
(1998) J. Biol. Chem. 273, 1166711674
423. Montelione, G. T., Wthrich, K., and Scheraga,
H. A. (1988) Biochemistry 27, 22352243
424. Schlessinger, J. (1988) Biochemistry 27, 31193123
425. Montelione, G. T., Wthrich, K., and Scheraga,
H. A. (1988) Biochemistry 27, 22352243
426. Chantry, A. (1995) J. Biol. Chem. 270, 30683073
427. Karasik, A., Pepinsky, R. B., Shoelson, S. E.,
and Kahn, C. R. (1988) J. Biol. Chem. 263,
1186211867
428. Kauffmann-Zeh, A., Klinger, R., Endemann,
G., Waterfield, M. D., Wetzker, R., and Hsuan,
J. J. (1994) J. Biol. Chem. 269, 3124331251
429. Kaplan, K. B., Bibbins, K. B., Swedlow, J. R.,
Arnaud, M., Morgan, D. O., and Varmus, H.
E. (1994) EMBO J. 13, 47454756
429a. Williams, J. C., Weijland, A., Gonfloni, S.,
Thompson, A., Courtneidge, S. A., Superti-
Furga, G., and Wierenga, R. K. (1997) J. Mol.
Biol. 274, 757775
430. Okada, M., and Nakagawa, H. (1989) J. Biol.
Chem. 264, 2088620893
431. Buss, J. E., and Sefton, B. M. (1985) Journal of
Virology 53, 712
432. Soriano, P., Montgomery, C., Geske, R., and
Bradley, A. (1991) Cell 64, 693702
433. Lowe, C., Yoneda, T., Boyce, B. F., Chen, H.,
Mundy, G. R., and Soriano, P. (1993) Proc.
Natl. Acad. Sci. U.S.A. 90, 44854489
434. Tezuka, K.-i, Denhardt, D. T., Rodan, G. A.,
and Harada, S.-i. (1996) J. Biol. Chem. 271,
2271322717
435. Bolen, J. B., and Veillette, A. (1989) Trends
Biochem. Sci. 14, 404407
436. Mustelin, T., and Burn, P. (1993) Trends
Biochem. Sci. 18, 215220
437. Vetrie, D., Vorechovsky, I., Sideras, P.,
Holland, J., Davies, A., Flinter, F.,
Hammarstrm, L., Kinnon, C., Levinsky, R.,
Bobrow, M., Smith, C. I. E., and Bentley, D. R.
(1993) Nature (London) 361, 226233
438. Hatakeyama, M., Kono, T., Kobayashi, N.,
Kawahara, A., Levin, S. D., Perlmutter, R. M.,
and Taniguchi, T. (1991) Science 252, 15231528
438a. Zou, X., and Calame, K. (1999) J. Biol. Chem.
274, 1814118144
439. Daley, G. Q., Van Etten, R. A., and Baltimore,
D. (1990) Science 247, 824830
440. Landis, C. A., Masters, S. B., Spada, A., Pace,
A. M., Bourne, H. R., and Vallar, L. (1989)
Nature (London) 340, 692696
441. Barbacid, M. (1987) Ann. Rev. Biochem. 56,
779827
442. Kato, G. J., and Dang, C. V. (1992) FASEB J. 6,
30653072
443. Marcu, K. B., Bossone, S. A., and Patel, A. J.
(1992) Ann. Rev. Biochem. 61, 809860
587
References
475. Ladbury, J. E., Hensmann, M., Panayotou, G.,
and Campbell, I. D. (1996) Biochemistry 35,
1106211069
476. Maignan, S., Guilloteau, J.-P., Fromage, N.,
Arnoux, B., Becquart, J., and Ducruix, A.
(1995) Science 268, 291293
477. Mikol, V., Baumann, G., Zurini, M. G. M., and
Hommel, U. (1995) J. Mol. Biol. 254, 8695
478. Zhou, M.-M., Ravichandran, K. S., Olejniczak,
E. T., Petros, A. M., Meadows, R. P., Sattler,
M., Harlan, J. E., Wade, W. S., Burakoff, S. J.,
and Fesik, S. W. (1995) Nature (London) 378,
584592
479. Bonfini, L., Migliaccio, E., Pelicci, G.,
Lanfrancone, L., and Pelicci, P. G. (1996)
Trends Biochem. Sci. 21, 257261
479a. Nantel, A., Mohammad-Ali, K., Sherk, J.,
Posner, B. I., and Thomas, D. Y. (1998) J. Biol.
Chem. 273, 1047510484
480. Burgering, B. M. T., and Bos, J. L. (1995) Trends
Biochem. Sci. 20, 1822
481. Pumiglia, K. M., LeVine, H., Haske, T., Habib,
T., Jove, R., and Decker, S. J. (1995) J. Biol.
Chem. 270, 1425114254
482. Thompson, M. J., Roe, M. W., Malik, R. K.,
and Blackshear, P. J. (1994) J. Biol. Chem. 269,
2112721135
483. Jhun, B. H., Haruta, T., Meinkoth, J. L.,
Leitner, J. W., Draznin, B., Saltiel, A. R., Pang,
L., Sasaoka, T., and Olefsky, J. M. (1995)
Biochemistry 34, 79968004
484. Jacob, K. K., Ouyang, L., and Stanley, F. M.
(1995) J. Biol. Chem. 270, 2777327779
485. Pause, A., Belsham, G. J., Gingras, A.-C.,
Donz, O., Lin, T.-A., Lawrence, J. C., Jr., and
Sonenberg, N. (1994) Nature (London) 371,
762767
486. Proud, C. G. (1994) Nature (London) 371, 747
748
486a. Wartmann, M., Hofer, P., Turowski, P., Saltiel,
A. R., and Hynes, N. E. (1997) J. Biol. Chem.
272, 39153923
486b. Weng, G., Bhalla, U. S., and Lyengar, R. (1999)
Science 284, 9296
486c. Petosa, C., Masters, S. C., Bankston, L. A.,
Pohl, J., Wang, B., Fu, H., and Liddington, R.
C. (1998) J. Biol. Chem. 273, 1630516310
487. Cobb, M. H., and Goldsmith, E. J. (1995) J.
Biol. Chem. 270, 1484314846
488. Inglese, J., Koch, W. J., Touhara, K., and
Lefkowitz, R. J. (1995) Trends Biochem. Sci. 20,
151156
489. van Biesen, T., Hawes, B. E., Raymond, J. R.,
Luttrell, L. M., Koch, W. J., and Lefkowitz, R.
J. (1996) J. Biol. Chem. 271, 12661269
490. Wu, J., Spiegel, S., and Sturgill, T. W. (1995) J.
Biol. Chem. 270, 1148411488
491. Nurse, P. (2000) Science 289, 17111716
492. Edgar, B. A., and Lehner, C. F. (1996) Science
274, 16461652
493. Kirschner, M. (1992) Trends Biochem. Sci. 17,
281285
494. Horne, M. C., Goolsby, G. L., Donaldson, K.
L., Tran, D., Neubauer, M., and Wahl, A. F.
(1996) J. Biol. Chem. 271, 60506061
Study Questions
1. Illustrated is a generalized metabolic pathway in
which capital letters indicate major metabolites in
the pathway, small letters indicate cofactors and
numbers indicate enzymes catalyzing the reac-
tions.
List and describe four different ways in which the
pathway might be regulated, referring to the
specific enzymes, reactants, and cofactors indi-
cated in the diagram. NOTE: Do not just refer to
four different reactions as possible sites of regula-
tion, but give four different general methods for
regulation.
2. Describe the role of -fructose-2,6-bisphosphate in
the control of the further breakdown and resynthe-
sis of glucose 1-phosphate.
3. It has been proposed that the substrate cycle
involving phosphofructokinase and fructose
bisphosphatase is used by bumblebees to warm
their flight muscles to 30C before flight begins.
Clark et al. (1973) Biochem. J. 134, 589 597 found
maximal rates of catalytic activity for both enzymes
to be about 44 mol / min / g of fresh tissue. In
flying bees glycolysis occurred at a rate of about 20
mol / min / g of tissue with no substrate cycling.
In resting bees at 27C no cycling was detected, but
at 5C substrate cycling occurred at the rate of 10.4
mol / min / g while glycolysis had slowed to 5.8
mol / min / g. If the cycling provides heat to warm
the insect, estimate how long it would take to reach
30C if a cold (5C) bee could carry out cycling at
the maximum rate of 40 mol / min / g and if no
heat were lost to the surroundings.
4. a) Compare the reaction cycle of a small GTPase
(G protein) that is regulated by the actions of
a GTPase-activating protein (GAP) and a
guanine-nucleotide exchange factor (GEF)
with that of a G protein linked to a receptor.
b) Some G proteins have been described as timed
switches and others as triggered switches.
In what ways might these two groups differ?
See Kjeldgaard et al. (1996) FASEB J. 10, 1347
1368.
A B
m n
1
C D
o p
3
2
E F
5 4
495. Tyson, J. J., Novak, B., Odell, G. M., Chen, K.,
and Thron, C. D. (1996) Trends Biochem. Sci. 21,
8996
496. Sherr, C. J. (1996) Science 274, 16721677
497. Hartwell, L. H., and Kastan, M. B. (1994)
Science 266, 18211828
498. Hennig, M., Jansonius, J. N., Terwisscha van
Scheltinga, A. C., Dijkstra, B. W., and
Schlesier, B. (1995) J. Mol. Biol. 254, 237246
499. Elledge, S. J. (1996) Science 274, 16641672
499a. Chin, L., Pomerantz, J., and DePinho, R. A.
(1998) Trends Biochem. Sci. 23, 291296
499b. Nead, M. A., Baglia, L. A., Antinore, M. J.,
Ludlow, J. W., and McCance, D. J. (1998)
EMBO J. 17, 23422352
499c. Bakiri, L., Lallemand, D., Bossy-Wetzel, E.,
and Yaniv, M. (2000) EMBO J. 19, 20562068
500. Kussie, P. H., Gorina, S., Marechal, V.,
Elenbaas, B., Moreau, J., Levine, A. J., and
Pavletich, N. P. (1996) Science 274, 948953
501. Gorina, S., and Pavletich, N. P. (1996) Science
274, 10011005
502. Galaktionov, K., Chen, X., and Beach, D.
(1996) Nature (London) 382, 511517
503. Agarwal, M. L., Taylor, W. R., Chernov, M. V.,
Chernova, O. B., and Stark, G. R. (1998) J. Biol.
Chem. 273, 14
504. Carr, A. M. (2000) Science 287, 17651766
505. Hirao, A., Kong, Y.-Y., Matsuoka, S., Wake-
ham, A., Ruland, J., Yoshida, H., Liu, D.,
Elledge, S. J., and Mak, T. W. (2000) Science
287, 18241827
506. Gatti, A., Li, H.-H., Traugh, J. A., and Liu, X.
(2000) Biochemistry 39, 98379842
588 Chapter 12. Transferring Groups by Displacement Reactions
A. Factors Affecting Rates of a Displacement Reaction
B. Nucleophilic Displacements on Singly Bonded
Carbon Atoms
1. Inversion as a Criterion of Mechanism
2. Transmethylation
3. Kinetic Isotope Effects
4. Glycosyltransferases
Inversion or retention?
Double-displacement mechanisms
Carbocationic intermediates
5. Lysozymes and Chitinases
Catalytic side chain groups
Kinetic isotope effect for lysozyme
Does lysozyme distort its substrate?
Help from a neighboring group
6. Cellulases and Other -Glycosidases
7. Glycogen Phosphorylase
8. Starch-Hydrolyzing Enzymes
C. Displacement Reactions on Carbonyl Groups
1. The Serine Proteases
Serine as a nucleophile
Acyl-enzyme intermediates
Three-dimensional structures
The catalytic cycle
The catalytic triad
The oxyanion hole
Stereoelectronic considerations
pH dependence
Substrate specificity
2. The Cysteine Proteases (Thiol Proteases)
3. N-Terminal Nucleophile Hydrolases and
Related Enzymes
4. The Aspartic Proteases
5. Metalloproteases
6. ATP-Dependent Proteases
7. The Many Functions of Proteases
8. Protease Inhibitors of Animals and Plants
9. Coagulation of Blood
D. Displacement on a Phosphorus Atom
1. Questions about Mechanisms
Geometric complexities
Metaphosphate ions
Coping with negative charges
2. Magnetic Resonance Studies
3. Stereochemistry
4. Phosphatases
5. Ribonucleases
6. Ribonuclease P, Ribozymes, and Peptidyl
Transferase
7. Deoxyribonucleases
8. Mutases
9. Molecular Properties of Kinases
589
590
590
591
592
593
593
595
598
599
599
600
601
601
602
604
605
608
609
609
610
611
613
614
614
616
616
616
618
620
621
625
627
628
629
631
634
637
637
638
638
639
639
639
642
645
647
649
652
653
654
657
658
659
660
660
660
661
662
663
674
594
596
622
630
636
658
603
E. The Adenylate Kinase Fold, the P Loop, and ATPases
and GTPases
F. Displacements on Sulfur Atoms
G. Multiple Displacement Reactions and the Coupling
of ATP Cleavage to Endergonic Processes
1. Transfer of Phospho, Pyrophospho, and Adenylyl
Groups from ATP
2. Acyl Phosphates
3. General Mechanism of Formation of Thioesters,
Esters, and Amides
4. Coenzyme A Transferases
References
Study Questions
Boxes
Box 12-A Carboxymethylation of Proteins
Box 12-B Arsenic
Box 12-C Synthetic Protease Inhibitors
Box 12-D Molecular Mousetraps
Box 12-E Insecticides
Box 12-F The Toxicity of Aluminum
Tables
Table 12-1 Acidic and Basic Catalytic Groups in
a Few Glycosyltransferases
10. Esterases and Lipases
11. Other Acyltransferases
10. Nucleotidyl Transferases
Contents
View of a modified bovine fibrinogen molecule. The 45-nm-long disulfide-
linked dimer is composed of three nonidentical polypeptide chains. The N
termini of the six chains from the two halves come together in the center in
a small globular disulfide knot. The C termini form globular domains at
the ends. The 340-kDa molecule has been treated with a lysine-specific
protease which has removed portions of two chains to give the ~ 285-kDa
molecule whose crystal structure is shown. Arrows point to attached
oligosaccharides. From Brown et al. (2000) Proc. Natl. Acad. Sci. U. S. A. 97,
85 90. Courtesy of Carolyn Cohen.
589
Transferring Groups by Displacement Reactions
12
The majority of enzymes that are apt to be men-
tioned in any discussion of metabolism catalyze nucleo-
philic displacement reactions (Type 1, Table 10-1). These
include most of the reactions by which the energy of
ATP cleavage is harnessed and by which polymers are
assembled from monomers. They include reactions
by which pieces, large or small, are transferred onto
or off of polymers as well as the reactions by which
polymers are cleaved into pieces.
13d
In these reactions a nucleophilic group (a base),
designated B

in Table 10-1, approaches an electrophilic


center, often an electron-deficient carbon or phosphorus
atom. It forms a bond with this atom, at the same time
displacing some other atom, usually O, N, S, or C. The
displaced atom leaves with its bonding electron pair
and with whatever other chemical group is attached,
the entire unit being called the leaving group. This is
designated YH in Table 10-1. Simultaneous or subse-
quent donation of a proton from an acidic group of the
enzyme, or from water, to the O, N, or S atom of the
leaving group is usually required to complete the
reaction. The entering base B, which may or may not
carry a negative charge, must often be generated by
enzyme-catalyzed removal of a proton from the conju-
gate acid BH. If BH is water, the enzyme is a hydrolase;
otherwise, it is called a transferase.
For purposes of classifying the reactions of metab-
olism, in this book the nucleophilic displacements are
grouped into four subtypes (Table 10-1). These are
displacements on (A) a saturated carbon atom, often
from a methyl group or a glycosyl group; (B) a carbonyl
group of an ester, thioester, or amide; (C) a phospho
group; or (D) a sulfur atom. In addition, many enzymes
employ in sequence a displacement on a carbon atom
followed by a second displacement on a phosphorus
atom (or vice versa).
A. Factors Affecting Rates of a Displacement
Reaction
In Chapter 9, the displacement of an iodide ion
from methyl iodide by a hydroxide ion (Eq. 9-76) was
considered. Can we similarly displace a methyl group
from ethane, CH
3
CH
3
, to break the C C bond and
form CH
3
OH? The answer is no. Ethane is perfectly
stable in sodium hydroxide and is not cleaved by a
simple displacement process within our bodies. Like-
wise, long hydrocarbon chains such as those in the
fatty acids cannot be broken by a corresponding process
during metabolism of fatty acids. Not every structure
will allow a nucleophilic displacement reaction to
occur and not every anion or neutral base can act to
displace another group.
At least four factors affect the likelihood of a displace-
ment reaction:
1 3d
(1) The position of the equilibrium in the overall reaction.
An example is provided by the hydrolases that catalyze
cleavage of amide, ester, and phosphodiester linkages
using water as the entering nucleophile. Because
enzymes usually act in an environment of high water
content, the equilibrium almost always favors hydro-
lysis rather than the reverse reactions of synthesis.
However, in a nonaqueous solvent the same enzyme
will catalyze synthetic reactions.
(2) The reactivity of the entering nucleophile. Nucleo-
philic reactivity or nucleophilicity is partly deter-
mined by basicity. Compounds that are strong bases
tend to react more rapidly in nonenzymatic displace-
ment reactions than do weaker bases; the hydroxyl ion,
HO

, is a better nucleophile than COO

. However,
enzymes usually act optimally near a neutral pH.
Under these conditions the COO

group may be
590 Chapter 12. Transferring Groups by Displacement Reactions
more reactive than strongly basic groups such as NH
2
or HO

because at pH 7 the latter groups will be almost


completely protonated and the active nucleophile will
be present in low concentrations.
A second factor affecting nucleophilic reactivity is
polarizability, which is the ease with which the elec-
tronic distribution around an atom or within a chemi-
cal group can be distorted. A large atomic radius and
the presence of double bonds in a group both tend to
increase polarizability. In most cases, the more highly
polarizable a group, the more rapidly it will react in a
nucleophilic displacement, apparently because a polar-
izable group is able to form a partial bond at a greater
distance than can a nonpolarizable group. Thus, I

is
more reactive than Br

, which is more reactive than Cl

.
Polarizable bases such as imidazole are often much
more reactive than nonpolarizable ones such as NH
2
.
Sulfur compounds also tend to have a high nucleo-
philic reactivity. In displacement reactions on carbonyl
groups (reaction type 1B), the less polarizable hard
nucleophiles are more reactive than polarizable ones
such as I

. Attempts have been made to relate quanti-


tatively nucleophilic reactivity to basicity plus polariz-
ability.
1
Certain chemical groups, e.g., those in which an
atom with unpaired electrons is directly bonded to
the nucleophilic center undergoing reaction, are more
reactive than others of similar basicity. This effect
has been invoked to explain the high reactivity of the
poisons hydroxylamine (NH
2
OH) and cyanide ion
4
and other puzzling results.
1
(3) The chemical nature of the leaving group that is
displaced. The chemistry of the leaving group affects
both the rate and the equilibrium position in nucleo-
philic displacements. The leaving group must accom-
modate a pair of electrons and often must bear a
negative charge. A methyl group displaced from
ethane or methane as CH
3

would be an extremely
poor leaving group; the pK
a
of methane as an acid
5
has been estimated as 47. Iodide ion is a good leaving
group, but F

is over 10
4
times poorer.
5
In an aqueous
medium, phosphate is a much better leaving group
than OH

, and pyrophosphate and tripolyphosphate


are even better.
(4) Special structural features present in the substrate.
Enzymes are usually constructed so that they recognize
unique features in substrates. As a consequence, they
have many ways of lowering the energy of the transi-
tion state and increasing the apparent nucleophilic
reactivity.
B. Nucleophilic Displacements on Singly
Bonded Carbon Atoms
The enzymatic counterpart of Eq. 9-76, the dis-
placement of I

from a saturated carbon atom by HO

,
is catalyzed by the haloacid dehalogenases of soil
pseudomonads (Eq. 12-1).
6 8
Even the poor leaving
group F

can be displaced by OH

in the active site of


these enzymes.
HO

+ F CH
2
COO

HO CH
2
COO

+ F

1. Inversion as a Criterion of Mechanism


An interesting result was obtained when a haloacid
dehalogenase was tested with a substrate containing a
chiral center.
6
Reaction of L-2-chloropropionate with
hydroxyl ion gave only D-lactate, a compound with a
chirality opposite to that of the reactant. A plausible
explanation is that the hydroxyl ion attacks the central
carbon from behind the chlorine atom (Eq. 12-2). The
resulting five-bonded transition state (center) loses a
chloride ion to form the product D-lactate in which
inversion of configuration has occurred. Inversion
always accompanies single displacement reactions in which
bond breaking and bond formation occur synchronously, as
in Eq. 12-2. This is true for both enzymatic and nonen-
zymatic reactions. However, the occurrence of inver-
sion does not rule out more complex mechanisms.
Indeed, in this case the displacing group is evidently
not HO

but a carboxylate group from the enzyme.


In one of the haloacid dehalogenases, a 232-residue
protein for which the three-dimensional structure is
known,
8,9
Asp 10 is in a position to carry out the initial
attack which would give an enzyme-bound intermedi-
ate with an ester linkage:
(12-1)
C Cl
H H
3
C
OOC

HO
l-2-Chloropropionate
HO Cl
C
O
O

H
C
H
3

Transition state
C
Enzyme C
O
O
HO in above structures is replaced by
in haloacid dehalogenases
Cl
C HO
H
CH
3
COO

d-Lactate

(12-2)
591
Hydrolytic cleavage, as indicated in the diagram, yields
the product D-lactate.
7
Thus, we have a direct displace-
ment with inversion followed by an additional hydro-
lytic step. This is an example of covalent catalysis,
the enzyme providing a well-oriented reactive group
instead of generating a hydroxyl group from a bound
water molecule.
Related haloalkane dehalogenases as well as
epoxide hydrolases and a large superfamily of other
enzymes utilize similar mechanisms.
10,11,11a
In the
active site of a haloalkane dehalogenase from Xantho-
bacter (Fig. 12-1) the carboxylate of Asp 124 acts as the
attacking nucleophile that displaces Cl

from the
substrate dichloroethane, which is held in a small
cavity with the aid of two tryptophan indole rings.
12 14
However, the histidineaspartate pair and the bound
water molecule shown in Fig. 12-1 are essential for the
subsequent hydrolysis of the intermediate ester. (see
also Fig. 12-11).
15
The substrate shown in Fig. 12-1 is
1,2-dichloroethane, a widespread environmental pol-
lutant that is not known to occur naturally. An inter-
esting question considered by Pries et al.
16
is how this
dehalogenase has evolved in the years since 1922
when industrial production of dichloroethane began.
2. Transmethylation
Nucleophilic attack on a methyl group or other
alkyl group occurs most readily if the carbon is attached
to an atom bearing a positive charge, for example, the
sulfur atom of a sulfonium ion such as that present in
S-adenosylmethionine (abbreviated AdoMet or SAM).
The ensuing transmethylation reaction results in the
transfer of the methyl group from the sulfur to the
attacking nucleophile (Eq. 12-3). Transmethylation is
an important metabolic process by which various
oxygen, nitrogen, and other atoms at precise positions
in proteins, nucleic acids, phospholipids, and other
small molecules are methylated.
17
The methyl group
donor is usually S-adenosylmethionine. This com-
pound has two chiral centers, one at the -carbon of
the amino acid and one at the sulfur atom, with an
unshared electron pair serving as the fourth group
around the S atom. In the naturally occurring Ado-
Met both centers have the S configuration.
18
The
reaction of Eq. 12-3, which is catalyzed by catechol
O-methyltransferase (COMT), inactivates the neuro-
transmitters adrenaline, dopamine, and related com-
pounds by methylation. When the substrate contains
a chiral methyl group ( C
1
H
2
H
3
H; see also Chapter 13),
the inversion of the methyl group expected for a simple
S
N
2-like reaction is observed.
19
Structural features similar to those of COMT
20
are
found in glycine N-methyltransferase
21
and guanidino-
acetate methyltransferase
22
from liver, and transferases
that place methyl groups on N-6 of
adenine
23 25a
and N-4 of cytosine or
on C-5 of cytosine
26
in nucleic acids.
27
A stereoscopic view of the active site
of glycine N-methyltransferase is
shown in Fig. 12-2. An acetate ion
present in the active site in the crystal
has been converted into glycine by
computer-assisted modeling. The
amino group of the bound glycine
is adjacent to the methyl group of
AdoMet. A nearby glutamic acid
side chain (E15) may have removed
a proton from the glycine dipolar
ion to create the free NH
2
group
required for the reaction. In COMT
a magnesium Mg
2+
ion binds to the
two aromatic hydroxyl groups of
the substrate and helps to hold it
correctly in the active site.
2829a
In the case of the more difficult
C-methylation of uracil 54 in transfer
RNA, an SH group from Cys 324
of the methyltransferase adds to the
C-6 position of U54 of the substrate,
which may be any of the E. coli
tRNAs (Eq. 12-4, step a) .
30
In the
Asp 10 CH
2
C
O
O C
COO
H
CH
3

OH
2
N
H
Cl Cl
N
H
N
H
O
O
O
N
N H O O
H
O
H
W175
W125

D260

H289
D124
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
Figure 12-1 The active site structure of haloalkane dehalogenase from
Xanthobacter autotrophicus with a molecule of bound dichloroethane. See
Pries et al.
13
The arrows illustrate the initial nucleophilic displacement. The
D260 H289 pair is essential for the subsequent hydrolysis of the intermedi-
ate ester formed in the initial step.
592 Chapter 12. Transferring Groups by Displacement Reactions
H
3
N
HO
H
OH
O
+
H
Adrenaline
COMT
O
OH OH
Adenine
S
COO

H
NH
3
+
S-Adenosylhomocysteine
(inactive)
H
3
N
HO
H
OH
O
+
CH
3
O
OH OH
Adenine
S
COO

H
NH
3
C
3
H
2
H
1
H
+
+
:
S-Adenosylmethionine
(AdoMet or SAM)
H
+
(12-3)
adduct carbon atom C-5 acquires substantial nucleo-
philic character which permits the transfer of the
methyl group in step b. The adduct breaks up (step c)
and the product is released.
Methylation of nucleic acids is considered further
in Chapers 27 and 28. Methylation of carboxyl groups
of certain proteins regulates motion of bacterial flagella
(Chapter 19) and other aspects of metabolism
31
while
methylation of isomerized aspartyl residues is part of
a protein repair process (Box 12-A). Methyl groups are
usually transferred from S-adenosylmethionine, but
sometimes from a folic acid derivative (Chapter 15)
or from a cobalt atom of a corrin ring (Chapter 16).
3. Kinetic Isotope Effects
An S
12
C bond breaks a little faster than an S
13
C
bond in a nucleophilic displacement reaction. This
primary kinetic isotope effect (KIE)
3a,3b,31ad
is
usually discussed first for breakage of a C H bond.
In a linear transition state, in which the C H bond is
being stretched, then cleaved, the difference between
the transition state barrier for a C
1
H bond and a C
2
H
bond is thought to arise principally from a difference
in the energies of the C H stretching vibration.
3a
This
vibrational energy, in the ground state of a molecule
(the zero-point energy) is equal to 1/ 2 h
0
where
0
may be observed in the infrared absorption spectrum
(see Fig. 23-2). For a C
2
H bond, with a stretching
vibration at a wave number of ~ 2900 cm
1
, the zero
point energy is about + 17.4 kJ mol
1
. For a C
2
H bond,
with a stretching wave number of ~ 2200 cm
1
, it is
about + 17.4 kJ mol
1
. This difference is a result of the
difference in mass of
1
H (1.67 x 10
24
g) and
2
H (2.34 x
10
24
g). The isotope effect arises because the vibration
occurs along the axis of the bond being broken, and
the vibrational energy is converted into translational
motion along the reaction coordinate, in effect lower-
ing the transition state barrier by G
0
. The difference
in this effect between
1
H and
2
H (G
0
) gives the ratio
of expected rate constants as follows:
For heavier nuclei, such as C, N, O the kinetic
isotope effect is much smaller.
When
13
C was introduced into the methyl
group of S-adenosylmethionine and its rate
of reaction with catechol O-methyltransfer-
ase was compared with that of the normal
12
C-containing substrate, the expected effect
on V
max
, expressed as a first-order rate constant,
was seen: k
12
/k
13
= 1.09 0.05. This effect is
small but it can be measured reliably and
establishes that the methyl transfer step rather
than substrate binding, product release, or a
conformational change in the protein is rate
limiting.
32
Substitution of
1
H by
2
H in the CH
3
group has a larger effect. This secondary
kinetic isotope effect (or -deuterium
HN
N O
O
HN
N O
O

S-Enzyme
H
H
3
C S
HN
N O
O
S-Enzyme
tRNA
tRNA
HN
N O
O
tRNA
S-Enzyme
H
5
+
H
CH
3
CH
3
5
6
tRNA
AdoMet
m
5
U

c
b
a
(12-4)
k
CH
k
CD
e
G
0
/RT
= = 7
593
Fig. 12-2 S-Adenosylmethionine
(AdoMet, solid bonds) bound in
the active site of glycine N-methyl-
transferase together with an acetate
ion bound in the glycine site. Gly-
cine was built by attaching an
amino group (open bond) to the
acetate (solid bonds). Possible
polar interactions (O O and O N
< 0.31 nm and O S < 0.4 nm) are
indicated by dotted lines. Tyrosine
residues located at the inner surface
of the active site are also shown.
From Fu et al.
21
effect) arises because of small differences in the vibra-
tional energies of the methyl group resulting from the
differences in mass of
1
H and
2
H. It often leads to a
more rapid reaction for the
1
H-containing substrates.
In a model nonenzymatic displacement reaction of a
similar type
32
the ratio of rate constants was k
1H
/k
2H
=
1.17 0.02. However, for COMT an inverse -deuteri-
um effect was seen: k
(C
1
H
3
)
/ k
(C
2
H
3
)
= 0.83 0.05. Theo-
retical calculations suggested that such an effect might
be observed if the enzyme compresses an S
N
2-like
transition state, shortening the bonds from the central
carbon atom to both the oxygen atom of the entering
nucleophile and the sulfur atom of the leaving
group.
33,34
However, more recent calculations suggest
that it is difficult to draw such conclusions from sec-
ondary isotope effects.
35,36
Calculations by Zheng,
Kahn, and Bruice predict that in the gas phase the two
substrates, upon collision, will react with a very low
energy barrier.
29,29a
For this enzyme, as for the haloal-
kane dehalogenase (Fig. 12-1), it has been concluded
that the enzyme excludes water from the active site
and binds the two substrates very close together and
in a correct orientation for reaction. The computations
predicted secondary isotope effects for the enzymatic
reaction similar to those measured by Hegazi et al.
32
suggesting that the transition states for the enzymatic
reactions closely resemble those for the gas phase.
4. Glycosyltransferases
The polarization of a single C O bond in an ether
is quantitatively much less than that of the C S
+
bond
of S-adenosylmethionine, and simple ethers are not
readily cleaved by displacement reactions. However,
glycosides, which contain a carbon atom attached to two
oxygen atoms, do undergo displacement reactions which
lead to hydrolysis, phosphorolysis, or glycosyl
exchange. The corresponding enzymes are glycosyl-
hydrolases, phosphorylases, and glycosyltransferases.
Two characteristics are commonly used to classify
glycosylhydrolases that act on polysaccharides:
Endoglycanases cut at random locations within the
chains of sugar units, while exoglycanases cut only
at one end or another, usually at the nonreducing
end. Inverting enzymes invert the configuration at
the anomeric carbon atom which they attack, while
retaining enzymes do not. Although simple glyco-
sides undergo acid-catalyzed hydrolysis readily, un-
catalyzed hydrolysis is extremely slow, the estimated
first order rate constant
37
at 25C being about 2 x 10
15
s
1
. Polysaccharides may be the most stable of all
bioploymers.
Glycosyltransferases are numerous. For example,
the amino acid sequences for about 500 glucosidases,
which hydrolyze linkages between glucose residues,
have been determined and this one group of enzymes
has been classified into over 60 families
38,39
and eight
larger groups.
40
Many three-dimensional structures
have been reported and numerous studies of the reac-
tion mechanism for both enzymatic and nonenzymatic
hydrolysis of glycosides have been conducted. Most
of the experimental results have been carefully verified.
Nevertheless, they serve to illustrate how difficult it is
to understand how enzymes catalyze this simple type
of reaction.
41
Inversion or retention? Equation 12-5 pictures
the reaction of a glycoside (such as a glucose unit at
HO
O
OH
O
H HO
OH
R

O
Y
BH
HO
O
OH
H
O
HO
OH

Y
B
HOR

(12-5)
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
594 Chapter 12. Transferring Groups by Displacement Reactions
BOX 12-A CARBOXYMETHYLATION OF PROTEINS
Methylation and demethylation of the
carboxyl groups of the side chains of specific
glutamyl residues play a role in bacterial
chemotaxis (Chapter 19, Section A). How-
ever, similar functions in eukaryotes have
not been found. There is a specific methyl-
transferase that methylates C-terminal
carboxyl groups on prenylated and some-
times palmitoylated peptides.
a
Another
type of carboxylmethyltransferase acts on
only a small percentage of many proteins
and forms labile methyl esters. These
methyltransferases have a dual specificity,
acting on l-isospartyl residues and usually
also on d-aspartyl residues. Both of these
amino acids can arise from deamidation of
asparagine, especially in Asn-Gly sequences
(Eq. 2-24 and steps a and b of following
scheme).
b g
A similar isomerization of
aspartyl residues occurs more slowly. Asn-
Gly sequences are present in many proteins
and provide weak linkages whose isomer-
ization is an inevitable aspect of aging.
e,h
Furthermore, the proton of the cyclic imide
(green box) is more readily dissociated than
in a standard peptide, leading to racemiza-
tion (step c). The isoaspartyl and D-aspartyl
residues provide kinks in the peptide chain
which may interfere with normal function
and turnover.
The L-isoaspartyl / D-aspartyl methyl-
transferase (step d), which is especially
abundant in the brain,
i,j
provides for partial
repair of these defects.
k o
The methyl esters
of isoaspartyl residues readily undergo
demethylation with a return to a cyclic imide (step e).
The cyclic imide is opened hydrolytically (step c) in
part to an isoaspartyl residue, but in part to a normal
aspartyl form. The combined action of the carboxyl-
methyltransferase and the demethylation reaction
tends to repair the isomerized linkages. Methylation
of D- aspartyl residues returns them to the cyclic imide
(steps f, e), allowing them to also return to the normal
L configuration. Nevertheless, the protein will have
a different net charge than it did originally and must
be considered a new modified form.
a
Dai, Q., Choy, E., Chiu, V., Romano, J., Slivka, S. R., Steitz, S. A.,
Michaelis, S., and Philips, M. R. (1998) J. Biol. Chem. 273, 15030
15034
b
Geiger, T., and Clarke, S. (1987) J. Biol. Chem. 262, 785 794
c
Tyler-Cross, R., and Schirch, V. (1991) J. Biol. Chem. 266, 22549
22556
d
Brennan, T. V., and Clarke, S. (1993) Protein Sci. 2, 331 338
Asp-Gly peptide
AdoMet
(on d-aspartyl
peptides)
Asn-Gly in a peptide
AdoMet,
l-Isoaspartyl/
d-aspartyl protein
methyltransferase
Partially
racemized
H
3
N
Cyclic imide
H
2
O
Partially
racemized
d
e
f
C
N
C
C
N
CH
2
C
N
O
O
O
H
H H
H
2
C
C
NH2
O
H
H
+
C H
2
C
C
C
N
O
O
CH
2
N
H
H
C
COO

N
C H
2
C
N
O
CH
2
H
H H
C
N
C
C
N
O
O
CH
2
H
2
C
H
H
COO

H
C
C O
N
C H
2
C
N
O
CH
2
O
CH
2
H
H H
H
2
O
CH
3
OH
c
b
a
e
Paranandi, M. V., Guzzetta, A. W., Hancock, W. S., and Aswad,
D. W. (1994) J. Biol. Chem. 269, 243 253
f
Donato, A. D., Ciardiello, M. A., de Nigris, M., Piccoli, R.,
Mazzarella, L., and DAlessio, G. (1993) J. Biol. Chem. 268, 4745
4751
g
Tomizawa, H., Yamada, H., Ueda, T., and Imoto, T. (1994)
Biochemistry 33, 8770 8774
h
Man, E. H., Sandhouse, M. E., Burg, J., and Fisher, G. H. (1983)
Science 220, 1407 1408
i
Orpiszewski, J., and Aswad, D. W. (1996) J. Biol. Chem. 271,
22965 22968
j
Najbauer, J., Orpiszewski, J., and Aswad, D. W. (1996) Biochem-
istry 35, 5183 5190
k
Johnson, B. A., Murray, E. D. J., Clarke, S., Glass, D. B., and
Aswad, D. W. (1987) J. Biol. Chem. 262, 5622 5629
l
Brennan, T. V., Anderson, J. W., Jia, Z., Waygood, E. B., and
Clarke, S. (1994) J. Biol. Chem. 269, 24586 24595
m
Mudgett, M. B., and Clarke, S. (1993) Biochemistry 32, 11100
11111
n
Mudgett, M. B., and Clarke, S. (1994) J. Biol. Chem. 269, 25605
25612
o
Aswad, D. W., ed. (1995) Deamidation and Isoaspartate Formation
in Peptides and Proteins, CRC Press, Boca Raton, Florida
595
the end of a starch chain) with a nucleophile Y O

as
the displacing group. An enzyme-bound acidic group
BH is shown assisting the process. Inversion of
configuration with formation of a product of the
configuration at the anomeric carbon atom would be
predicted and is observed for many of these enzymes.
However, many others do not cause inversion.
41,42
An example is the reaction catalyzed by sucrose
phosphorylase from Pseudomonas saccharophila:
Sucrose (an -glucoside of fructose) +
inorganic phosphate (P
i
)
-D-glucose 1-phosphate + fructose
Two possible explanations for the lack of inversion
during this reaction are that the enzyme acts by a
double-displacement reaction or through a stabilized
carbocationic intermediate. Let us consider these
possibilities in turn.
Double-displacement mechanisms. In a double-
displacement mechanism sucrose phosphorylase would
catalyze two consecutive single displacements, each
with inversion. A nucleophilic group of the enzyme
would react in Eq. 12-7, step a. In step b, a phosphate
would react to regenerate the enzyme with its free
nucleophilic group B

.
a
Fructose
b
HO
O
HO
OH O
O
OH
OH
HO
CH
2
OH
2

B-Enzyme

H
H
+
HOCH
2
HO
O
HO
O
B-Enzyme

O
P
OH
O O

Gycosyl enzyme
HOCH
2

H
H
HO
O
HO
OH
HOCH
2 H

OPO
3
H

Glucose 6-P

B-Enzyme
Four kinds of experiments were used to identify
this double-displacement mechanism.
(1) Kinetics. In a double-displacement mechanism
the enzyme shuttles between free enzyme and the inter-
mediate carrying the substrate fragment (here, the
glycosyl enzyme). With sucrose phosphorylase the
maximum velocity varies with the concentrations of
sucrose and HPO
4
2
in the characteristic fashion ex-
pected for this ping-pong mechanism (Eq. 9-47).
43
(2) Exchange reactions. In a double-displacement
mechanism sucrose containing
14
C in the fructose por-
tion of the molecule should react with free enzyme E
to form glycosyl enzyme and free radioactive fructose
(Eq. 12-8). The
14
C-containing groups are designated
here by the asterisks.
If a very low molar concentration of enzyme is present,
and a large excess of nonradioactive fructose is added,
the enzyme will catalyze no net reaction but will
change back and forth repeatedly between the free
enzyme and glucosyl enzyme. Each time, in the reverse
reaction, it will make use primarily of unlabeled fruc-
tose. The net effect will be catalysis of a sucrosefructose
exchange:
This exchange reaction, as well as other predicted
exchanges, has been observed.
44
Although the exchange
criterion of the mechanism is often applied to enzy-
matic processes, the observation of exchange reactions
does not prove the existence of a covalently bound
intermediate. Furthermore, enzymes using double-
displacement mechanisms may not always catalyze
the expected exchanges.
(3) Arsenolysis. Sucrose phosphorylase also cata-
lyzes the cleavage of sucrose by arsenate and promotes
a rapid cleavage (arsenolysis) of glucose 1-phosphate
to free glucose. This reaction is evidently a result of a
displacement by arsenate on a glycosyl enzyme inter-
mediate. The resulting unstable glucose 1-arsenate
(see Box 12-B) is hydrolyzed rapidly. Arsenolysis is a
general way of trapping reactive enzyme-bound inter-
mediates that normally react with phosphate groups.
Arsenate is one of many substrate analogs that can
be used to siphon off reactive enzyme-bound inter-
mediates into nonproductive side paths.
(4) Identification of glycosyl-enzyme intermediates.
Studies with pure enzymes often make it possible to
confirm directly the existence of enzyme-bound inter-
mediates. The intermediates detected are frequently
glycosyl esters of glutamate or aspartate side chain
d-Glucosyl-fructose* + E glucosyl-E + fructose*
(Sucrose)
(12-8)
Sucrose* + fructose sucrose + fructose*
(12-9)
(12-7)
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
(12-6)
596 Chapter 12. Transferring Groups by Displacement Reactions
Arsenate, AsO
4
3
, is chemically similar to phos-
phate in size and geometry and in its ability to enter
into biochemical reactions. However, arsenate esters
are far less stable than phosphate esters. If formed
in the active site of an enzyme, they are quickly
hydrolyzed upon dissociation from the enzyme.
This fact accounts for some of the toxicity of arsenic
compounds.
a
Arsenate will replace phosphate in all phospho-
rolytic reactions, e.g., in the cleavage of glycogen
by glycogen phosphorylase, of sucrose by sucrose
phosphorylase, and in the action of purine nucleo-
side phosphorylase.
b
Glucose 1-arsenate or ribose-
1-arsenate is presumably a transient intermediate
which is hydrolyzed to glucose. The overall process
is called arsenolysis. Another reaction in which
arsenate can replace phosphate is the oxidation of
glyceraldehyde 3-phosphate in the presence of P
i
to form 1,3-bisphosphoglycerate:
The subsequent transfer of the 1-phospho group to
ADP is an important energy-yielding step in metab-
olism (Chapter 12). When arsenate substitutes for
phosphate the acyl arsenate (1-arseno-3-phospho-
glycerate) is hydrolyzed to 3-phosphoglycerate.
As a consequence, in the presence of arsenate oxida-
tion of 3-phosphoglyceraldehyde continues but
ATP synthesis ceases. Arsenate is said to uncouple
phosphorylation from oxidation. Arsenate can also
partially replace phosphate in stimulating the respira-
tion of mitochondria and is an uncoupler of oxida-
tive phosphorylation (Chapter 18). Enzymes that
normally act on a phosphorylated substrate will
usually catalyze a slow reaction of the correspond-
ing unphosphorylated substrate in the presence of
arsenate. Apparently, the arsenate ester of the sub-
strate forms transiently on the enzyme surface,
permitting the reaction to occur.
Arsenite is noted for its tendency to react rapidly
with thiol groups,
c
especially with pairs of adjacent
(vicinal) or closely spaced SH groups
d
as in lipoic
acid. By blocking oxidative enzymes requiring
BOX 12-B ARSENIC
lipoic acid (Chapter 15), arsenite causes the accumu-
lation of pyruvate and of other 2-oxo acids. Similar
chemistry underlies the action of the mustard gas
Lewisite, which also attacks lipoic acid.
de
Many, perhaps all, organisms have enzyme
systems for protection against arsenic compounds.
O
O
O
P
O
CH
2
C
C
O
P
H OH
O
OH
O
O

1,3-Biphosphoglycerate
This phospho group is normally
transferred to ADP to form ATP
COOH
SH SH
Lipoic acid
S
As
S
COOH
OH
H
+
2H
2
O
H
2
ASO
3

O
CH
2
O
CH
2
C
CH
2
H OH
As
CH
3
O
CH
3
OH OH
+

SO
3

2-Hydroxy-3-sulfopropyl-5-deoxy-5-
(dimethylarseno)-ribofuranoside
CHCl
HC
AsCl
2
Lewisite
In E. coli arsenate is reduced to arsenite by a glutare-
doxin- and NADH-dependent system.
e g
The arsenite
as well as antimonite and tellurite are pumped out
by an ATP-dependent transporter. The genes for
reductase, periplasmic-binding protein, and trans-
porter components are encoded in a conjugative plas-
mid.
h,i
A quite similar system functions in yeast.
j
Marine algae as well as some higher aquatic
plants detoxify and excrete arsenate by conversion
to various water-soluble organic forms such as
trimethyarsonium lactic acid (Chapter 8) and the
following ribofuranoside.
a
Arsenic-containing phospholipids are also formed
(Chapter 8).
k
Arsenic is present at high levels in some soils
and contamination of drinking water with arsenic is
a major problem is some areas of the world. In West
Bengal, India, millions of people drink contaminated
well water. As a result hundreds of thousands have
developed debilitating nodular keratoses on their
feet.
l,m
The problem is made worse by the increasing
597
BOX 12-B (continued)
use of fresh water for irrigation and the difficulty of
removing arsenic contamination.
Although it is most known for its toxicity, arsenic
may be an essential nutrient. Data from feeding of
chicks, goats, rats, and miniature pigs suggest a
probable human need for arsenic of ~12 g / day.
n
Compounds of arsenic have been used in medi-
cine for over 2000 years, but only in the past century
a
Knowles, F. C., and Benson, A. A. (1983) Trends Biochem. Sci. 8,
178180
b
Kline, P. C., and Schramm, V. L. (1995) Biochemistry 34, 1153
1162
c
Lam, W.-C., Tsao, D. H. H., Maki, A. H., Maegley, K. A., and
Reich, N. O. (1992) Biochemistry 31, 1043810442
d
Li, J., and Pickart, C. M. (1995) Biochemistry 34, 1582915837
de
Ord, M. G., and Stocken, L. A. (2000) Trends Biochem. Sci. 25,
253256
e
Silver, S., Nucifora, G., Chu, L., and Misra, T. K. (1989) Trends
Biochem. Sci. 14, 7680
f
Ji, G., Garber, E. A. E., Armes, L. G., Chen, C.-M., Fuchs, J. A.,
and Silver, S. (1994) Biochemistry 33, 72947299
g
Rosen, B. P., Weigel, U., Karkaria, C., and Gangola, P. (1988) J.
Biol. Chem. 263, 30673070
h
Gladysheva, T. B., Oden, K. L., and Rosen, B. P. (1994) Biochem-
istry 33, 72887293
i
Chen, Y., and Rosen, B. P. (1997) J. Biol. Chem. 272, 1425714262
j
Wysocki, R., Bobrowicz, P., and Ulaszewski, S. (1997) J. Biol.
Chem. 272, 3006130066
k
Cooney, R. V., Mumma, R. O., and Benson, A. A. (1978) Proc.
Natl. Acad. Sci. U.S.A. 75, 42624264
l
Bagla, P., and Kaiser, J. (1996) Science 274, 174175
m
Saha, D. P., and Subramanian, K. S. (1996) Science 274, 1287
1288
n
Nielsen, F. H. (1991) FASEB J. 5, 26612667
carboxyl groups. Such an intermediate, fructofuranosyl-
enzyme, was identified tentatively for sucrose phospho-
rylase and also for a related levan sucrase.
45
Recently,
identification of glycosyl-enzyme intermediates has been
accomplished for many other glycosyltransferases.
Among these are human pancreatic and salivary
-amylases,
42,46
-glucosidases, and some cellulases
and xylanases.
For these hydrolytic enzymes the glycosyl-enzyme
would be attacked by a hydroxyl ion derived from
H
2
O, whose deprotonation would presumably be
assisted by the conjugate base (B: in Eq. 12-5) of
the catalytic acidic group.
A convenient way to form and identify covalently
linked glucosyl-enzyme intermediates, developed by
Withers and coworkers, employs enzyme-activated
inhibitors such as 2-deoxy-2-fluoroglycosyl fluorides
(Eq. 12-10).
42,47,48
The 2-F substituent greatly decreases
both the rate of formation of a glycosyl-enzyme and its
rate of breakdown by hydrolysis or transglycosylation.
This may be in part because these compounds lack the
2 OH group which helps to stabilize, by hydrogen
bonding, the complexes of normal substrates. A second
factor is the high electronegativity of F versus OH,
which leads to significant loss of stability for a transi-
tion state in which the anomeric carbon atom carries a
significant amount of positive charge (see next section).
Having a good leaving group, such as fluorine or 2,4-
dinitrophenyl, the compounds react rapidly to give
stable glucosyl-enzymes which can be characterized.
In the example shown in Eq. 12-10, the mannosyl-
enzyme was identified by the chemical shifts and line-
widths of the
19
F resonances of the intermediates and
the anomeric configuration was established. More
recently, mass spectrometry has been used.
49
For
example, a maltotriosyl-enzyme intermediate in the
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
H
2
N As OH
O
O

Sodium arsanilate
Na
+
HO As O
Oxophenarsine
H
2
N
H
2
N
HO As As
NH
2
OH 2HCl
Salvarsan
Substitution by
CH
2
CONH
2
yields
tryparsamide, a
more effective anti-
trypanosome drug
Two of Ehrlich's
anti-syphilis drugs
have specific arsenicals been created as drugs. In
1905, it was discovered that sodium arsanilate is
toxic to trypanosomes. The development by P.
Ehrlich of arsenicals for the treatment of syphilis
(in 1909) first focused attention on the possibility of
effective chemotherapy against bacterial infections.
O
CH
2
O
CH
2
OH
C CH
2
Protein
O
HO
OH
HO

(Glu or Asp)
Fructofuranosyl - enzyme
598 Chapter 12. Transferring Groups by Displacement Reactions
action of glycogen-debranching enzyme was identified
by separation of an active site peptide by HPLC fol-
lowed by mass spectrometry (Chapter 3).
50
Carbocationic intermediates. In a second mech-
anism of nucleophilic displacement the leaving group
departs (often in a protonated form) before the entering
nucleophile reacts.
A carbocation is formed as shown in Eq. 12-11, which
represents just one-half of the overall displacement
reaction. In the common terminology of physical
organic chemistry this is an S
N
1 reaction rather than an
S
N
2 reaction of the kind shown in Eqs. 12-3 and 12-5.
This terminology is not quite appropriate for enzymes
because the breakdown of ES complexes to product is
usually a zero-order process and the numbers 1 and 2
in the symbols S
N
1 and S
N
2 refer to the order or mole-
cularity of the reaction. It is better to speak of S
N
1-like
or S
N
2-like reactions.
The carbocation in Eq. 12-11 is depicted as a
resonance hybrid of two forms. One of these is an
oxocarbenium (or oxonium) ion which contains a
double bond between carbon and oxygen.
50a
This
double-bonded structure can be visualized as arising
from the original structure by an internal displacement
or elimination by the unshared electrons on oxygen, as
indicated by the small arrows. In the oxocarbenium
ion, the ring atoms C-2, C-1, and C-5 and the oxygen
atom are almost coplanar and the ring conformation
is described as half-chair or sofa. As pointed out in
Chapter 9, the theory of stereoelectronic control pre-
dicts that elimination of the group OR of Eq. 12-11
should occur most easily if a lone pair of electrons on
the ring oxygen atom are antiperiplanar to the O R
bond. This is impossible for a -glycoside with a ring
in the chair conformation shown in Eq. 12-11. This fact
suggested that enzymes cleaving -glycosides may
preferentially bind substrate with the appropriate
sugar ring in a less stable half-chair or flexible boat
conformation prior to bond cleavage (Eq. 12-12).
51,52
This would allow an unshared pair of electrons on
oxygen antiperiplanar to the C-1 to O R bond to par-
ticipate in the elimination reaction as is indicated in
the following diagrams.
On the other hand, Bennet and Sinnott provided evi-
dence that an antiperiplanar lone electron pair is not
needed in acid-catalyzed cleavage of glycosides via a
carbocationic intermediate.
53,54
Theories of stereoelec-
tronic control must be applied to enzymes with caution!
O
O
R
H
B Enzyme
C
O
O
C
O
O
C
O
O
H O R
+

Enzyme

Enzyme
C
O
Enzyme
+

H
Oxocarbenium ion, a stabilized carbocation
C
O
H
B-Enzyme

B-Enzyme

(12-11)
O
O
R
HO
6
Half chair
O
HO
O
R
Boat
1
2
4
6
O
HO
O
R
a
b
(12-12)
HO
O
HO
HOH
2
C
F
H
F
HO
O
HO
HOH
2
C

2-Deoxy-2-F--d-mannosyl fluoride
H
Y
Y

Enzyme

Enzyme
4
(12-10)
599
5. Lysozymes and Chitinases
Polysaccharide chains in the
peptidoglycan layer (Fig. 8-29) of
the cell walls of bacteria are attacked
and cleaved by lysozymes,
55
en-
zymes that occur in tears and other
body secretions and in large amounts
in egg white. Some bacteria and
fungi, and even viruses, contain
lysozymes.
56
Their function is usually
to protect against bacteria, but lyso-
zyme of phage T4 is a component of
the baseplate of the virus tail (Box 7-C).
Its role is to cut a hole in the bacterial
cell wall to permit injection of the
virus own DNA. Egg white lyso-
zyme, the first enzyme for which a
complete three-dimensional struc-
ture was determined by X-ray dif-
fraction,
55
is a 129-residue protein.
The active site is in a cleft between a
large domain with a nonpolar core
and a smaller -sheet domain that
contains many hydrogen-bonded polar side chains
(Figs. 12-3, 12-4). Human lysozyme has a similar
structure and properties.
57 59
The T4 lysozyme has an
additional C-terminal domain whose function may be
to bind the crosslinking peptide of the E. coli peptido-
glycan. Goose lysozyme is similar in part to both hen
lysozyme and T4 lysozyme. All three enzymes, as
well as that of our own tears, may have evolved from
a common ancestral protein.
60
On the other hand,
Streptomyces erythaeus has developed its own lysozyme
with a completely different structure.
61
An extensive
series of T4 lysozyme mutants have been studied in
efforts to understand protein folding and stability.
61 63
Catalytic side chain groups. Six N-acetylgluco-
samine (GlcNAc) or N-acetylmuramic acid (MurNAc)
rings of the polysaccharide substrate are able to fit
precisely into six subsites (designated A to F) in a
groove between the two structural domains of egg
white lysozyme (Fig. 12-4). The bond between the
fourth and fifth rings (subsites D and E) is then cleaved.
At the active site, the side chain carboxyl group of Glu
35 is positioned correctly to serve as the proton donor
BH of Eq. 12-11, while the carboxyl of Asp 52 lies on
the opposite side of the groove. Both Glu 35 and Asp
52 have relatively high pK
a
values for carboxyl (micro-
scopic pK
a
s are ~ 5.3 and 4.6, respectively, in the fully
protonated active site when the ionic strength is ~ 0.2)
64
as a result of the hydrophobic environment and hydro-
gen bonding to other groups. If the pH is raised from
an initially low value, a proton from Asp 52 usually
dissociates first and the electrostatic field of the resulting
anion keeps Glu 35 largely protonated until the pH
Figure 12-3 Stereoscopic MolScript view of hen egg white lysozyme com-
plexed with a trisaccharide of N-acetylglucosamine (GlcNAc)
3
in binding
subsites A, B, and C. The side chain of Trp 62 and the trisaccharide above it
are shown in ball-and-stick form. From Maenaka et al.
65
Courtesy of Izumi
Kumagai.
approaches 6. Positively charged basic groups nearby
affect the pK
a
values; hence, the behavior of the en-
zyme is sensitive to the ionic strength of the medium.
63
The Asp 52 anion lies only ~ 0.3 nm from the center of
positive charge expected for an oxocarbenium ion
intermediate and presumably stabilizes it. Replace-
ment of Glu 35 by Gln destroys all catalytic activity
and replacement of Asp 52 by Ala or Asn decreases
activity to ~ 4 5% of the original.
66 68
Less than 1%
activity remained for the D52S mutant.
69
Nevertheless, Asp 52 is not absolutely essential for
lysozyme activity. Goose lysozyme lacks this catalytic
aspartate.
70
Matsumura and Kirsch suggested that
carboxyl groups of glycine residues covalently attached
to N-acetylmuramic acid rings in the natural substrates
may participate in catalysis.
66
Many mutant forms of
lysozyme have been studied. Of special interest is the
D52E mutant. In this enzyme the carboxylate of the
longer glutamate side chain reacts with the oxocarbe-
nium ion intermediate to form a covalent adduct and
apparently alter the basic mechanism.
71
Replacement
of asparagine 46, which can hydrogen bond to Asp
52, also decreases k
cat
greatly, suggesting a role in
catalysis.
72
The lysozyme-catalyzed reaction is completed by
stereospecific addition of a hydroxyl ion to the oxocar-
benium ion with the original configuration being
retained in the product. Such stereospecificity for reac-
tions of enzyme-bound carbocations is not surprising
because the enzyme probably assists in generation, on
the appropriate side of the sugar ring, of the attacking
hydroxyl ion.
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
600 Chapter 12. Transferring Groups by Displacement Reactions
Kinetic isotope effect for lysozyme. A secondary
kinetic isotope effect is expected because a molecule
with
1
H in the number 1 position can be converted to
the corresponding oxocarbenium ion somewhat more
easily than the molecule with
2
H in the same position
(Eq. 12-13). For example, in the nonenzymatic acid-
catalyzed hydrolysis of a methyl- glucoside, a reaction
also believed to proceed through a carbocation inter-
mediate,
41,75
the ratio k
1
H
/ k
2
H
is 1.14 for the anomer
and 1.09 for the anomer.
53
In the base-catalyzed
hydrolysis of the same compound, which is believed
to occur by a double-displacement reaction involving
participation of the neighboring OH group on C-2, the
ratio k
1
H
/ k
2
H
is 1.03. The corresponding ratio measured
Figure 12-4 Schematic drawing of the active site of egg white lysozyme with substrate in place and about to be cleaved.
Three strands of the small -sheet domain, which contains an extensive hydrogen bond network, are seen at the top. Notice
that this view into the active site is different from that in Fig. 12-3. A 180 rotation of either figure will make the views more
similar. The three-strand sheet can be seen (in stereo) at the lower right in Fig. 12-3 and the helix end carrying Glu 35 in the
right center foreground. A segment of a chitin oligosaccharide (green), whose reducing end is to the left, is bound into subsites B
F. Cleavage occurs between rings D and E as indicated by the heavy arrow which points to the anomeric carbon atom of ring
D. The side chain of Glu 35 is shown protonating the bridge oxygen. Ring D has been distorted into a twist conformation to
facilitate cleavage. A larger domain, which contains one long helix, is at the bottom of the drawing. The active site lies in
a cleft between the domains. Notice the chain of hydrogen bonds between the carbonyl of residue 107 in the large domain
and the peptide NH of residue 59 in the small domain. Also notice the aromatic side chains which are usually present in
carbohydrase active sites. Drawing is based in part on those of Irving Geis
73
and Levitt
74
and on a sketch by author from a
three-dimensional model.
HN
O
H
3
C
O
H
N
H
H
N
H
+
O
H
O
O
O O
O
O
O
H
N
H
3
C
H
H
O
H
107
108
N
H
N
H
O
H
N
H
3
C
O
O
O
O
N
H
O
O
N
H
H
O
O C
O
H
3
C
H
3
C
62
63
N
H
N
H
O
H
O
O
O
C
N
H
H
H
H
N
H
C O
O
O
N
H
N
O
52
59
N

N
H
O
H
O
H
H
35
O
H
N
N
H
O
O
O
H
F
Helix end
N
H
N
O
34
O
O
H
O
O
N
H
O
O
H N
N
H
H
H
N
H
N
H
O
H
O
O
H
N
N
N
O
H
H
N
H
O
O
A2
B
C
D
E
H
N
N
61
O
O
H
O
O
O
H
H
N
N
N
H
H
O
44
O
O
46
54
57
O
50
Three strands
O
OH
HO
HO
OH
O
HO
HO
OH
OCH
3
1
H or
2
H
OH
1
H or
2
H
+

CH
3
O
(12-13)
601
for the action of lysozyme is 1.11, much closer to that
of the oxocarbenium ion mechanism than to that of the
double-displacement mechanism. Similar observations
have been made with amylases.
76
Kinetic isotope effects
have also been measured for
12
C vs
13
C in the anomeric
position,
16
O vs
18
O in the leaving group (OCH
3
) of
the methyl glucosides, and for other locations
53
as well
as for hydrolysis of glucosyl fluorides.
77 79
The results
are generally supportive of the oxocarbenium ion
mechanism for lysozyme. However, as mentioned in
Section B,3, the interpretation of secondary isotope
effects is difficult. Such effects cannot reliably identify
a carbohydrase mechanism.
80,81
For acid-catalyzed hydrolysis of methyl gluco-
sides
53
the kinetic isotope effect observed for the oxy-
gen of the leaving group was k
16
O
/k
18
O
= 1.024 1.026.
Observation of similar effects for enzymes supports
the participation of an acidic group of the protein (Glu
35 of lysozyme) in catalysis but does not eliminate the
possibility of concerted involvement of a nucleophilic
group, e.g., Asp 52 in lysozyme.
81,82
Does lysozyme distort its substrate? An early
study of models indicated that for six sugar rings of a
substrate to bind tightly into the active site of lysozyme,
the ring in subsite D, which contains the carbon atom
on which the displacement occurs, had to be distorted
from its normal chair conformation into the half-chair
conformation.
83
This is illustrated in Fig. 12-4. It was
suggested that by binding the substrate chain at six
different sites the enzyme provides leverage to distort
the ring in subsite D into a conformation similar to
that of the transition state. This idea was criticized on
the basis that an enzyme would be too flexible to act
in this manner.
74
Furthermore, the non-hydrolyzed
trisaccharide MurNAc GlcNac MurNAc was shown
to fit into subsites B, C, and D of the active site groove
without distortion.
84
However, it is bound weakly.
Can electrical forces acting within the active site
help to distort the substrate and assist in formation
of the carbocation intermediate? Levitt and Warshel
suggested this possibility and proposed that the neces-
sary electrostatic force arises from the arrangement of
dipoles within the peptide backbone of the protein and
in the amino acid side chains.
74
As can be seen from
Fig. 12-4, the enzyme forms many hydrogen bonds
with the substrates. Of special interest is a chain of
hydrogen bonds that passes from the backbone carbo-
nyl of Ala 107 through the 2-acetamido group of the
substrate in subsite C and into the edge of the sheet
at the backbone NH of Asn 59. This interaction pro-
vides specificity toward GlcNac-containing substrates.
Perhaps oscillation of charge within such polarizable
chains of hydrogen bonds can also help a substrate to
move toward its transition state structure.
85,86
From simulations of lysozyme action by the methods
of molecular dynamics, Post and Karplus observed
that Glu 35 tended not to hydrogen bond to the exocy-
clic bridge oxygen, even though that would be a logi-
cal step in the protonation (by BH) shown in Eq.
12-11. They suggested that Glu 35 may protonate the
ring oxygen.
87
The ring could then open to form an
exocyclic oxonium ion, which could hydrolyze and
cyclize to the final product. The initial elimination could
receive stereoelectronic assistance from the anti-
periplanar lone pair of electrons on the bridge oxygen.
This proposal has been criticized.
81,88
For example, it is
hard to explain the rapid cleavage of glycosyl fluorides
by this exocyclic oxonium mechanism.
As attractive as the arguments for a oxocarbenium
mechanism in lysozyme are, is it still possible to explain
the experimental observations by a double displacement
in which Asp 52 serves as the nucleophile to form a
glycosyl enzyme?
38,82,82a
Sucrose phosphorylase and
other glucosyl transferases, like lysozyme, apparently
have a carboxylate ion at the active site. In some
enzymes, the carboxylate ion forms a covalent glucosyl
enzyme, whereas in lysozyme it apparently only
stabilizes a carbocation. Is there really a difference
in mechanism? Do glucosyl enzymes form only with
certain substrates or upon denaturation of the enzymes?
Nature hides her secrets well. The difficulty in pinning
down the fine mechanistic details of enzymatic action
makes it essential to be skeptical, to examine data
critically, and to try to imagine all possible alternatives
even when things seem to be proven beautifully.
Help from a neighboring group. In the 1960s it
was suggested that the acetamido group of N-acetyl-
glucosamine residues might participate as a nucleo-
phile, either stabilizing an oxocarbenium ion or
forming an oxazoline intermediate.
89 91
The proposal
received little support, as applied to lysozyme, until
sequences and structures of many larger carbohydras-
es were determined. Among these are chitinases,
enzymes that act on the same substrates as the small
lysozymes. One group of plant chitinases have a
structure similar to that of egg white lysozyme. The
243-residue enzyme from barley seeds apparently has
Glu 67 as a proton donor and Glu 89 as a possible
stabilizing nucleophile.
92
Another group of chitinases
from both plants and bacteria have active sites at ends
of ()
6
barrels.
93 95
They all have a proton-donating
glutamate in a conserved position but no aspartate that
could serve as a nucleophile. Study of complexes with
substrates and inhibitors with these enzymes has
provided direct evidence of ring distortion and of the
probable role of the acetamido group as indicated in
Eq. 12-14.
94,95
The distortion is beyond that in a sofa
conformation and allows for maximum stereoelectron-
ic assistance as well as participation of the acetamido
group.
In these relatively large enzymes the substrate
is deeply buried and cannot reach a conformation
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
602 Chapter 12. Transferring Groups by Displacement Reactions
approaching that of the transition state without having
a strained ring conformation. These enzymes may use
binding forces exerted on many parts of the substrate
to stabilize the transition state structure.
6. Cellulases and Other -Glycosidases
Cellulose, the most abundant of all biopolymers, is
extremely stable but is attacked by a host of bacterial
and fungal -glycanases.
96
Animals do not ordinarily
produce cellulases but some termites do.
97
Cellulase
structures are varied, being represented by 10 of 57
different glycosylhydrolase families.
98
Most, like lyso-
zyme, retain the configuration in their products but
some invert.
98 100
Among the cellulases are endoglycanases, which
cleave chains at random positions, and exoglycanases
(also called cellobiohydrolases) that cleave cellobiose
units from ends of chains. Some act on the nonreduc-
ing ends and some on reducing ends; a mixture of
enzymes is most effective.
99,101,102
In some bacteria a
whole series of different cellulases, together with a
large (197-kDa) organizing protein, form a cellusome,
a complex with high catalytic activity for crystalline
cellulose.
103 105
Cellulases usually have tightly packed
catalytic domains which may vary in size from ~ 200
residues
98
to over 400.
106
In most cases the catalytic
domain is connected by a flexible linker to one or more
small, globular cellulose-binding domains. These
vary in size from 36 to 200 residues and often have a
-barrel fold.
104,107108a
Their function is to hold the
enzymes to the cellulose surfaces. They may also
facilitate disruption of the tightly hydrogen bonded
cellulose structure (Fig. 4-5). As with other carbohy-
drases, carboxyl groups of amino acid side chains
provide the major catalytic groups (Table 12-1). An
extensive hydrogen-bond network which often includes
imidazole groups may influence activity.
109,110
A striking feature of a 411-residue endoglucanase
from Fusarium was revealed by the binding of a non-
hydrolyzable thiooligosaccharide substrate analog.
The pyranose ring at the cleavage site was distorted in
an identical manner to that mentioned in the preceding
section for a chitinase thought to use the substrates
acetamido group as a nucleophile (Eq. 12-14).
106
The
distortion observed is beyond that required for a sofa
conformation and allows for the maximum stereoelec-
tronic assistance (Eq. 12-12b). An oxygen atom of the
E197 carboxylate, the catalytic nucleophile, occupies a
position in the complex that is coincident with that of
the C2 acetamido oxygen in the catalytic site of the
chitinase discussed in the previous section.
The inverting -glucanases also have two catalytic
acid base groups but they are ~0 .9 1.0 nm apart rather
than ~ 0.6 nm for retaining enzymes. This allows space
for a water molecule whose

OH is the nucleophilic
reactant (

OY in Eq. 12-5) and in which a carboxylate


group assists in dissociating the water molecule.
98
(This mechanism is illustrated for glucoamylase in
Fig. 12-7).
Structural features met in some cellulases include
an , barrel
111
similar to that of glucoamylase (Fig. 2-29)
and, in a cellobiohydrolase,
101
a 5-nm-long tunnel into
which the cellulose chains must enter. Ten well-defined
subsites for glycosyl units are present in the tunnel.
101
A feature associated with this tunnel is processive
action, movement of the enzyme along the chain with-
out dissociation,
105
a phenomenon observed long ago
for amylases (see Section 9) and often observed for
enzymes acting on nucleic acids.
Another group of -glucanases, found in plants
and their seeds, hydrolyze -1,3-linked glucans
112
and,
in some cases, also mixed 1,3- and 1,4-linked polysac-
charides. A characteristic enzyme is found in barley
(Table 12-1).
113,114
Similar enzymes are produced by
some bacteria
113
and also by molluscs.
115
Xylanases act on the -1,4-linked xylan, the most
abundant of the hemicelluloses that constitutes over
30% of the dry weight of terrestrial plants.
116
They
resemble cellulases and cooperate with cellulases and
xylosidases
117
in digestion of plant cell walls.
110,116,118 121
Galactanase digests the -1,4-linked component of
pectins.
122
The final step in degradation of cellulose is hydro-
lysis of cellobiose to glucose. This is accomplished
by -glucosidases, enzymes that also hydrolyze
lactose, phosphorylated disaccharides, and cyano-
genic glycosides.
123 126
Lactose is also cleaved by
-galactosidases.
127
The large 1023-residue -galactosi-
dase from E. coli
128
is famous in the history of molecular
biology as a component of the lac operon.
129 132
Its
properties are also employed to assist in the cloning of
genes (Chapter 26).
O
N
HO
HO
OH
O
HO
HO
OH
O
R
HB
H
3
C
O
N
O
CH
3
+
H
O
R
H
H
+
Oxazoline
B
(12-14)
603 B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
TABLE 12-1
Acidic and Basic Catalytic Groups in a Few Glycosyltransferases
a
Lysozyme
Human, hen
b
130 No E35 D53(52)
Bacteriophage T4
c
No E11 D20
Chitinase
Rubber plant
d
273 No E127
Cellulases
E1 endocellulase,
Acidothermus
e
521
catalytic domain 358 No E282 E162, D252*
Endoglucanase I
Fusarium
f
411 No E197 E202
Endoglucanase Cen A
Cellulomonas
g
>351 Yes D392 D78
1,3--D-Glucanase
Barley
h
No E231 E288
Xylanase
B. circulans
i
No Yes E78 E172
-Glucosidase
Agrobacterium
j
458 No Yes E358 E170, Y298*
-Galactosidase
E. coli
k
1023 No Yes E537 E461, Y503*
-Amylase
Human and pig
l
496 No D197 E233, D300*
Barley
m
No D179 E204, D289*
Aspergillus
n
No D206 E230
Cyclodextrin
glucosyltransferase
B. circulans
o
D229 E257, D328
-Glucosidase
Saccharomyces
p
No Yes D214 E233, D300
Glycogen
debranching
enzyme, rabbit
s
No Yes D549
Glucoamylase
Aspergillus
q
616 Yes H
2
O, E400 E179
-Amylase
Soybean
r
Yes E186 E380
Glucocerebrosidase
Human
t
E340
Enzyme Number of
residues
Inverting? Glycosyl-
enzyme
identified?
Nucleophile
(COO

)
Electrophile
(proton donor)
* Assisting group
a
For classification of glycosyl hydrolases into families, see Henrissat, B.,
Callebaut, I., Fabrega, S., Lehn, P., Mornon, J.-P., and Davies, G. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 70907094
b
Matsumura, I., and Kirsch, J. F. (1996) Biochemistry 35, 18811889; Harata,
K., Muraki, M., Hayashi, Y., and Jigami, Y. (1992) Protein Sci. 1, 14471453
c
Brzozowski, A. M., and Davies, G. J. (1997) Biochemistry 36, 1083710845;
Hardy, L. W., and Poteete, A. R. (1991) Biochemistry 30, 94579463 Kuroki,
R., Weaver, L. H., and Matthews, B. W. (1993) Science 262, 20302033
d
Tews, I., Terwisscha van Scheltinga, A. C., Perrakis, A., Wilson, K. S., and
Dijkstra, B. W. (1997) J. Am. Chem. Soc. 119, 79547959
e
Sakon, J., Adney, W. S., Himmel, M. E., Thomas, S. R., and Karplus, P. A. (1996)
Biochemistry 35, 1064810660; Ghidoni, R., Sonnino, S., Tettamanti, G.,
Baumann, N., Reuter, G., and Schauer, R. (1980) J. Biol. Chem. 255, 69906995
f
Sulzenbacher, G., Schlein, M., and Davies, G. J. (1997) Biochemistry 36,
59025911; Sulzenbacher, G., Driguez, H., Henrissat, B., Schlein, M., and
Davies, G. J. (1996) Biochemistry 35, 1528015287; Mackenzie, L. F., Davies,
G. J., Schlein, M., and Withers, S. G. (1997) Biochemistry 36, 58935901
g
Damude, H. G., Withers, S. G., Kilburn, D. G., Miller, R. C., Jr., and Warren,
R. A. J. (1995) Biochemistry 34, 22202224
h
Chen, L., Garrett, T. P. J., Fincher, G. B., and Hoj, P. B. (1995) J. Biol. Chem.
270, 80938101
i
Wakarchuk, W. W., Campbell, R. L., Sung, W. L., Davoodi, J., and Yaguchi,
M. (1994) Protein Sci. 3, 467475; Lawson, S. L., Wakarchuk, W. W., and
Withers, S. G. (1997) Biochemistry 36, 22572265; Sidhu, G., Withers, S. G.,
Nguyen, N. T., McIntosh, L. P., Ziser, L., and Brayer, G. D. (1999) Biochemistry
38, 53465354
j
Wang, Q., Trimbur, D., Graham, R., Warren, R. A. J., and Withers, S. G.
(1995) Biochemistry 34, 1455414562
k
Gebler, J. C., Aebersold, R., and Withers, S. G. (1992) J. Biol. Chem. 267,
1112611130; Jacobson, R. H., Zhang, X.-J., DuBose, R. F., and Matthews, B.
W. (1994) Nature (London) 369, 761766; Richard, J. P., Huber, R. E., Lin, S.,
Heo, C., and Amyes, T. L. (1996) Biochemistry 35, 1237712386
l
Brayer, G. D., Luo, Y., and Withers, S. G. (1995) Protein Sci. 4, 17301742;
Qian, M., Haser, R., Buisson, G., Due, E., and Payan, F. (1994) Biochemistry
33, 62846294
m
Kadziola, A., Sogaard, M., Svensson, B., and Haser, R. (1998) J. Mol. Biol.
278, 205217
n
Brzozowski, A. M., and Davies, G. J. (1997) Biochemistry 36, 1083710845;
Matsuura, Y., Kusunoki, M., Harada, W., and Kakudo, M. (1984) J. Biochem.
95, 697702
o
Knegtel, R. M. A., Strokopytov, B., Penninga, D., Faber, O. G., Rozeboom, H.
J., Kalk, K. H., Dijkhuizen, L., and Dijkstra, B. W. (1995) J. Biol. Chem. 270,
2925629264
p
McCarter, J. D., and Withers, S. G. (1996) J. Biol. Chem. 271, 68896894
q
Christensen, U., Olsen, K., Stoffer, B. B., and Svensson, B. (1996) Biochemistry
35, 1500915018
r
Mikami, B., Degano, M., Hehre, E. J., and Sacchettini, J. C. (1994) Biochemistry
33, 77797787; Adachi, M., Mikami, B., Katsube, T., and Utsumi, S. (1998) J.
Biol. Chem. 273, 1985919865
s
Braun, C., Lindhorst, T., Madsen, N. B., and Withers, S. G. (1996) Biochemistry
35, 54585463
t
Withers, S. G., and Aebersold, R. (1995) Protein Sci. 4, 361372
604 Chapter 12. Transferring Groups by Displacement Reactions
7. Glycogen Phosphorylase
A large number of different glycosyltransferases
act on the 1,4 linkages of glycogen, starch, and related
polysaccharides. Among these, one of the most studied
is glycogen phosphorylase. It is not a hydrolase, but it
catalyzes cleavage of 1,4 linkages at the nonreducing
ends of glycogen molecules by displacement with
inorganic phosphate to give -D-glucose-1-phosphate.
It is a very large enzyme (841 residues in rabbit muscle)
whose structure is shown in Fig. 11-5. Its complex
regulatory mechanisms were discussed briefly in
Chapter 11.
Neither partial exchange reactions nor inversion
of configuration occur when glycogen phosphorylase
acts on its substrates. The enzyme apparently does
nothing until both substrates are present. These are
glycogen + inorganic phosphate or, for the reverse
reaction, glycogen (shifted over by one sugar binding
subsite) + glucose-1-phosphate. The active site is in
a deep groove in the enzyme. Evidently, the protein
must close and fold around the substrates before it
becomes active.
133
An oxocarbenium ion mechanism
has also been proposed for this enzyme, partly on
the basis of strong inhibition of phosphorylase by
5-gluconolactone,
134,135
a compound having a half-
chair conformation and perhaps acting as a transition
state inhibitor (Chapter 9). This gluconolactone also
inhibits many other carbohydrases.
123,136
An unexpected discovery was that glycogen phospho-
rylase contains a molecule of the coenzyme pyridoxal
5'-phosphate (PLP) bound into the center of the protein
behind the active site
134,135,137 139
with its phosphate
group adjacent to the binding site of the phosphate of
glucose-1-phosphate. It probably serves as a general
base catalyst, e.g., assisting the attack of a phosphate
ion on the oxocarbenium formed by cleavage of the
glycogen chain (Eq. 12-15).
137,140 142
A key observation
by Graves and associates was that pyridoxal alone does
not activate apo-phosphorylase but that pyridoxal
plus phosphite, phosphate, or fluorophosphate does
provide up to 20% of full activity.
143
X-ray crystallo-
graphy confirmed that these activating anions are
bound into the active site at the approximate position
of the phosphate group of PLP.
139,143a
Glycogen phos-
phorylase is being studied by X-ray diffraction tech-
niques that allow observation of structural changes in
as short a time as a few milliseconds (see Chapter 3,
Section H).
144
The regulation of glycogen phosphorylase, like
that of many other allosteric proteins, depends upon
the existence of two distinct conformational states,
whose structures have been established by crystallo-
graphy.
137
It is not immediately evident how they can
affect the active site. In the R-state the enzyme has a
low affinity for both substrates and activators such as
AMP. In the T-state the affinities are much higher. For
example, that of inorganic phosphate is raised by a
factor of fifteen.
137
As we have already seen (Figs. 11-4,
11-5) the relatively inactive phosphorylase b is converted
to the active phosphorylase a by phosphorylation of
the side chain of serine 14, a structural change which
favors the R-state. In the inactive T-state of phospho-
rylase b, an 18-residue N-terminal segment of the poly-
peptide is not seen by X-ray diffraction, presumably
because it does not assume a fixed conformation but
projects into a solvent channel in the crystal and moves
freely within it. However, in the active R-state of phos-
phorylase a, in which Ser 14 has been phosphorylated,
the N-terminal segment is rigid. The phospho group
on Ser 14 binds to Arg 69 and other arginine, lysine,
and histidine side chains from both subunits.
137,145
The phosphorylation of Ser 14 occurs 1.5 nm from the
active sites. The conformational changes induced by
phosphorylation of Ser 14 cause a rotation of about
10 between subunits, somewhat reminiscent of the
changes accompanying oxygenation of hemoglobin
(Fig. 7-25).
O
OH
HO
HO
OH
O R
O
OH
HO
HO
OH
O
HO R
P
O O
O
H
(Glycogen, n glucosyl units)
O
+
P
O O
O
Pyridoxal
H

(Glycogen, (n1) glucosyl units)


+

(Schiff base)
Glucose-1-phosphate
(12-15)
HO
HO
O
O
CH
2
OH
OH
HO
HO
O
O
CH
2
OH
OH
+

5-Gluconolactone
605
A related structural change accompanies binding
of caffeine, adenosine, or AMP at high concentrations.
These substances inhibit the enzyme by binding at a
site next to the catalytic site (Fig. 11-5), stabilizing the
T-state and causing a loop of protein to move into the
catalytic site and to block it.
137
The catalytic site lies
deeply buried in the protein between the two large
structural domains (Fig. 11-5). The substrate is held by
a network of hydrogen bonds, some of which are
shown in Fig. 12-5, in which the active site contains an
inhibitory substrate analog nojirimycin tetrazole,
which is viewed toward the edges of the two rings.
Figure 12-5 (A) Stereoscopic view of the structure of the catalytic site of
phosphorylase b in the inhibited T-state with the inhibitor nojirimycin tetra-
zole bound into the active site. Inorganic phosphate (P
i
) as well as the coen-
zyme pyridoxal 5'-phosphate (PLP) are also shown. (B) Details of interactions
of the inhibitor, P
i
, and PLP with the protein and with water molecules (small
circles). This is a weak-binding state but the P
i
has displaced the negatively
charged side chain carboxylate of Asp 283 (visible at the lower right in A).
This carboxylate blocks access to the P
i
site in the free T-state enzyme. Here,
the positively charged Arg 569 guanidinium group has swung in to interact
with the P
i
. From Mitchell et al.
133
Courtesy of Louise N. Johnson.
The enzyme is in the weak-binding T-state but inor-
ganic phosphate (P
i
) is bound below it and next to the
phosphate group of PLP, as required by the mechanism
of Eq. 12-15.
The difference in binding affinities for the T- and
R-states lies in a flexible loop of residues 280288, which
in the T-state blocks access to the substrate-binding
cleft. The universally conserved Asp 238 behaves as
a substrate mimic, occupying the P
i
site.
137,146,147
In
the R-state this residue moves, allowing P
i
to enter
and bind (Fig. 12-5).
Most of the structure of mammalian phosphory-
lases is conserved in species as diverse as E. coli and
the potato.
148
However, E. coli maltodextrin phos-
phorylase
143a,149 151
and potato phosphorylases have
less sophisticated regulatory mechanisms than do the
animal enzymes. Another feature of glycogen phos-
phorylase is a glycogen storage site about 2.5 nm
from the active site (Fig. 11-5).
152
This provides a
means for the enzyme to hold onto the giant glycogen
molecule while nibbling off the outside ends of
nearby branches.
8. Starch-Hydrolyzing Enzymes
Among the hydrolases are the widely distributed
-amylases, endo-glycosidases which hydrolyze
N
HO
HO
OH
OH
N
N
N
N
HO
HO
OH
OH
N
N
N
Nojirimycin tetrazole
+

B. Nucleophilic Displacements on Singly Bonded Carbon Atoms


A
B
606 Chapter 12. Transferring Groups by Displacement Reactions
starch chains by random attack at points far from
chain ends to form short polysaccharide chains known
as dextrins as well as simpler sugars.
153
The catalyzed
reactions proceed with retention of the original
configuration. Alpha-amylases are found in fungi,
plants, and animals. One powerful enzyme of this
class is present in the saliva of most humans and other
isoenzymes are formed by the pancreas.
154
They are
encoded by a family of genes, a fact that accounts for
the existence of some healthy individuals completely
lacking salivary amylase.
155
Structures are known for human
154
and porcine
156 159
-amylases as well as for corresponding enzymes from
barley,
160
mealworms,
161
fungi, and bacteria.
162 164
The
-amylase from Aspergillus oryzae (Taka-amylase),
widely used in laboratory work, was the first for which
a structure was determined.
165,166
The -amylases fold
into three domains (Fig. 12-6), with the active site in
the center of an (/)
8
barrel. All of the -amylases
contain one or more bound Ca
2+
ions. Some, including
the human -amylases, also require a chloride ion.
The Cl

is held by a pair of arginine guanidinium


groups
154
and interacts with adjacent carboxyl groups,
inducing pK
a
shifts and allosteric activation.
163
The sequences of -amylases vary widely but a
conserved cluster of one glutamate and two aspartates
is usually present (Table 12-1).
Studies of the action patterns, i.e., the distribution
of products formed by -amylases when acting on a
variety of 1,4-linked oligosaccharides, suggested that
the substrate binding region of the porcine pancreatic
enzyme has five subsites, each binding one glucose
residue.
153
The -glucan chain is cleaved between
the residues bound at the second and third subsites
(numbered from the reducing end of the substrate)
by a lysozyme-like mechanism. Endolytic enzymes,
which cleave biopolymer chains internally, are usually
thought to carry out random attack. However, after
the initial catalytic reaction one of the polysaccharide
products of porcine pancreatic amylase action often
does not leave the enzyme. The polysaccharide simply
slides over until it fully occupies all of the subsites
of the substrate binding site and a second attack
occurs. An average of seven catalytic events occur
each time it forms a complex with amylase.
167
Is there
a mechanism by which the enzyme deliberately pro-
motes the sliding of the substrate in this multiple
attack or processive mechanism
168,169
or does the
dissociated product simply diffuse a short distance
while enclosed in a solvent cage? The latter explana-
tion may be adequate for some enzymes. In contrast
to pancreatic -amylase a bacterial maltogenic
-amylase produces principally maltose as a
product.
169a,b
Digestion of starch and glycogen by -amylases
produces a mixture of glucose, oligosaccharides, and
dextrins, which in the human body are further degraded
by -glucosidases of the brush border membrane of
the small intestine.
154
A lysosomal form of the enzyme
is missing in Pompes disease (Box 20-D).
170,171
The
-glucosidases are also members of the -amylase
family.
46,172
For digestion of the branched amylo-
pectin and glycogen a debranching enzyme and
-1,6-glucosidase activity are required. In mammals
these activities are found in a single polypeptide chain
with separate but adjacent active sites.
50,173,174
The
debranching enzyme catalyzes transfer of oligosaccha-
ride chains from 1,6-linked branch positions to new
locations at ends of chains with 1,4 linkages. A bacte-
rial oligo-1,6-glucosidase has a catalytic site formed from
Figure 12-6 Drawing
showing the overall
polypeptide chain fold
and relative positioning
of the three structural
domains of human
pancreatic -amylase.
Also drawn are the
locations of the calcium
and chloride binding
sites. Overlaid is the
placement of a modified
form of the inhibitor
acarbose (p. 607) that
binds in the active site
cleft. MolScript draw-
ing courtesy of G. Sidhu
and G. Brayer.
607
D199, E255, and D329 similar to that of -amylases.
175
Another member of the -amylase family has
principally a glycosyltransferase activity. Cyclodextrin
glucanotransferase forms cyclodextrins (Box 4-A)
by a transferase reaction in which a 6- to 8- member
oligo-saccharyl group is transferred from a straight
amylase chain onto a protein side chain (Glu 257) and
then joins the ends of the oligosaccharide to form the
cyclo-dextrin rings (an overall double-displacement
process).
176 177b
However, large circular dextrins are
produced initially and are then converted into the
smaller cyclodextrins.
178
A starch-digesting enzyme of great industrial
importance is glucoamylase, whose (,)
6
-barrel
structure is shown in Fig. 2-29. That figure also shows
the tetrasaccharide inhibitor acarbose in the active
site. The ring at the nonreducing end is deeply em-
bedded in the protein and, as shown in Fig. 12-7 for
the related D-gluco-dihydroacarbose, is held by many
hydrogen bonds. This slow enzyme (k
cat
~ 50 s
1
at
45C)
179
cuts off a single glucose unit, then releases
itself from the starch before releasing the glucose and
rebinding to the starch. The catalytic acid has been
identified as the carboxyl group of Glu 179. In Fig.
12-7 it is seen, presumably as a carboxylate group,
tightly hydrogen bonded to what is doubtless the
bridge NH
2
+
between rings A and B of the inhibitor
D-gluco-dihydroacarbose. Thus, the complex mimics
that of a true substrate protonated on the bridge oxy-
gen, a possible first step in normal catalysis. In accord
with this mechanism (Eq. 12-16), glucoamylase is an
OH
HO
HO
OH
N
O
OH
CH
3
HO
O
O
OH
HO
OH
O
O
OH
HO
OH
OH
HO
HO
OH
N
O
OH
CH
3
HO
O
O
OH
HO
OH
O
O
OH
HO
OH
H
1
H
H, OH
H, OH
d-Gluco-dihydroacarbose
Acarbose
A
B
C
D
1
2
3
4
5
6
7
2
3
4
5
6
7
inverting enzyme.
Notice in Fig. 12-7 the abundance of polar groups,
many of which are charged. Four carboxyl groups,
from D55, E179, E180, and E400, are present in the active
site region and participate in the hydrogen bonded
network. Active sites of other carbohydrases differ
from that of glucoamylase. However, the presence of
many hydrogen-bonded polar groups, including two or
more carboxyl groups, is characteristic. As illustrated
in Figs. 9-8 and 9-9, the pH dependence of the maximum
velocity v
max
is often determined by these carboxyl
groups. At least one of them must be protonated for
maximum catalytic activity and if two are protonated
activity may fall again. Under these circumstances a
single proton bound to a molecule of enzyme may
spend a fraction of its time on each of several different
carboxylate, imidazole, or other groups.
181
In the case
of glucoamylase it was concluded from the X-ray struc-
ture that the catalytic acid group, E179, is probably
unprotonated most of the time.
181
However, it can still
bind to starch. Then, after a fraction of a second a
proton may be transferred onto the
Glu 179 carboxylate and from there
to the bridge oxygen of the substrate,
inducing reaction according to Eq.
12-16. It may even be necessary to
have enzyme protonated initially
on a group other than E179 to allow
small conformational changes to
occur prior to formation of the final
activated complex. Such essential
conformational changes have often
been invoked for glucoamylase.
182
Just as most cellulases have
special cellulose-binding domains,
glucoamylase has a compact C-
terminal starch-binding domain
(residues 509 616) similar to the
O
O
HO
HO
O
O
H
O
H
H
H
H
O
O
E179
E400
O O
+

(12-16)
B. Nucleophilic Displacements on Singly Bonded Carbon Atoms
608 Chapter 12. Transferring Groups by Displacement Reactions
L177
W178
R54
E180
#Man-S455
PH-4
#A205
R305
PH-6
W120
E179
L319
S119
W417
D55
W317
W52
Wat500
Y311
#T148
E400
L415
Q401
Y48
L177
R54
#Man-S455
W
E180
W120
PH-4
PH-6
E179
#A
R
S119
D55
W417
L319
W52
Wat500
W
Y3
#T148
L415
Y48
E400
Q40
corresponding domain of cellulases.
183
It is connected
by a glycosylated linker to the 470-residue catalytic
subunit. Cyclodextrin glucanotransferase also has a
starch-binding domain.
184
Beta-amylases, characteristic plant and bacterial
enzymes
184a
, have an exo action, cutting off chain ends
two sugar units at a time as maltose. The original
linkage is inverted, the product being -maltose. The
(/)
8
-barrel structure is unlike that of glucoamylase,
but the spacing of active site carboxyl groups suggests
that a water molecule is held and activated as in glu-
coamylase.
185,186
The multiple subsite structure of the
active site may permit the substrate amylase to slip
forward after a maltose product molecule leaves the
site. This may account for the observed processive
action.
186
Many specialized glycosyl transferases synthesize
glycogen, starch, cellulose, and other polysaccharides
and add glycosyl groups to glycoproteins and glyco-
lipids.
187,188
Often the glycosyl group is transferred
from a carrier such as uridine diphosphate (UDP). An
example is glycogen synthase (Fig. 11-4), which trans-
fers glucosyl groups of UDP-glucose to the 4' OH
groups at the nonreducing ends of the bushlike glyco-
gen molecules. Other similar synthetic reactions are
considered in Chapter 20.
Figure 12-7 Stereoscopic
view of the inhibitor D-gluco-
dihydroacarbose in its com-
plex with glucoamylase of
Aspergillus. Residues from
symmetry-related molecules
of the enzyme are shown as
dotted lines. Ring A at the
nonreducing end of an amy-
lase chain is thought to bind
in a similar way (with ring A
at the bottom in this figure).
Cleavage is between the A
and B rings E178 acting as a
proton donor to the bridge
oxygen (or NH for the inhibi-
tor). The attacking nucleo-
phile is thought to be a water
molecule, which is labeled
Wat 500 and is held by the
assisting carboxylate of
E400.
180
Courtesy of
Alexander Aleshin.
C. Displacement Reactions on Carbonyl
Groups
A second major class of nucleophilic displacement
reactions (Type 1B in Table 10-1) involve replacement
of a group Y attached to a carbonyl carbon:
Group Y may be OR (esters), SR (thioesters) or NHR
(amides or peptides). If B

is a hydroxyl ion formed


from H
2
O, the reaction is hydrolysis. If B

is the
anion of an alcohol, thiol, or amine, the reaction is
transacylation. Transacylation is an essential process
in biosynthesis of proteins and lipids, but it is the
digestive enzymes, which catalyze hydrolysis, that
have been studied most intensively.
Uncatalyzed hydrolysis of a peptide linkage is
very slow with t
1/2
at neutral pH and 25C of ~ 300600
years.
189
Both acids and bases catalyze hydrolysis, but
enzymes are needed for rapid digestion. The carbonyl
group C = O is highly polarized, with the resonance
form C
+
O

contributing substantially to its structure.


An attack by a base will take place readily on the
electrophilic carbon atom. While the reactivity of the
carbonyl group in esters and amides is relatively low
R C
O
Y + + R C
O
B B:
+
H + YH
(12-17)
609
because of the resonance stabilization of these groups,
the carbon atom still maintains an electrophilic charac-
ter and may combine with basic groups. Thus, in the
base-catalyzed hydrolysis of esters a hydroxyl ion
adds to the carbonyl group to form a transient single-
bonded tetrahedral intermediate (Eq. 12-18). Similar
intermediates are believed to form during the action
of many enzymes. However, for purposes of classifi-
cation, we can regard them as simple displacement
reactions on a carbon atom with the understanding
that there are probably transient single-bonded inter-
mediates.
A large number of hydrolytic enzymes, the
proteases, and peptidases act on peptide linkages of
proteins.
190
At present the traditional name protease,
which implies proteolysis, is most often used. How-
ever, the IUB encourages use of the name proteinase.
Although this seems less specific in meaning, its use
will probably increase. Some proteases trim newly
formed peptide chains, others convert proteins from
precursor forms into biologically active molecules, and
others digest proteins. In addition to endopeptidases,
such as trypsin and chymotrypsin, which cleave at
positions within a long peptide chain, there are many
enzymes that cleave amino acids from the ends of
chains. These are usually called peptidases and are
designated aminopeptidases if they cleave from the
N terminus and carboxypeptidases if they cleave
from the C terminus. Most of these enzymes can be
classified into serine proteases, cysteine proteases,
aspartic proteases, metallo proteases, or N-terminal
nucleophile hydrolases, depending upon the chemical
nature of the active site. These groups are further
divided into families or clans.
190
1. The Serine Proteases
The digestive enzymes trypsin, chymotrypsin,
elastase, and proteinase E are related serine proteases.
All three are synthesized in the pancreas which secretes
5 10 g per day of proteins, mostly the inactive pro-
enzymes (zymogens) of digestive enzymes.
191,192
(12-18)
These proenzymes are synthesized and packaged
as zymogen granules which travel to the surfaces of
the secretory cells. The contents of the granules are
secreted into the extracellular medium and are dis-
charged via the pancreatic duct into the small intestine.
At their sites of action, the zymogens are converted
into active enzymes by the cutting out of one or more
pieces from the precursor. This occurs in a cascade-type
process triggered by enteropeptidase (historically
enterokinase), another serine protease which is secreted
by the intestinal lining.
192a
Human enteropeptidase
consists of a 235-residue catalytic subunit bonded
through a disulfide bridge to a larger 784-residue
membrane-anchoring subunit.
193 195
It attacks specifi-
cally trypsinogen, converting it to active trypsin.
196,197
Trypsin in turn activates the other zymogens, as is
indicated in Fig. 12-8. Trypsin can also activate its
own zymogen, trypsinogen, in an autocatalytic process.
Chymotrypsinogen consists of a single 245-residue
chain. The amino acid residues in chymotrypsin, tryp-
sin, and elastase are usually all numbered according to
their position in this zymogen. Inactive proenzymes
are formed as precursors to enzymes of many different
classes and are activated in a variety of ways. A part
of the polypeptide chain of the proenzymes is often
folded over the active site, interacting in a nonsub-
strate-like fashion and blocking the site.
197a
Serine as a nucleophile. An early clue to the
mechanism of action of chymotrypsin came from
investigation of the related acetylcholinesterase.
This key enzyme of the nervous system is inactivated
irreversibly by powerful phosphorus-containing poisons
that had been developed as insecticides and as war
gases (nerve gases, Box 12-C). Around 1949, the nerve
gas diisopropylfluorophosphate (DFP) was shown
also to inactivate chymotrypsin. When radioactive
32
P-containing DFP was allowed to react the
32
P became
C. Displacement Reactions on Carbonyl Groups
Trypsinogen
Chymotrypsinogen
Proelastase
Procarboxypeptidases
A and B
Carboxypeptidases
A and B
Elastase
Chymotrypsin
Trypsin
Enteropeptidase (present in intestinal brush
border membrane)
Figure 12-8 Cascade of reactions that activate pancreatic
proteases. Enteropeptidase, or trypsin, cleaves the proen-
zyme (zymogen) at specific sites.
R C
O
OR' R C
O
OR'
OH
R C
O
OH
HO

Tetrahedral intermediate
H
+
HOR
610 Chapter 12. Transferring Groups by Displacement Reactions
covalently attached to the enzyme. When the labeled
enzyme was denatured and subjected to acid hydrolysis
the phosphorus stuck tightly; the radioactive fragment
was identified as O-phosphoserine. It was evident
that this product could be formed by an attack of the
hydroxyl group of the serine side chain on the phos-
phorus with displacment of the fluoride ion. This is a
nucleophilic displacement on phosphorus, occurring
on an enzyme that normally catalyzes displacement
on C = O. The DFP molecule acts as a pseudosubstrate
which reacts with the enzyme in a manner analogous
to that of a true substrate but which does not complete
the reaction sequence normally.
From study of peptides formed by partial hydroly-
sis of the
32
P-labeled chymotrypsin, the sequence of
amino acids surrounding the reactive serine was estab-
lished and serine 195 was identified as the residue whose
side chain hydroxyl group became phosphorylated.
The same sequence Gly-Asp-Ser-Gly was soon discov-
ered around reactive serine residues in trypsin, throm-
bin, elastase, and in the trypsin-like cocoonase used
by silkmoths to escape from their cocoons.
198
We
know now that these are only a few of the enzymes in
a very large family of serine proteases, most of which
have related active site sequences.
199,200
Among these
are thrombin and other enzymes of the blood-clotting
cascade (Fig. 12-17), proteases of lysosomes, and se-
creted proteases.
Numerous serine proteases, including trypsin-like
enzymes called tryptases
201 204a
and chymotrypsin-
like chymases,
205 207
are found within tissues in which
they are stored in granules of mast cells,
208
neutro-
phils, lymphocytes, and cytotoxic T cells.
205
Secretory
granules of mast cells present in skin and other tissues
contain high concentrations of tryptase and chymase
precursors
202,206
which may be released as part of an
inflammatory response. Tryptase may be involved in
asthma and other allergic resposes.
201
Cathepsin G
209
(proteinase II), neutrophil elastase, and proteinase
III
210
are found in granules of neutrophils and mono-
cytes as well as in mast cells.
209
Cytoplasmic granules
of cytotoxic T cells contain at least seven proteases
called granzymes that can be released to attack target
cells.
211 213a
Many secreted proteins, as well as smaller peptide
hormones, are acted upon in the endoplasmic reticu-
lum by tryptases and other serine proteases. They
often cut between pairs of basic residues such as KK,
KR, or RR.
214 216
A substilisin-like protease cleaves
adjacent to methionine.
217
Other classes of proteases
(e.g., zinc-dependent carboxypeptidases) also partici-
pate in this processing. Serine carboxypeptidases
are involved in processing human prohormones.
218
Among the serine carboxypeptidases of known struc-
ture is one from wheat
219
and carboxypeptidase Y, a
vacuolar enzyme from yeast.
220
Like the pancreatic
metallocarboxypeptidases discussed in Section 4, these
enzymes remove one amino acid at a time, a property
that has made carboxypeptidases valuable reagents for
determination of amino acid sequences. Carboxypep-
tidases may also be used for modification of proteins
by removal of one or a few amino acids from the ends.
The variety of bacterial serine proteases known
include the 198-residue -lytic protease of Myxo-
bacter,
221
a family of at least 80 subtilisins which are
produced by various species of Bacillus
222 225a
as well
as by many other organisms,
226
and a trypsinlike
enzyme from Streptomyces griseus.
227,228
Tripeptidyl
peptidases, subtilisin-like enzymes, cut tripeptides
from the N-termini of proteins.
228a,b,c
One participates
in lysosomal protein degradation and the other, an
oligomer of 138 kDa subunits, cuts precursor proteins
to form neuropeptides and other hormones (Chapter
30).
Acyl-enzyme intermediates. Serine proteases
are probably the most studied of any group of en-
zymes.
229
Early work was focused on the digestive
enzymes. The pseudosubstrate, p-nitrophenyl acetate,
reacts with chymotrypsin at pH 4 (far below the opti-
mum pH for hydrolysis) with rapid release of p-nitro-
phenol and formation of acetyl derivative of the
enzyme.
This acetyl enzyme hydrolyzes very slowly at pH 4
but rapidly at higher pH. These experiments suggest-
ed a double displacement mechanism:
CH
3
C O
O
NO
2
p-Nitrophenyl acetate
C O P F
O
O
CH
H
3
C CH
3
H
3
C
H
3
C
P
O
CH
2
C
COO
H
3
N H
O
O
H
32
Enzyme
32
P-containing
enzyme
32
+
O-Phosphoserine
HO
Diisopropyfluorophosphate (DFP), also
called diisopropylphosphofluoridate

(12-19)
611
Although the experiments with DFP suggested
that the O

group from Ser 195 might be the base B

in this equation, there was reluctance to accept this


deduction because of the very weak acidity of the
CH
2
OH group. Furthermore, the pH dependence of
catalysis suggested an imidazole group of a histidine
side chain as the attacking nucleophile B. Indeed,
imidazoles catalyze the nonenzymatic hydrolysis
168
97
102
228
214
182
220
188
136
245

65
120
36
+
16
191 201
195

150
76
O
O
42
57
87
S
S
N
H
Figure 12-9 Alpha-carbon diagram
of the three-dimensional structure of
pancreatic elastase. A principal struc-
tural feature is a pair of cylinders.
One of these, at the top of the figure, is
viewed end-on, while the other, at the
bottom of the figure, is viewed from a
side. The prominent interface between
them is seen in the center. The -carbon
positions of the catalytic triad serine
195, histidine 57, and aspartate 102 are
marked. As shown in Fig. 12-10 the
catalytic triad is located across the
interface between cylinders, which
may allow for easier conformational
alterations during the action of the
enzyme. The four disulfide bridges
and the long C-terminal helix are also
emphasized. The cross-hatched loop
regions are residues present in elastase
but not present in chymotrypsin. The
green shaded strand (residues 214 216)
is a segment that joins residues P
1
and P
2
of the substrate to the edge of the lower
cylinder in an extended structure.
The ion pair formed by aspartate 194
and isoleucine 16 during zymogen
activation is also shown, as are two
peptide carbonyl groups that protrude
into the interface area, one forming a
hydrogen bond across the interface.
Modified from a drawing of Sawyer
et al.
234
which was in turn based on
the original chymotrypsin drawing of
Annette Snazle.
230
C. Displacement Reactions on Carbonyl Groups
of p-nitrophenyl acetate with formation of unstable
N-acetyl imidazoles as intermediates. Thus, while the
stable end products of reactions with pseudosubstrates
were unquestionably derivatives of serine, the possi-
bility remained that these were side products and that
histidine was involved in transient, rapidly forming,
and reacting intermediates. It remained for the results
of the then newly developed science of X-ray crystal-
lography to clarify this question.
230
Three-dimensional structures. The structures of
chymotrypsin,
199,230,231
trypsin,
232,233
elastase,
234,235
thrombin,
236
kallikrein,
237,238
and many other enzymes
are similar, with the basic fold shown in Fig. 12-9.
199,228
Both Ser 195 and His 57 (or corresponding residues)
are present in the active site (Figs. 12-9, 12-10). From
the observed positions of competitive inhibitors occu-
pying the active site, the modes of binding depicted in
Fig. 12-10A for the chymotrypsin family and in Fig.
12-10B for the subtilisin family have been deduced.
Bear in mind that the X-ray diffraction results do not
R C
O
O R
H
B
Enzyme
R C
O
B
Enzyme
R C
O
OH
+

B
Enzyme
Acyl
enzyme

HOR
OH

(12-20)
612 Chapter 12. Transferring Groups by Displacement Reactions
O
N
H
O
C
Asp
102
57
43
Cylinder 1
S
197
H
S
N H
O
N
N
O
C
H
O
40
H
N
O
N
H
H
N
H O
H
O
O
O
H H
Ser 32
Gln 30
Gly 193
H
O
O
H
N
143
O
O
192
Ile 16
N
H
O
O
N
H
195
O
H
N
H
N
H
O
O
H
2
O
O
O
H N
N
N
H
N
O
Pro
H
O
Interface
Substrate
Cylinder 2

NH
3
+
N
N
C
P
3
P
2
P
1
N
H
H
O
H
213
N
H
O
O
O
O
N
H
N
O
O
H
H
N
N
H
H
N
O
H
N
N
H
N
H
214
227
216
Figure 12-10 (A) Part of
the hydrogen-bonding net-
work of trypsin and other
serine proteases with a bound
trypsin substrate (green).
Residues P
1
to P
3
, as defined in
Fig. 12-14, have been marked.
The view is similar to that in
Fig. 12-9 but some background
lines have been omitted. Note
the two competing hydrogen-
bonded chains passing through
C = O of residue 227 in
cylinder 2. One of these
chains passes through the
backbone of the substrate and
the other through residues
214 and 195 across the inter-
face between domains into
cylinder 2. Asp 102, His 57,
and Ser 195 of the catalytic
triad are emphasized. Arrows
indicate probable movement of
electrons from the negative
charge of Asp 102 into the
oxyanion hole. After
Metzler.
85
Based on papers
of Sawyer et al.
234
and Huber
and Bode.
240
(B) Stereoscopic
view showing the model sub-
strate N-succinyl-L-Ala-L-Ala-
L-Pro-L-Phe-p-nitroanilide
bound into the active site of
subtilisin BPN. Residues of
the catalytic triad (Ser 221,
His 64, Asp 32) and some
others are labeled. Subsites
P
1
', P
1
, P
2
, P
3
, and P
4
(see Fig.
12-14) are also labeled. Notice
that site P
3
, which is near the
top in this drawing, is at the
bottom in (A). Based on X-ray
data of R. Bott and M. Ultsch
at 0.2 nm resolution. From
Wells and Estell.
223
show where the hydrogen atoms are and that these
have been added in Fig. 12-10. The imidazole group
of His 57 is located next to the side chain OH group
of Ser 195 and is able to form an N-- -HO hydrogen
bond to it. The obvious conclusion is that His 57 acts
as a general base catalyst that assists in removing the
proton from the OH of Ser 195, making that hydroxyl
group more nucleophilic than it would be otherwise.
This may happen after the OH group has started to
add to the substrate carbonyl.
The second nitrogen atom of His 57 is hydrogen
bonded to the carboxylate group of Asp 102, which is
in turn hydrogen bonded to two other groups. Aspar-
tate 102 has one of the few carboxylate side chains
that is buried inside the protein. To Blow,
230,231,239
the
structure suggested a charge-relay system by which
negative charges might move synchronously from Asp
102 to the imidazole which could then deprotonate the
hydroxyl group of Ser 195, allowing the serine oxygen
to add to the substrate carbonyl to form the tetrahedral
A
B
613
or oxyanion intermediate, which is depicted in step b
of Fig. 12-11. The small arrows in Fig. 12-10 also indi-
cate the movement of charge. Blow suggested that in
the extreme case a proton might be transferred also
from the His 57 imidazole to the Asp 102 carboxylate.
However, a variety of experiments, including studies
by
15
N NMR
241,242
and by neutron diffraction,
243
sug-
gest that the imidazole does not transfer its proton to
the carboxylate of Asp 102,
244,245
unless it does so
transiently, e.g., in a transition state complex. Instead,
the carboxylate and the imidazolium ions formed by
protonation of His 57 probably exist as a tight ion pair:
Support for this concept is provided by
1
H NMR
studies which have identified a downfield resonance
of the hydrogen-bonded proton in this pair at ~18 ppm
in chymotrypsinogen and chymotrypsin at low pH and
at ~ 14.9 15.5 ppm at high pH values.
246,247
Similar
resonances are seen in the -lytic protease,
248
in sub-
tilisin,
249
in adducts of serine proteases with boronic
acids
250,251
or peptidyl trifluoromethyl ketones,
252
in
alkylated derivative of the active site histidine,
253
and
in molecular complexes that mimic the Asp-His pair
in the active sites of serine proteases.
254
The catalytic cycle. Figure 12-11 depicts the
generally accepted sequence of reactions for a serine
protease. If we consider both the formation and the
subsequent hydrolysis of the acyl-enzyme intermediate
with appropriate oxyanion intermediates, there are at
least seven distinct steps. As indicated in this figure,
His 57 not only accepts a proton from the hydroxyl
Figure 12-11 Sequence
of chemical reactions
involved in the action of
a serine protease. The
oxyanion hole structure
has been omitted in the
right-hand column. The
imidazole ring may
rotate, as indicated by
the green arrow, to
provide two different
conformations.
c e
Acyl-enzymeproduct complex
Second conformation of oxyanion intermediate
Oxyanion intermediate
a
Free enzyme and substrate
Michaelis complex
b
b
f
Second oxyanion
Acyl-enzymeH
2
O complex
d
H
2
O
RNH
2
(product)
g
Product complex
Free enzyme + RCOOH
Possible
conformational
change
N N
H
His 57
H C
O
O
O
CH
2
C
R
N
O
R'
H N
N
H
H
Ser 195
Oxyanion hole

Asp 102
N N
H
H C
O
O
O
CH
2
C
R
N
O
R'
H N
N
H

+
N N H C
O
O
O
CH
2
C
R
N
O
R'
H

+
H
N N C
O
O
O
CH
2
C
H
2
N R'

H
R O

N N C
O
O
O
CH
2
C

H
R
O
H
O
H
N N C
O
O
O
CH
2
C

H
R

H
+
OH
O
N N C
O
O
O
CH
2

H
H
OH
C
R O
?
C. Displacement Reactions on Carbonyl Groups
N N H O C
O
H
+
614 Chapter 12. Transferring Groups by Displacement Reactions
group of Ser 195 (step b) but probably also functions in
protonation of the NH R leaving group (step c). An
unprotonated NH R would be such a poor leaving
group that the oxyanion intermediate would not go on
to acyl-enzyme. However, donation of a proton to that
leaving group from the protonated His 57 (general
acid catalysis) permits elimination of H
2
N R (step c).
The product of this step is the acyl-enzyme intermedi-
ate which must be hydrolyzed to complete the cata-
lytic cycle. This is accomplished through steps d f of
Fig. 12-11. The product R NH
2
is replaced by H
2
O,
which, in steps paralleling steps b and c, is converted
to an HO

ion that serves as the attacking nucleophile


to form (in step e) a second oxyanion intermediate
which is cleaved to the second product R COOH.
The water molecule that enters in step d, and which
participates in hydrolysis of the acyl enzyme, has
apparently been observed directly by time-resolved
Laue crystallography at low temperature.
255
The catalytic triad. The significance of the
charge-relay effect may not be fully understood but
the importance of the SerHisAsp cluster, which has
become known as the catalytic triad, cannot be doubted.
It has evolved independently in several subfamilies of
bacterial and plant proteases, a well-known example
of convergent evolution. This triad is also found
throughout a broad range of many different kinds of
enzymes and other proteins. In pancreatic enzymes
the catalytic triad consists of Ser 195His 57Asp 102
but in the bacterial subtilisin it consists of Ser 221His
64Asp 32 and in a wheat serine carboxypeptidase it
is Ser 146His 397Asp 338.
219
In all three cases the
folding patterns of the polypeptides are entirely differ-
ent but the geometry of the triad is the same. Another
folding pattern is seen in a protease encoded by cyto-
megalovirus, which contains an active site Ser 132His
63 pair.
256,257
Investigation of a host of mutant proteins also
demonstrates the importance of the catalytic triad.
For example, if either the histidine or the serine of the
triad of subtilisin was replaced by alanine the catalytic
activity decreased by a factor of 2 x 10
6
and replace-
ment of the aspartate of the triad by alanine decreased
activity by a factor of 3 x 10
4
.
229,258
When Asp 102 of
trypsin is replaced by asparagine the catalytic activity
falls by four orders of magnitude.
259
This may be in
part because the histidine in this mutant is hydrogen
bonded to Asn 102 as the tautomer with a proton on
N

, the nitrogen that should serve as the catalytic base


in step b (Fig. 12-11).
260
A mutant in which Ser 214
(see Fig. 12-10) was replaced with alanine is fully
active but charged residues in this position interfere
with catalysis.
261
Does the low-barrier hydrogen bond in the
catalytic triad play any special role in catalysis?
Blows suggestion of a charge relay from Asp 102 to
Ser 195 of chymotrypsin is probably correct. Some
theoretical calculations have indicated the possibility
of synchronous movement of the two protons in the
system during step a of the sequence shown in Fig.
12-11, with the proton in the strong hydrogen bond
to Asp 102 moving away from His 57 and toward the
midpoint distance of the hydrogen bond. Could the
presumed high energy of the short hydrogen bond
be harnessed to lower the transition state energy? A
realistic possibility is that the hydrogen bond increases
the polarizability of the catalytic triad, facilitating
movement of the substrate to the transition state.
Cassidy et al. suggested that the strong hydrogen bond
may be formed by compression of the triad resulting
from binding of substrate in the S
1
and S
2
subsites.
252
They suggested that this would raise the pK
a
of His 57
from ~ 7 in the free enzyme to 10 12, high enough to
enable it to remove the proton from the Ser 195 OH
group and low enough to allow the protonated form to
be the proton donor to the leaving group (step c, Fig.
12-11).
The oxyanion hole. A third mechanism by
which an enzyme can assist in a displacement reaction
on a carbonyl group is through protonation of the
carbonyl oxygen atom by an acidic group of the en-
zyme (Eq. 12-21). This will greatly increase the posi-
tive charge on the carbon atom making attack by a
nucleophile easier and will also stabilize the tetrahedral
intermediate. Although the carbonyl oxygen is very
weakly basic, it can interact with a suitably oriented
acidic group of the enzyme. In many serine proteases
this acidic function is apparently fulfilled by NH
groups of two amide linkages. In chymotrypsin these
are the backbone NH groups of Ser 195 and Gly 193
(Figs. 12-10,12-12). No actual transfer of a proton to
the carbonyl oxygen of the substrate is expected.
However, the NH groups are positive ends of amide
dipoles and can interact electrostatically with the
negative charge that develops on the oxyanion. The fit
of substrate into the oxyanion hole between the two
R
N
C
R
O
HB
R
N
C
R
O
B
H
R
N
C
R
O
H H
H
+
B

H
+
(12-21)
615
NH groups is apparently good only for the tetrahe-
drally bonded oxyanion intermediate,
262
a structure
thought to be close to that of the transition state. The
importance of the oxyanion hole for catalysis has been
supported by theoretical calculations
245
and by the
study of mutant enzymes.
263
In subtilisin, in which a
side chain of asparagine forms part of the oxyanion hole,
replacement of Asn with the isosteric Leu causes k
cat
to
fall by a factor of about 200 while K
m
is unaffected.
264
It is also of interest that thiono ester substrates, in
which the C = O of an oxygen ester has been replaced
by C = S, bind with normal affinity to chymotrypsin
but are not hydrolyzed at significant rates.
265
The formation of the oxyanion intermediate during
serine protease action is also supported by the existence
of tetrahedral forms of enzymes inhibited by substrate-
like aldehydes. The OH group of Ser 195 can add to
the carbonyl group to form hemiacetals. For example,
a
13
C-enriched aldehyde whose carbonyl carbon had a
chemical shift of 204 ppm gave a 94 ppm resonance as
it formed the tetrahedral hemiacetal with one of the
inhibitory aldehydes, N-acetyl-L-Leu-L-Leu-L-arginal
(leupeptin; Box 12-C). A natural product from a
species of Streptomyces, this aldehyde inhibits trypsin
and several other enzymes strongly.
266
Adducts of
wheat serine carboxypeptidases with aldehyde inhibi-
tors have also been observed.
267
While the carbonyl
group of the substrate amide linkage that is to be
cleaved apparently cant form strong hydrogen bonds
to the NH groups of the oxyanion hole, that of the
acyl-enzyme intermediate can, as judged by resonance
Raman spectroscopy.
268,269
The strength of the hydro-
gen bonding, as judged by the stretching frequency of
the C = O group, is correlated with the reactivity of the
acyl group.
269
Chymotrypsinogen and related proenzymes have
extremely low catalytic activity even though a major
part of the substrate binding site as well as the catalytic
triad system are already in place. However, the oxy-
anion hole is created during activation of the proen-
zyme by a subtle conformational change
197,262,271
that
involves the chain segment containing Gly 193 (Fig.
12-12). This is further evidence of the importance of
this part of the active site structure.
C. Displacement Reactions on Carbonyl Groups
Figure 12-12 Formation of the oxyanion hole following cleavage of trypsinogen between Lys 15 and Ile 16. (A) Stereoscopic
view. (B) Schematic representation. The newly created terminal NH
3
+
of Ile 16 forms a hydrogen-bonded ion pair with the
carboxylate of Asp 194. This breaks the hydrogen bond between Asp 194 and His 40 in trypsinogen, inducing the peptide
segment 192194 to shift from an extended conformation to a helical form in which the NH groups of Gly 193 and Ser 195 form
the oxyanion hole. Notice that the positions and interactions of Asp 102, His 57, and Ser 195, the catalytic triad, are very little
changed. From Birktoft et al.
270
N
H
His 40
N
O
C
O
H
O
H
H
N
C O
Gln 30
Met 192
S
CH
3
O
C
N
H H
N
O
C
O
Gly 193
C
H
Asp 194
Ser 95
Trypsinogen
O
H
N
His 57
H
N
O
C O
H
O
Asp 102
Ser 214
N
H
O
O
CH
3
Ser 214
Trypsin
192
S
O
O
N
N
N
H
H
C
R
O
H
H
N
C
O
N
C O
H
O
O
C
H
H
C
O
H
H
H
O
H
N
H
C
O
H
N
H
N
N
Gln 30
O Gly
193

+
194
Bound acyl
group from
substrate

Ile 16
A B
616 Chapter 12. Transferring Groups by Displacement Reactions
Stereoelectronic considerations. The amide
group that is cleaved by a protease is a resonance hybrid
of structures A and B of Fig. 12-13. The unshared pair
of electrons on the nitrogen atom of structure A and the
third unshared pair on the oxygen atoms of structure B
(shaded orbitals) have been drawn in this figure in such
a way that they are anti-periplanar to the entering
serine oxygen. This is required in the transition state
according to stereoelectronic theory. The newly created
(shaded) electron pair on oxygen is one of those that
hydrogen bonds to an NH group of the oxyanion hole.
The tetrahedral intermediate has another unshared pair,
which is not hydrogen bonded, and is antiperiplanar to
the HN-R leaving group. Thus, it appears to be set up
for easy elimination. Nevertheless, there is some doubt
about the need for adherence to the stereoelectronic
rule that two antiperiplanar lone pairs are necessary
for elimination.
There is another complication. The leaving group
NH R cannot be eliminated from the oxyanion until
it is protonated, presumably by the imidazolium group
of His 57. However, the proton on His 57 will be
adjacent to the proton that is already on this nitrogen
rather than to the unshared pair of electrons on the
same nitrogen atom. A conformational change in
which the tilt of the catalytic imidazole ring is altered
(step b, Fig. 12-11) or in which the ring rotates, may
have to precede the protonation of the leaving group
(step c, Fig. 12-11).
272 274b
This change may be assisted
by the presence of positive charge on the protonated
imidazole, but it still does not solve the problem.
Bizzozero and Butler suggested that a rapid inversion
of this chiral center on the NH R group may be
required prior to protonation and elimination.
273
This would explain their observation that N-alkylated
peptide linkages of otherwise fast substrates are not
cleaved by chymotrypsin. A high-resolution structure
determination demonstrated that a very slow elastase
substrate that contains N-methylleucine at the cleavage
site forms a normal ES complex.
275
However, inversion
would be hindered by the N-methyl group. An alter-
native in inversion would be a torsional rearrangement
of the substrate during binding.
276
The structure of
an acyl-enzyme intermediate with elastase has been
determined by crystallographic cryoenzymology. The
intermediate was allowed to accumulate at 26C,
after which the temperature was lowered to 50C and
the structure determined. The structure shows the
carbonyl group in the oxyanion hole as anticipated.
277
pH dependence. A plot of k
cat
/K
m
for chymo-
trypsin is bell shaped with a maximum around pH 7.8
and pK
a
values of 6.8 and 8.8. These represent pK
a
s of
the free enzyme (Eq. 9-57). That of 6.8 has been shown
to represent His 57. As the pH is lowered from the
optimum the affinity for substrate falls off as His 57
becomes protonated. The high pK
a
of 8.8 is thought
to belong to the N-terminal amino group of Ile 16, a
group that is generated during the conversion of the
proenzyme to active enzyme.
278
The Ile 16 amino
group forms an ion pair with Asp 194 (Fig. 12-12B)
which is next to the serine at the active center. This
salt linkage helps to hold the enzyme in the
required conformation for reaction and its
deprotonation at high pH causes a decrease
in substrate affinity.
279
The value of k
cat
is also pH dependent
and falls off at low pH around a pK
a
value
of from 6 7 depending upon the substrate.
However, no higher pK
a
affects k
cat
in the
experimentally accessible range. These
results provide the basis for believing that
an unprotonated His 57 is needed in the ES
complex for catalysis to occur. Similar
conclusions have been reached for other
serine proteases.
Substrate specificity. Like most other
enzymes, proteases display distinct prefer-
ences for certain substrates. These are often
discussed using the nomenclature of Fig.
12-14. The substrate residue contributing
the carbonyl of the amide group to be cleaved
is designated P
1
and residues toward the
N terminus as P
2
, P
3
, etc., as is shown in
Fig. 12-14. Residues toward the C terminus
from the peptide linkage to be cleaved are
designated P
1
, P
2
, etc. Chymotrypsin acts
H O
Ser 195
N H
N
His 57
H N
H
R
O
C

H N
H
R
O
C
+
H N
O
C
Tetrahedral
intermediate
O
Ser 195

N
H
H
N
C
Must be protonated
before elimination
Resonance
forms
Oxyanion
hole
A B
Figure 12-13 The stereochemistry of formation of a tetrahedral
intermediate by a serine protease. The most probable orientation of
groups as deduced by model building is shown. The shaded orbitals in
A and B are antiperiplanar to the entering oxygen of Ser 195. See
Polgr and Halsz.
272
617
most rapidly if the P
1
residue is one of the aromatic
amino acids.
280
Thus, the S
1
part of the substrate
binding site must bind preferentially to large, flat
aromatic rings. The crystal structure of chymotrypsin
showed this site to be composed of nonpolar side chain
groups. On the other hand, in trypsin the specificity
portion of the S
1
site is a deep specificity pocket
containing a fixed negative charge provided by the
carboxylate side chain group of Asp 189. This explains
why trypsin acts only upon peptide linkages containing
the positively charged arginine or lysine residues in
the P
1
position. In elastase the specificity pocket is
partly filled by nonpolar side chains and the enzyme
can accommodate only small P
1
side chains such as the
methyl group of alanine. Replacement of Asp 189 of
trypsin by lysine led to the predicted loss of specificity
for basic side chains. However, the mutant enzyme
did not become specific for negatively charged side
chains.
281
This is presumably because the lysine NH
3
+
group was not located in the same position as the Asp
189 carboxylate as a result of a different packing of
side chains between native and mutant enzymes.
Residues P
1
and P
2
also have major effects on substrate
binding by serine proteases.
282 284
Now many mutant
forms of subtilisin
225,285
and other serine proteases are
being made and are yielding a more sophisticated
understanding of the basis of the specificity of these
enzymes. An important factor that has emerged is
flexibility of surface loops in allowing an enzyme to
adjust its structure to give a better fit to some sub-
strates.
286,287
The specificity of the serine proteases is
also being exploited in the design of specific inhibitors
(Box 12-D).
Many serine proteases react with p-nitrophenyl-
acetate to give acetyl enzymes. However, its rate of
C. Displacement Reactions on Carbonyl Groups
Cleavage
S
5
S
4
S
3
S
2
S
1
S
1
S
2
S
3

N
H
N
H
O R
3
P
3
P
4
R
4
O
N
H
N
H
O R
1
P
1
P
2
R
2
O
N
H
N
H
O R
2

P
2

P
1

R
1
O
N
H
N
H
O
P
3

R
3

R
5
P
5
O
N
H
N
H
N
H
N
H
N
H
O O O
O O
N
H
N
H
Val 216
Ser 214 41 218
S
5
S
4
S
3
S
2
S
1
S
1
S
2

Gly 193
Ser 195
S
3

Figure 12-14 Standard nomenclature used to define the residues P


1
, P
2
. . .
toward the N terminus and P
1
', P
2
' . . . toward the C terminus of a peptide
substrate for a protease. The corresponding subsites of the protease are desig-
nated S
1
, S
2
. . . S
1
', S
2
'.
hydrolysis to give acetate is orders of magnitude slower
than that of acyl-enzymes derived from small substrates
such as the chymotrypsin substrate N-acetyltyrosine
amide.
In addition to the specificity-determining P
1
aromatic
side chain, the amide groups of this substrate can form
specific hydrogen bonds to the protein (Fig. 12-10).
These hydrogen bonds presumably help the enzyme
to recognize the compound, which is bound with
K
m
of ~ 0.03 M and is hydrolyzed (with liberation of
NH
3
)
288,289
with k
cat
~ 0.17 s
1
.
What happens when the length of the substrate
is extended in the direction of the N terminus? The
tyrosyl residue in the foregoing compound may be
designated P
1
. For extended substrates which contain
P
2
, P
3
, and additional residues (this includes most
natural substrates) the K
m
values decrease very little
from that of short substrates despite the larger number
of subsites to which the substrate is bound. However,
the maximum velocity is often much greater for the
extended substrates than for short ones. Thus, for
N-acetyltyrosyl-glycine amide K
m
is 0.017 M, only a
little less than for N-acetyltyrosine amide, but k
cat
is
7.5 s
1
, 440 times greater than for the shorter sub-
strate.
229,288,289
Other examples have been tabulated
by Fersht.
279
These observations suggest
that the binding energy that would be
expected to increase the tightness of
binding is, instead, causing an increase
in V
max
279,290
that is, it is reducing the
Gibbs energy of activation. How can
this be? Imagine that as the extended
substrate binds, for example, into
the subsite S
2
(which binds residue
P
2
), it must compress a spring in the
enzyme. Could not the compressed
spring now provide a source of
energy for assisting in peptide bond
cleavage? If this is the case, we must
ask what are the springs? Are
there amide linkages of the peptide
backbone that are distorted when
the substrate hydrogen bonds into
the site? How is the distortion
transmitted to the active site and
how does it stabilize the transition
H
3
C
C
N
C
C
O
H CH
2
O
NH
2
OH
H
N-Acetyltyrosine amide
618 Chapter 12. Transferring Groups by Displacement Reactions
state? We are far from understanding the answers to
these questions. However, it is of interest that one chain
of H-bonds that passes through the amide between P
1
and P
2
also passes through a carbonyl group of a
bulge (Fig. 12-10). A second chain of H-bonds that
connects to the same bulge passes through the back-
bone amide of the active site Ser 195, across the inter-
face between the two domains, and through the back-
bone of the second cylinder.
85
Perhaps when substrate
binds, the strengthening of the first H-bond chain
weakens the second through competition at the bulge,
allowing some subtle rearrangement in the active site
structure. We are talking here about the P
1
P
2
amide.
What happens at P
2
P
3
, and further along the chain in
both directions? It is important to understand these
effects; the phenomenon of increased reaction rate for
longer, more specific substrates is observed with many
proteases and other enzymes as well.
Another difference between small substrates and
longer, more specific substrates has been found in
studies of the effects of changing the solvent from pure
H
2
O to mixtures containing an increasing mole fraction
of
2
H
2
O. This is called a proton inventory.
291
For
the serine proteases the rates are decreased as the
2
H
content of the solvent increases, a fact that suggests
that some step involving a proton transfer, for example,
the deprotonation of Ser 195, is rate limiting. For simple
substrates, the effect is directly proportional to the mole
fraction of
2
H. However, for extended substrates, a
quadratic dependence on the mole fraction of
2
H is
observed. This suggests that a process involving syn-
chronous transfer of two protons, as in the postulated
charge relay system, may be more important in extended
substrates than in simple ones and may account for
the more rapid action on these substrates.
290,292
How-
ever, other interpretations of the data are possible,
leaving this conclusion uncertain.
229,291
2. The Cysteine Proteases (Thiol Proteases)
Papain from the papaya is one of a family of
enzymes that includes bromelain of the pineapple,
ficin of the fig, and actinidin of the kiwifruit.
293
Additional cysteine proteases
294
from the latex of
the papaya tree Carica papaya are known as caricain,
chymopapain,
294
and glycyl endopeptidase.
295
All
are members of a large superfamily which includes at
least 12 mammalian enzymes and many others from
both eukaryotic and prokaryotic organisms.
296
All
share with papain a characteristic structure which was
determined by Drenth and coworkers in 1968.
297
The
participating nucleophile in the active sites of these
enzymes is an SH group, that of Cys 25 in papain. An
adjacent imidazole from His 159 removes the proton
from the SH group to form a thiolate-imidazolium
pair.
298 300
The close proximity of the imidazolium group greatly
lowers the microscopic pK
a
of the Cys 25 thiol group
and the proximity of the resulting hydrogen-bonded
S

group greatly raises the microscopic pK


a
of the
imidazolium group.
Studies of the pH dependence of V
max
/k
cat
(Eq. 9-57)
reveal a bell-shaped dependence on pH with pK
a
values
301,302
of ~ 4 and ~ 9. However, the ion pair is
formed at a pH below four with apparent pK
a
values
of 2.5, 2.9, and 3.3 for ficin, caricain, and papain, re-
spectively.
302
These low values can be assigned princi-
pally to Cys 25 with only very small contributions from
His 159 (see Eq. 6-75). A third pK
a
, perhaps of a nearby
carboxylate from Glu 50, affects the rate. For caricain
the nearby Asp 158 (Fig. 12-15) has been implicated.
303,304
N
N
H
NH
C
O
O
C
N
H
H
O
Asn 175
H S
N
H
S
C
O
H
N
H
C
O
H
O
Ser 176
+
His 159
Asp 158
Gln 19

Cys 25

O
C
S
O
Figure 12-15 Schematic drawing of the active site of a
cysteine protease of the papain family with a partial struc-
ture of an acyl-enzyme intermediate in green. The thiolate-
imidazolium pair of Cys 25His 159 lies deep in the substrate-
binding cleft and bridges an interface between two major
structural domains, just as the SerHis pair does in serine
proteases (Fig. 12-10). This may facilitate small conforma-
tional changes during the catalytic cycle. Asn 175 provides a
polarizable acceptor for positive charge, helping to stabilize
the preformed ion pair, and allows easy transfer of an imida-
zolium proton to the product of substrate cleavage. The pep-
tide NH of Cys 25 and the side chain of Gln 19 form an
oxyanion hole.
N
N
H
H S
Cys 25
+
His 159

619
As shown in Fig. 12-15, the side chain of Asn 175
provides papain with a third member of a catalytic triad
analogous to that of serine proteases.
305
Glutamine 19,
together with the peptide backbone NH of Cys 25, pro-
vides an oxyanion hole.
306 308
Many studies, including
structure determinations on bound aldehyde and other
inhibitors, and observation by
13
C NMR
309
indicate that
thiol proteases act by addition of the thiolate anion to the
peptide carbonyl of the P
1
residue, just as in step b of
Fig. 12-11 (see also Fig. 12-12) for serine proteases.
308,310
However, alternative sequences of proton transfer have
been suggested.
311
A possible role for a strong hydrogen
bond has also been proposed.
310
For a detailed discussion
see Brocklehurst et al.
312
Most of the lysosomal proteases called cathepsins
are small 20- to 40-kDa glycoproteins found in all
animal tissues.
313
Most are cysteine proteases which
function best and are most stable in the low pH reducing
environment of lysosomes. They resemble papain in
size, amino acid sequence, and active site structures.
Papain is nonspecific but most cathepsins have definite
substrate preferences. Cathepsin B is the most abun-
dant. There are smaller amounts of related cathepsins
H (an aminopeptidase)
314
and L
315
and still less of cathep-
sins C, K, and others. Cathepsin B is both an endopep-
tidase and an exopeptidase.
316
It acts on peptides with
arginine at either P
1
or P
2
but also accepts bulky hydro-
phobic residues in P
1
and prefers tyrosine at P
3
.
317
Cathepsin S is less stable at higher pH than other
cathepsins and has a more limited tissue distribution,
being especially active in the immune system.
318,319
Cathepsin K is especially abundant in the bone
resorbing osteoclasts (Chapter 8). It is essential to
normal bone structure and its absence is associated
with the rare hereditary disease pycnodysostosis
(pycno) which causes short stature, fragile bones, and
skull and skeletal deformities.
320
It may also play a
role in the very common bone condition osteoporosis.
Cathepsin C is also called dipeptidyl peptidase. It
removes N-terminal dipeptides from many intracellular
proteins activating many enzymes, including some
other cathepsins.
321,322
A prohormone thiol protease
cleaves peptide chains between pairs of basic residues,
e.g., in the brain peptide precursor proenkephalin
(Chapter 30), and also on the N-terminal side of argi-
nine residues.
323
Pyroglutamate aminopeptidase
removes pyroglutamyl (5-oxoprolyl) groups from amino
termini of some peptides and proteins (see Fig. 2-4).
324
Another cysteine protease cleaves isopeptide linkages
such as those formed by transglutaminase or those
involving ubiquitin (Box 10-C).
325
Another cysteine
protease present in animal tissues was recognized by
its ability to hydrolyze the anticancer drug bleomycin.
This bleomycin hydrolase is a hexamer with a central
channel lined with papainlike active sites as in the
proteasome structure (Box 7-A)
326
The enzyme also
binds to DNA.
327
Unfortunately, cancer tissues often
contain high levels of the enzyme, whereas it is low in
skin and lung tissues, which are damaged by bleomycin.
The Ca
2+
-dependent neutral proteases called
calpains are found within the cells of higher animals.
The 705-residue multidomain peptide chain of a chicken
calpain contains a papain-like domain as well as a
calmodulin-like domain.
328
It presumably arose from
fusion of the genes of these proteins. At least six
calpains with similar properties are known.
329
Some
have a preference for myofibrillar proteins or neuro-
filaments.
330
They presumably function in normal
turnover of these proteins and may play a role in
numerous calcium-activated cellular processes.
331332a
A group of cysteinyl aspartate-specific proteases
(caspases) play an essential role in programmed cell
death (apoptosis).
333 335
Recall that nematodes are cell-
constant organisms. For maturation of Caenorhabditis
elegans 131 programmed cell deaths must occur at
specific stages of development. An essential gene
for this process was identified and named CED-3.
Deletion of this gene completely blocked the death
of these cells. CED-3 encodes a cysteine protease that
is highly homologous to the mammalian interleukin-
1-converting enzyme (ICE or caspase 1) which
cleaves the 31-kDa pro-interleukin-1 to form the
active 175 kDa species of this cytokine (Chapter 30).
At least ten caspases are known and many observations
have confirmed their role in apoptosis (see also Chap-
ter 32). In caspases 1 and 2 the side chains of Cys 285
and His 237 form the catalytic dyad and peptide NH
groups of Cys 285 and Gly 238 form the oxyanion
hole.
333
Parasites often use proteases in attacks on their
hosts. The cysteine protease cruzain is secreted by
the trypanosomes that cause Chagas disease and is
essential to their survival within the human body.
336,337
Cruzain is consequently an attractive target for devel-
opment of drugs for treating this major disease.
Among cysteine proteases of bacteria is a papain-
like enzyme from Clostridium histolyticum with a speci-
ficity similar to that of trypsin.
338
The anaerobic
Porphyromonas gingivalis, which is implicated in perio-
dental disease, produces both arginine- and lysine-
specific cysteine proteases designated gingipains.
339,339a
Some virally encoded cysteine proteases, including
one from the polio virus, have trypsin-like sequences
with the serine of the catalytic triad replaced by cys-
teine.
340,341
A human adenovirus protease also has a
CysHisGlu triad but a totally different protein fold.
342
Zymogens of cysteine proteases usually have a
long terminal extension which is removed, some-
times by autoactivation. Propapain has a 107-residue
extension.
343
The 322-residue cathepsin B carries an
unusually short 62-residue extension in its proenzyme
form.
315,343,344
In every case the N-terminal extension
folds into a domain, one of whose functions is to block
the active site cleft.
C. Displacement Reactions on Carbonyl Groups
620 Chapter 12. Transferring Groups by Displacement Reactions
3. N-terminal Nucleophile Hydrolases and
Related Enzymes
The most recently discovered group of proteases
are the N-terminal threonine hydrolases of the multi-
catalytic protease complex (MPC) of proteasomes.
The enzymes are arranged in a regular array inside
proteasomal compartments as shown in Box 7-A. The
active site is a catalytic dyad formed from the amino
group at the N terminus of the subunits.
345346a
Proteasomes of Thermoplasma contain a single type of
subunit but eukaryotic proteasomes contain subunits
with at least three distinct substrate preferences.
347 349c
They all appear to use the same hydrolytic mechanism
but in their substrate specificities they are chymotrypsin-
like, peptidylglutamyl-peptide hydrolyzing, branched
chain amino acid preferring, and small neutral amino
acid preferring based on the P
1
amino acid residue. In
the spleen some of the subunits of the proteasomes
appear to have been replaced by proteins encoded by
the major histocompatibility complex of the immune
system (Chapter 31).
347
This may alter the properties
of the proteasome to favor their function in antigen
processing. Proteasomes are also ATP- and ubiquitin-
dependent, as discussed in Section 6.
The enzyme glucosylasparaginase (aspartylgluco-
saminidase) is one of a group of other enzymes that use
N-terminal threonyl groups as catalytic dyads.
346,350 353
It removes N-linked glycosyl groups from asparagine
side chains of proteolytically degraded proteins and,
as indicated on the accompanying structural formula,
releases free aspartate and a 1-amino-N- acetylgluco-
samine-containing oligosaccharide. The amino group
is then released as NH
3
from the product by acid
catalysis in the lysosome.
350
Glutamine PRPP amidotransferase (Fig. 25-15) and a
penicillin acylase have similar active sites and overall
structures.
354
The NH
2
group is basic enough in the
environment of the protein to remove the proton from
the threonine OH group, activating it as indicated
below. Several serine proteases use an NH
2
group of
a lysine to form a serinelysine dyad.
346
Among these are the well-known E. coli leader pepti-
dase
355,356
and other signal peptidases.
357
These are
integral membrane proteins that cleave N-terminal
signal sequences from proteins incorporated into plasma
membranes. Another enzyme of this class is the lexA
repressor and protease discussed in Chapter 28.
A specific inhibitor of the major proteasomal activi-
ties is lactacystin, a compound formed by Streptomyces.
Lactacystin is converted reversibly, by loss of N-acetyl-
cysteine, into a -lactone known as clasto-lactacystin.
The N-terminal amino group attacks the reactive four-
membered ring of the lactone (Eq. 12-22).
358,359
N
C
C
N
Enzyme
H O
C
O
H
CH
3
H
H
H
C O (substrate)
H
Hydroxyl -amine
catalytic dyad
N
C
COO
NH
3
O
H
O
HO
NH
Oligosaccharide
OH
C O
CH
3
H
2
O

+
N
H
H
O CH
2
H
Serine
Lysine
NH
O
H
3
C
HO
C S
COOH
N
C
CH
3
O
O
CH
3
CH
3
HO
NH
O
H
3
C
O
C
O
N
CH
3
O
Enzyme
H
N
H
O
H
H
NH
O
H
3
C
HO
C
H
O
OH
N
CH
3
O
Enzyme
H
H
3
N
O
Lactacystin
H
clasto-Lactacystin
-lactone
N-Terminal threonine
Inhibited enzyme
H
OH
+
N-Acetylcysteine
(12-22)
621
The active sites of the N-terminal nucleophile
hydrolases are generated autocatalytically.
360 362
A single
peptide chain is cleaved to form and chains as in
the following diagram. An activating nucleophile such
as histidine removes a proton from an adjacent threo-
nine OH and the resulting alkoxide ion attacks the
adjacent peptide linkage, presumably via a tetrahedral
intermediate, to form an ester linkage. Compare this
sequence with reactions in Box 12-A and Eq. 14-41. Hydro-
lysis generates the N-terminal threonine as indicated:
4. The Aspartic Proteases
A fourth large group of protein-hydrolyzing en-
zymes consists of pepsin of the stomach
363
and related
enzymes.
364
Each of these ~ 320-residue proteins is
folded into two domains which associate with a pseudo-
twofold axis of symmetry that passes through the active
site.
365
A second human gastric aspartic proteinase is
gastricsin.
365
The related chymosin (rennin),
366
which
is obtained from the fourth stomach of the calf, causes
a rapid clotting of milk and is widely used in manufac-
ture of cheese. Pepsin has a broad specificity but cleaves
preferentially between pairs of hydrophobic residues,
converting proteins into soluble fragments. It is un-
usual in being able to cleave X-Pro peptide bonds.
367
Chymosin has a more restricted specificity, cutting
the -casein of milk between a PheMet bond. This
decreases the stability of the milk micelles, inducing
clotting.
366
The serum protein renin (distinct from
rennin), the lysosomal cathepsins D and E,
368 370
an
aspartyl aminopeptidase,
371
and various fungal
proteases are also closely related.
372 375
Renin,
376,377
which is synthesized largely in the kidneys, is involved
in blood pressure regulation (Box 22-D). More distantly
related aspartic proteases are encoded by retroviruses.
The pepsin family is most active in the low pH
range 1 5. All of the enzymes contain two especially
reactive aspartate carboxyl groups.
378
One of them
(Asp 215 in pepsin) reacts with site-directed diazoni-
um compounds and the other (Asp 32) with site-
directed epoxides.
379
It is attractive to think that one
of these carboxyl groups might be the nucleophile in a
double displacement mechanism. The second carboxyl
could then be the proton donor to the cleaving group.
The acyl-enzyme would be an acid anhydride. How-
ever, X-ray studies on pepsin
363,380
and on related fungal
enzymes such as penicillopepsin
381
and others
373,375
suggested a different possibility: A water molecule,
hydrogen bonded to one of the active site Asp carb-
oxylates or bridging between them, becomes the
nucleophile. A proton, held by the carboxylate pair,
protonates the substrate carbonyl to facilitate nucleo-
philic attack.
381
This is illustrated in Eq. 12-23 but with-
out detail. Several intermediate sequences of reaction
steps are possible.
382
At the beginning of the sequence
one of the two symmetrically placed carboxylates is
protonated while the other is not. Also notice that
although the active site is symmetric, the substrate is
bound asymmetrically as determined by its hydrogen
bonding into an extended binding site.
A characteristic feature of catalysis by the aspartic
proteases is a tendency, with certain substrates, to cata-
lyze transpeptidation reactions of the following type.
Acetyl-Phe-Tyr + Acetyl-Phe* Acetyl-Phe*-Tyr
Here Phe* is an isotopically labeled residue. Although
such reactions suggested the possibility of some kind
of activated amino group on the tyrosine that is cut off
in the initial cleavage of the unlabeled substrate, it is
more likely that the released tyrosine stays in the
active site,
383
while the acetyl-Phe fragment exchanges
with acetyl*-Phe.
C. Displacement Reactions on Carbonyl Groups
C
N C
chain
O CH
3
O H
O
H
N
COO
O
N
N
HN
His 150
Thr 152

H
chain
chain chain
H
C
N
C O
NH
P
1
H
H
H
C
P
1
'
H
O
O
D 32
H
O
H
O
O
D 215
C
N
C O
NH
P
1
H
H
C
P
1
'
H
O
O
D 32
H
O
O
D 215
Cleavage of
CN bond
O H
H
Formation of
tetrahedral
intermediate

H
H
O
H
H O
(12-23)
(12-24)
622 Chapter 12. Transferring Groups by Displacement Reactions
BOX 12-C SYNTHETIC PROTEASE INHIBITORS
One of the goals of synthetic medicinal chemis-
try is to design potent inhibitors of clinically impor-
tant proteases. Elastase inhibitors may be useful for
treatment of emphysema, pancreatitis, and arthritis,
a,b
while inhibitors of the angiotensinogen-converting
enzyme or of renin (Box 22-D) can help control blood
pressure. Inhibition of thrombin, factor Xa, or other
blood clotting factors (Fig. 12-17) may prevent blood
clots and inhibition of the cytosolic tryptase may
provide a new treatment for asthma. Inhibition of
the cysteine protease cathepsin K may help combat
osteoporosis and inhibition of cysteine proteases of
corona viruses may fight the common cold. Cysteine
proteases of schistosomes are also targets for pro-
tease inhibitors.
c
Many chemical approaches are used in design-
ing inhibitors. Often, a naturally occurring inhibitor
provides a starting point. The availability of high-
resolution structures of the target enzymes and of
various enzyme-inhibitor complexes assists in the
rational design of tight-binding inhibitors. Use of
combinatorial chemistry (Chapter 3)
d
is another
source of potential inhibitors. To be of practical use
in medicine many criteria of stability, solubility, and
low toxicity must be met. While most inhibitors
are disappointing as drugs, their use in laboratory
experimentation has clarified a great deal of bio-
chemistry.
A straightforward approach is to hunt for short
polypeptides that meet the specificity requirement
of an enzyme but which, because of peculiarities
of the sequence, are acted upon very slowly. Such
a peptide may contain unusual or chemically
modified amino acids. For example, the peptide
Thr-Pro-nVal-NMeLeu-Tyr-Thr (nVal = norvaline;
NMeLeu = N-methylleucine) is a very slow elastase
substrate whose binding can be studied by X-ray
diffraction and NMR spectroscopy.
e
Thiol proteases
are inhibited by succinyl-Gln-Val-Val-Ala-Ala-p-
nitroanilide, which includes a sequence common to
a number of naturally occurring peptide inhibitors
called cystatins.
f
They are found in various animal
tissues where they inhibit cysteine proteases.
A group of inhibitors such as leupeptin have
C-terminal aldehyde groups. Small oligopeptides
with this structure and with appropriate specificity-
determining side chains form tetrahedral hemiace-
tals, which may mimic transition state structures,
at the active sites of the target enzymes.
g,h
Leupeptin is a slow, tight-binding inhibitor of
trypsin. Some peptide aldehydes are potent, revers-
ible inhibitors of cysteine proteases forming hemith-
ioacetals with the active site cysteine.
i
Similarly,
peptide nitriles form thioimidate adducts.
h
The peptide boronic acids form adducts with
the active site serine of serine proteases.
j l
However, both X-ray crystallography
m
and
11
B
NMR
n
have shown that imidazole of the catalytic
triad may also add to the boronic acid and that a
tetrahedral adduct with both the serine oxygen and
histidine nitrogen covalently bonded to boron can
also be formed.
m
Thus, in reversibly inhibited
enzymes a mixture of different chemical species
may exist. Inhibitors can be designed to bind more
tightly by providing additional bonding opportuni-
ties. For example, a suitably placed cyano group on
a phenylalanine ring in the P
1
position of thrombin
C
N
N NH
2
NH
2
C
O
N
C
C
O
H
H
C
H O
H
N H
3
C
O
+
H
H H
H
Leupeptin (Actinomyces)
OH
C
R S Enzyme
H
NH
C
R S Enzyme
Hemithioacetal Thioimidate
Peptide N CH B
R
OH
OH
H
E Ser
OH
Peptide N CH B
R
H
H
+
OH
OH
O
Ser

Adduct
623
BOX 12-C (continued)
or other serine proteases can form a hydrogen bond
to the peptide NH of Gly 219 (see Fig. 12-9).
o
Numerous synthetic active-site directed or
enzyme-activated irreversible inhibitors have been
designed.
p
For example, the following chloroketone
inhibits chymotrypsin but does not act on trypsin.
The corresponding structure with a lysine side
chain (TLCK) inhibits only trypsin. These affinity
labeling compounds initially bind noncovalently
at the active site.
However, -chloroketones are powerful alkyl-
ating agents and the bound inhibitor attacks His
57 of the catalytic triad system. The reaction is
probably more complex than is indicated in the fore-
going equation and may involve an epoxy ether
intermediate.
q
Many other peptide chloromethyl
ketone inhibitors have been devised.
q,r
Isocoumarins inactivate many serine proteases.
For example, 7-amino-4-chloro-3-methoxyisocoumarin
acylates serine 195 of elastases as follows.
s
Many other enzyme-activated inhibitors are being
developed.
c,d,t
Epoxy groups, such as that of E-64, a compound
isolated from the culture medium of a species of
Aspergillus, react irreversibly with the active site
thiolate group of cysteine proteases.
i,u,v
Related
epoxides may become useful medications against
abnormal cathepsin levels.
All of the aspartic proteases are inhibited by
pepstatin, a peptide produced by some species of
Actinomyces and which contains two residues of the
unusual amino acid statine (sta).
w
Pepstatin has
the sequence Isovaleryl-L-Val-L-Val-Sta-L-Ala-Sta.
The statine residue mimics the noncovalently bonded
tetrahedral intermediate, permitting formation of a
very tight complex. Pepstatin is a poor inhibitor of
human renin but its existence has inspired the syn-
thesis of numerous related compounds, some of
which are effective renin inhibitors.
x,y
Some of these
inhibitors use the human angiotensinogen sequence
with a secondary alcohol group mimicking the
tetrahedral intermediate.
x,z
C. Displacement Reactions on Carbonyl Groups
O
O
O
O
N N CH
2
Cl
C N
H CH
2
S
H
CH
3
O
H
N N CH
2
C N
CH
2
S
H
CH
3
O
His 57

Cl
l-1-(p-Toluenesulfonyl)-amido-2-
phenylethyl chloromethyl ketone (TPCK)
H
His 57
O
O
OCH
3
Cl
H
2
N
O
Ser 195
CH
O
O
C
H
2
N Ser 195
Cl
OCH
3
O
C
O
O
C
H
2
N Ser 195
O
OCH
3
O
H
C
O
CH
3
C CH
3
O
O
7
H

Cl

C
N
N NH
2
NH
2
C
O
N
C
C
O
H CH
2
CH
H
3
C CH
3
C
O
H C O
O
H
+
H
H H

E-64, from Aspergillus japonicum


H
3
C COO
CH
3
H
H
NH
3
OH
+

Statine (Sta)
624 Chapter 12. Transferring Groups by Displacement Reactions
BOX 12-C SYNTHETIC PROTEASE INHIBITORS (continued)
These aspartic protease inhibitors are also lead
compounds in the development of inhibitors of
HIV protease.
aa cc
As in statine-based inhibitors,
the site of occupancy by the catalytic H
2
O (green in
Eq. 12-23) is occupied in the inhibitor by something
that mimics a tetrahedral intermediate with CHOH ,
PO
2
H, etc.
bb
Tremendous efforts are being expended
in designing these inhibitors and considerable
a
Powers, J. C., Oleksyszyn, J., Narasimhan, S. L., Kam, C.-M.,
Radhakrishnan, R., and Meyer, E. F., Jr. (1990) Biochemistry 29,
3108 3118
b
Mattos, C., Giammona, D. A., Petsko, G. A., and Ringe, D. (1995)
Biochemistry 34, 3193 3203
c
Seife, C. (1997) Science 277, 1602 1603
d
Peisach, E., Casebier, D., Gallion, S. L., Furth, P., Petsko, G. A.,
Hogan, J. C., Jr., and Ringe, D. (1995) Science 269, 66 69
e
Meyer, E. F., Jr., Clore, G. M., Gronenborn, A. M., and Hansen, H. A.
S. (1988) Biochemistry 27, 725 730
f
Yamamoto, A., Tomoo, K., Doi, M., Ohishi, H., Inoue, M., Ishida, T.,
Yamamoto, D., Tsuboi, S., Okamoto, H., and Okada, Y. (1992)
Biochemistry 31, 11305 11309
g
Ortiz, C., Tellier, C., Williams, H., Stolowich, N. J., and Scott, A. I.
(1991) Biochemistry 30, 10026 10034
h
Dufour, E., Storer, A. C., and Mnard, R. (1995) Biochemistry 34,
91369143
i
Mehdi, S. (1991) Trends Biochem. Sci. 16, 150 153
j
Bone, R., Shenvi, A. B., Kettner, C. A., and Agard, D. A. (1987)
Biochemistry 26, 7609 7614
k
Takahashi, L. H., Radhakrishnan, R., Rosenfield, R. E., Jr., and
Meyer, E. F., Jr. (1989) Biochemistry 28, 7610 7617
l
Nienaber, V. L., Mersinger, L. J., and Kettner, C. A. (1996) Biochemis-
try 35, 9690 9699
m
Stoll, V. S., Eger, B. T., Hynes, R. C., Martichonok, V., Jones, J. B., and
Pai, E. F. (1998) Biochemistry 37, 451 462
n
Zhong, S., Jordan, F., Kettner, C., and Polgar, L. (1991) J. Am. Chem.
Soc. 113, 9429 9435
o
Lee, S.-L., Alexander, R. S., Smallwood, A., Trievel, R., Mersinger, L.,
Weber, P. C., and Kettner, C. (1997) Biochemistry 36, 13180 13186
p
Bode, W., Meyer, E., Jr., and Powers, J. C. (1989) Biochemistry 28,
1951 1963
q
Kreutter, K., Steinmetz, A. C. U., Liang, T.-C., Prorok, M., Abeles, R.
H., and Ringe, D. (1994) Biochemistry 33, 13792 13800
Stereoscopic ribbon structure of the
HIV-1 protease with the synthetic
inhibitor Sequinivir
cc
bound in the
active site. One of the two identical
subunits (top) is shaded darker than
the second (bottom). When mutated,
the amino acid side chains shown in
ball-and-stick form with residue num-
bers shown for the top subunit led to
drug-resistant viruses. Courtesy of
Alex Wlodawer, National Cancer
Institute.
cc
success has been achieved. However, the rapid
development of mutant strains of the virus with
drug-resistant proteases presents a major challenge.
cc,dd
Mercaptans of suitable structure bind tightly
to Zn
2+
in the active sites of metalloproteases. For
example, captopril
ee
is a tight-binding competitive
inhibitor of the angiotensinogen-converting enzyme
that is effective in lowering blood pressure and the
first of many related inhibitors.
ff
As mentioned in the text, a vari-
ety of inhibitors that mimic the
geometry of a tetrahedral inter-
mediate or transition state are
also potent inhibitors of metallo-
proteases.
r
Wolf, W. M., Bajorath, J., Mller, A., Raghunathan, S., Singh, T. P.,
Hinrichs, W., and Saenger, W. (1991) J. Biol. Chem. 266, 17695 17699
s
Meyer, E. F., Jr., Presta, L. G., and Radhakrishnan, R. (1985) J. Am.
Chem. Soc. 107, 4091 4094
t
Groutas, W. C., Kuang, R., Venkataraman, R., Epp, J. B., Ruan, S.,
and Prakash, O. (1997) Biochemistry 36, 4739 4750
u
Yamamoto, D., Matsumoto, K., Ohishi, H., Ishida, T., Inoue, M.,
Kitamura, K., and Mizuno, H. (1991) J. Biol. Chem. 266, 14771 14777
v
Varughese, K. I., Su, Y., Cromwell, D., Hasnain, S., and Xuong, N.-h.
(1992) Biochemistry 31, 5172 5176
w
Gmez, J., and Freire, E. (1995) J. Mol. Biol. 252, 337 350
x
Cooper, J., Quail, W., Frazao, C., Foundling, S. I., Blundell, T. L.,
Humblet, C., Lunney, E. A., Lowther, W. T., and Dunn, B. M. (1992)
Biochemistry 31, 8142 8150
y
Tong, L., Pav, S., Lamarre, D., Pilote, L., LaPlante, S., Anderson, P. C.,
and Jung, G. (1995) J. Mol. Biol. 250, 211 222
z
Cooper, J. B., Foundling, S. I., Blundell, T. L., Boger, J., Jupp, R. A.,
and Kay, J. (1989) Biochemistry 28, 8596 8603
aa
Hui, K. Y., Manetta, J. V., Gygi, T., Bowdon, B. J., Keith, K. A.,
Shannon, W. M., and Lai, M.-H. T. (1991) FASEB J. 5, 2606 2610
bb
Abdel-Meguid, S. S., Zhao, B., Murthy, K. H. M., Winborne, E., Choi,
J.-K., DesJarlais, R. L., Minnich, M. D., Culp, J. S., Debouck, C.,
Tomaszek, T. A., Meek, T. D., and Dreyer, G. B. (1993) Biochemistry
32, 7972 7980
cc
Ridky, T., and Leis, J. (1995) J. Biol. Chem. 270, 29621 29623
dd
Chen, Z., Li, Y., Schock, H. B., Hall, D., Chen, E., and Kuo, L. C.
(1995) J. Biol. Chem. 270, 21433 21436
ee
Vidt, D. G., Bravo, E. L., and Fouad, F. M. (1982) N. Engl. J. Med. 306,
214 219
ff
Gros, C., Nol, N., Souque, A., Schwartz, J.-C., Danvy, D.,
Plaquevent, J.-C., Duhamel, L., Duhamel, P., Lecomte, J.-M., and
Bralet, J. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 4210 4214
625
Pepsin is secreted as the inactive pepsinogen, which
is activated by H
+
ions at a pH below 5. Determination
of its crystal structure revealed that in the proenzyme
the N-terminal 44-residue peptide segment lies across
the active site, blocking it.
384
At low pH the salt bridges
that stabilize the proenzyme are disrupted and the
active site is opened up to substrates.
While the cellular aspartate proteases are over 300
residues in length, the retroviral proteases are less than
one-half this size.
385 388
That of the human HIV-I protease
contains only 99 residues. These enzymes are cut from
a polyprotein (encoded by the viral gag and pol genes
(Fig. 28-26). The pol gene encodes four other essential
enzymes as well, and these are cut apart at eight dif-
ferent sites by action of the protease.
388
Despite its
small size, it displays sequence homologies with the
larger cellular aspartate proteases and has a related
three-dimensional structure. Each chain has only one
active site aspartate; the functional enzymes are dimers
and the catalytic mechanism appears to be similar to
that of pepsin.
388 392
A great deal of effort is being
devoted to designing synthetic HIV protease inhibitors,
which are used in the treatment of AIDS (Box 12-C).
5. Metalloproteases
The pancreatic carboxypeptidases are character-
ized by the presence of one firmly bound zinc ion in
each molecule. The Zn
2+
can be removed and can be
replaced by other metal ions such as Co
2+
and Ni
2+
,
in some cases with reconstitution of catalytic activity.
The human pancreas synthesizes and secretes proen-
zymes for two forms of carboxypeptidase A, with a
preference for C-terminal hydrophobic residues, as
well as carboxypeptidases B, which prefer C-terminal
basic residues. Additional A and B forms
393
as well as
more specialized nondigestive carboxypeptidases are
also known. In eukaryotic cells the latter participate
in processing of proteins. Following removal of N-
terminal signal sequences, processing often continues
with removal of basic residues from the C termini by
carboxypeptidases N, H (also called E or enkephalin
convertase),
394
and M, all of which are metalloen-
zymes.
395,396
Carboxypeptidase N removes C-terminal
arginines from many biologically important peptides.
It also circulates in the plasma and protects the body
by inactivating such potent inflammatory peptides as
the kinins and anaphylotoxins.
397
Carboxypeptidase
H is located in secretory granules, while carboxypep-
tidase M is membrane associated.
396
Dipeptidyl car-
boxypeptidase (angiotensin-converting enzyme)
removes the C-terminal ProPhe dipeptide from
angiotensinogen to generate the potent pressor agent
angiotensin I (Box 22-D) and cleaves dipeptides from
many other substrates as well.
398
A D-alanyl-D-alanyl
carboxypeptidase cleaves D-alanine from the ends of
cell wall peptides (Chapter 20).
399
Another well-known zinc-containing enzyme is
thermolysin, a nonspecific endopeptidase widely used
in laboratories. Produced by a themophilic bacterium,
it is unusually resistant to heat. It contains four bound
calcium ions in addition to the active site zinc.
400 402
The active site structure resembles that of pancreatic
carboxypeptidase A and the two enzymes appear to
act by similar mechanisms.
401,403,404
The mammalian
zinc endopeptidase neprilysin, an integral membrane
protein involved in inactivation of enkephalins and
other signaling peptides, also resembles thermolysin.
405
A related neutral endopeptidase is the product of a
gene called PEX (phosphate-regulating gene with
homologies to endopeptidases on the X chromosome).
The absence of the PEX gene product causes X-linked
hypophosphatemic rickets which leads to excessive
loss of phosphate from the body with defective miner-
alization of bone.
406
The mitochondrial processing
peptidase, which removes signal sequences from the
N termini of mitochondrial proteins, also contains Zn
2+
at its active site.
407,408
However, it is an heterodimer
and a member of an additional family of enzymes, one
of which includes human insulin-degrading enzyme.
In both carboxypeptidase A and thermolysin the
active site Zn
2+
is chelated by two imidazole groups
and a glutamate side chain (Fig. 12-16). In carboxypep-
tidase A, Arg 145, Tyr 248, and perhaps Arg 127 form
hydrogen bonds to the substrate. A water molecule is
also bound to the Zn
2+
ion. The presence of the posi-
tively charged side chain of Arg 145 and of a hydro-
phobic pocket accounts for the preference of the enzyme
for C-terminal amino acids with bulky, nonpolar side
chains. The Zn
2+
in thermolysin is also bound to two
imidazole groups and that in D-alanyl-D-alanyl carboxy-
peptidase to three.
The presence of a zinc ion in the metalloproteases
immediately suggested a role in catalysis. Unlike
protons, which have a weak affinity for the oxygen
of an amide carbonyl group, a metal ion can form a
strong complex. If held in position by other ligands
from the protein, a properly placed zinc ion might be
expected to greatly enhance the electrophilic nature of
the carbon atom of the C = O group. However, it has
been difficult to establish the exact mechanism of
action.
410
Carboxypeptidase A cleaves both peptides
and ester substrates. For peptides, K
m
is the same for
various metals while k
cat
changes, but the converse is
true for ester substrates.
411
From its position Glu 270
(Glu 143 in thermolysin) seems to be the logical nucleo-
phile to attack the substrate to form an acyl-enzyme
intermediate, an anhydride. Using the following
specific ester substrate, Makinen et al. showed that at
very low temperatures of 40 to 60C, in solvents
such as 50:50 ethylene glycol:water, an acyl-enzyme
intermediate can be detected spectroscopically. It
could even be separated from free enzyme
412
by gel
C. Displacement Reactions on Carbonyl Groups
626 Chapter 12. Transferring Groups by Displacement Reactions
filtration at 60C and its cyanoborohydride reduction
product was characterized.
413
The intermediate appears to be the acid anhydride
formed by Glu-270.
414,415
The conformation of the
intermediate was deduced by ENDOR spectroscopy
and its formation and reaction interpreted according
to stereoelectronic principles.
416
If the anhydride mechanism is correct, the water mole-
cule bound to the Zn
2+
probably provides an HO

ion
necessary for the hydrolysis of the intermediate anhy-
dride.
417
Although these results seem convincing there are
objections.
410
The mechanism deduced for hydrolysis
of an ester may be different than that for a peptide.
418
N
H
N
H196
O
O
Zn O
H

N
NH
O
C
H
H
N
H
N
H
P
1
O
H
P
2
E270
O

O
Y248
O
H
H
O
H
C
O
H
N H
O H N
N
H
H
+
R145
Hydrophobic cavity
Peptides with C-terminal
amino acids with aromatic
or bulky, branched side
chains are best substrates
E72
Attacking nucleophile
may be COO

or H
2
O
with general base
catalysis by COO

Bond cleaved
by enzyme
H69
2+
Figure 12-16 Structure of the active site of carboxypeptidase
A with a peptide substrate present. See Christianson and
Lipscomb.
409
C
O
O
C
COO
CH
2 H

O-(trans-p-cinnamoyl)-l--phenyllactate
C
O
O
C
O
Glu 270
Furthermore, many observations favor an alternative
mechanism. A hydroxide ion derived from a water
molecule bound to the zinc ion may be the initial
attacking nucleophile.
404,419 422
Both X-ray crystallo-
graphic studies and EPR investigations
403,414
show that
the zinc in carboxypeptidases can coordinate at least
five surrounding atoms. As shown in Fig. 12-16, the
Zn
2+
could hold the attacking water molecule and, at
the same time, provide the positive charge for stabiliz-
ing the resulting tetrahedral intermediate. Glu 270 (or
in thermolysin His 231) acts as a base to deprotonate
the bound H
2
O. Mock and Stanford argue that the
H
2
O is probably displaced when the peptide carbonyl
binds and that the H
2
O is then deprotonated and adds
to the polarized carbonyl.
404
The pH dependence of the action of carboxypep-
tidase A is determined by pK
a
values of ~ 6 and ~ 9.5
for the free enzyme
422
and of ~ 6.4 and ~ 9 for k
cat
.
420
For thermolysin the values for k
cat
are ~ 5 and ~ 8.
404
Assignment of pK
a
values has been controversial. They
may all be composites of two or more microscopic
constants but probably, at least for carboxypeptidase,
the low pK
a
is largely that of Glu 270 while the high
one represents largely the dissociation of a proton
from the zinc-bound H
2
O.
Earlier studies of carboxypeptidase had indicated
that Tyr 248 moves its position dramatically upon sub-
strate binding, and it was suggested that its phenolic
OH group protonates the leaving group in the acylation
step. However, a mutant in which Tyr 248 was replaced
by phenylalanine still functions well.
423
Phosphonamidates,
424,425
phosphonate esters, and
oxo-methylene substrate analogs, which presumably
mimic the geometry of the tetrahedral intermediate or
transition state of the intermediate or transition state
627
of the catalytic cycle, are effective inhibitors of zinc
proteases. X-ray studies of complexes of such inhibitors
with both thermolysin and carboxypeptidase A sup-
port the suggestion that the Zn
2+
binds the carbonyl
oxygen of the amide bond that is to be cleaved in a
substrate and that a glutamate side chain could activate
a Zn
2+
-bound water to form an attacking nucleophile.
A similar conclusion was reached from the structure of
a thermolysinproduct complex.
426
Another large group of zinc proteases are the
matrix metalloproteases which act on proteins of
the extracellular matrix, such as collagen, proteogly-
cans, and fibronectin.
427 429c
These enzymes have been
classified as collagenases, gelatinases (which act on
denatured collagen), stromelysins (which are activated
in response to inflammatory stimuli),
430 432
and a mem-
brane type. The group also includes the digestive en-
zyme astacin, from the crayfish,
433
and metalloproteases
from snake venoms.
429,434
The matrix metalloproteases
are essential to the remodeling of the extracellular
matrix that occurs during wound healing, tissue growth,
differentiation, and cell death. An example of tissue
remodeling is the development of dental enamel (Box
8-G). The proteinaceous matrix formed initially must
be digested away, perhaps by the metalloprotease
enamelysin, and replaced by the dense mineral of
enamel.
435
Excessive secretion of collagenase by fibro-
blasts is observed in rheumatoid arthritis and other
inflammatory conditions.
At least 64 different matrix metalloproteins are
known.
427
Each enzyme consists of three domains.
An 80- to 90-residue N-terminal propeptide domain
contains a cysteine whose S

group binds to the active


site zinc, screening it from potential substrates. The
central catalytic domain is followed by a hinge region
and a C-terminal domain that resembles the serum
iron binding and transporting hemopexin.
427,436
The
mechanism of action is probably similar to that of
thermolysin.
430
C. Displacement Reactions on Carbonyl Groups
C
P
N
C
O O
H
C
P
O
C
O O

C
C
CH
2
C

1
Phosphonamidate

O
Phosphonate ester
Oxo-methylene analog
Many aminopeptidases are metalloenzymes.
437
Most studied is the cytosolic leucine aminopeptidase
which acts rapidly on N-terminal leucine and removes
other amino acids more slowly. Each of the subunits
of the hexameric enzyme contains two divalent metal
ions, one of which must be Zn
2+
or Co
2+
.
438,439
A me-
thionine aminopeptidase from E. coli contains two
Co
2+
ions
440,441
and a proline-specific aminopeptidase
from the same bacterium two Mn
2+
.
442
In all of these
enzymes the metal ions are present as dimetal pairs
similar to those observed in phosphatases and discussed
in Section D,4 and to the Fe Fe pairs of hemerythrin
and other diiron proteins (Fig. 16-20). A hydroxide ion
that bridges the metal ions may serve as the nucleophile
in the aminopeptidases.
438
A bound bicarbonate ion
may assist.
438a
A metalloenzyme peptide deformylase removes
the formyl groups from the N termini of bacterial pro-
teins. Although the active site is similar to that of
thermolysin,
443
the Zn
2+
form of peptide deformylase
is unstable. Both Ni
2+
and Fe
2+
form active, stable
enzymes.
444,445
6. ATP-Dependent Proteases
Much of metabolism is driven by the Gibbs energy
of hydrolysis of ATP so it shouldnt be surprising that
ATP is sometimes rather directly involved in hydrolytic
degradation of polypeptide chains. Much of protein
processing occurs in the endoplasmic reticulum,
446
which also assists in sorting unneeded and defective
proteins for degradation in the ubiquitinproteasome
system. Proteasomal degradation occurs in the cytosol,
in the nucleus, and along the ER. However, the pre-
dominant location is the centrosome.
446a
Polyubiquitination of proteins requires ATP (Box
10-C) and additional ATP is utilized in the proteasomes
(Box 7-A) during the selection of polyubiquitinated
proteins for hydrolysis.
447,448
With 28 subunits the 26S
proteasome is complex and not fully understood.
448
The ATP-hydrolyzing subunits appear to all be in the
cap regions. Is the ATP used to open and close the
entry pores? To induce conformational changes in all
subunits as part of a catalytic cycle? Or to unfold
folded proteins to help them enter the proteasomes?
449
N
H
C
N H
Substrate
O
H
O
H
R
H
+ 2
M
+ 2
M

628 Chapter 12. Transferring Groups by Displacement Reactions


Some answers may be obtained from smaller
bacterial, mitochondrial, and chloroplast ATP-dependent
proteases. Cells of E. coli contain at least nine proteases,
which have been named after the musical scale as Do,
Re, Mi, Fa, So, La, Ti, Di, and Ci.
450,451
Most are serine
proteases but two, Ci and Pi, are metalloproteins.
Protease La (Lon protease, encoded by gene lon)
has attracted particular attention because the hydrolysis
of two molecules of ATP occurs synchronously with
cleavage of a peptide linkage in the protein chain.
452
This enzyme, as well as protease Ti (more often
called Clp, for caseinolytic protease),
451,453,454
is
ATP-dependent.
451
The Lon protease of E. coli is a large 88-kDa serine
protease with the catalytic domain, containing active
site Ser 679, in the C-terminal half. The N-terminal
portion contains two ATP-binding motifs and a linker
region.
455,456
A homologous protein known either as
Lon or as PIM1 is present in mitochondria.
457,458
The
E. coli proteases Clp (Ti) include ClpAP and ClpXP,
which are heterodimers, each containing the catalytic
subunit ClpP and either ATPase ClpA or ClpX. The
active enzyme may be designated ClpAP.
459 461
A
homolog of ClpP has been found in human mitochon-
dria.
461a
The ATPase Clp forms seven-subunit rings
resembling the rings of proteasomes (Box 7-A), while
the catalytic subunit ClpA forms six-membered
rings.
451,454
Despite the mismatch in symmetry and
the fact that they are serine proteases, these enzymes
appear similar to proteasomes. However, each catalytic
subunit has an ATPase neighbor. Why is it needed?
The related ClpB is both an ATPase and a chaperonin. It
is essential for survival of E. coli at high temperatures.
461b
Another ATP-dependent protease identified
among heat shock proteins of E. coli is known as
Hs1VHs1U or (ClpQClpY). It has a threonine pro-
tease active site and is even more closely remeniscient
of eukaryotic proteasomes.
462463a
Also active in E. coli
is another ring-like protease, a membrane-bound zinc
endopeptidase FtsH (or HflB).
463b
Similar eukaryotic
proteases also exist.
460
7. The Many Functions of Proteases
Many of the enzymes considered in the preceding
sections function within the digestive tract. Others
function in the processing of newly formed peptide
chains, while others act in the intracellular degradation
of proteins. Yet others are secreted from cells and
function in the external surroundings. Both processing
and intracellular degradation can be viewed as parts
of biosynthetic loops (Chapter 17) that synthesize mature
proteins, and then degrade them when they have
served their function or have become damaged. The
pathways involved are varied and complex, a natural
result of thousands of enzymes and other compounds
acting on accessible and chemically appropriate parts
of the proteins. Evolutionary selection has evidently
led to the particular set of pathways that we observe
for any organisms.
Because they must often cleave large polyproteins,
many viruses encode processing proteases.
464 466b
For
example, the entire RNA genome of the poliovirus
encodes a large polyprotein which is cut by two virally
encoded chymotrypsin-like cysteine proteases within
Tyr-Gly and Gln-Gly sequences;
341,465
Asn-Ser sequenc-
es are also cut, apparently autocatalytically. As we
have already seen, retroviruses encode their own
aspartic protease. Most cellular and secreted proteins
of bacteria or eukaryotes also undergo processing.
This ranges from removal of N-formyl groups to cutting
off signal peptides, addition or formation of prosthetic
groups, internal cleavages, and modifications at the
C termini. A variety of peptidases are required.
Degradation of proteins, which converts them
back to amino acids as well as other products, takes
place in part in the cytosol via the ubiquitin-protease
system. Proteolysis also occurs in ER and external cell
spaces through the action of membrane-bound and
secreted proteases. Loosely folded proteins, which can
arise in various ways, are subject to rapid degradation.
For example, synthesis of a polypeptide chain on a
ribosome may be accidentally disrupted with formation
of a protein with a shorted chain. Mature proteins
may become damaged. For example, certain histidine
residues are readily attacked by oxidizing reagents.
Oxidative damage may mark proteins for rapid degen-
eration.
450
In E. coli, proteases So and Re attack oxi-
dized glutamine synthetase much more rapidly than
the intact native enzyme.
450,467
In mammalian cells the
proteasomes degrade oxidized as well as poorly folded
proteins.
468
However, proteasomal digestion is not
complete and peptide fragments may be secreted.
Peptide fragments of proteins are also formed in the ER
and may be secreted. Thus, a variety of small peptide
hormones and other biologically active peptides are both
generated and inactivated in or around cells.
469 471
Some receptors are activated by proteases.
471a
Circulating proenzymes of the blood clotting factors,
of the complement system (Chapter 31), represent a
specialized group of secreted signaling proteins that
are able to initiate important defensive cascades. Pro-
teases also act more directly in defense systems of the
body. For example, serine proteases cause lysis of the
target cells of cytotoxic T lymphocytes
472
(Chapter 31)
and activated neutrophils
473
(Chapter 18). At the same
time, pathogenic bacteria often secrete proteases that
assist in attack on their hosts
474
and schistosomes secrete
an elastase that helps them penetrate skin and invade
their hosts.
475
629
8. Protease Inhibitors of Animals and Plants
Premature conversion of proenzymes such as
trypsinogen into active proteases in the pancreas
would be disastrous. To prevent this, the pancreas also
produces inhibitors. The complex cascades that control
blood clotting would also be unstable were it not for
the presence in blood of numerous inhibitory proteins.
Indeed, inhibitors of proteases are found everywhere
in animals, plants, and microorganisms.
476 478
They
are usually proteins but small antibiotic protease
inhibitors are also produced by microorganisms. Pro-
tein inhibitors are usually specific for a given type of
enzyme: The serpins inhibit serine proteases, and the
cystatins inhibit cysteine proteases.
479 483
Other
inhibitors block the action of metalloproteases
484 486
and aspartic proteases. Inhibitors help not only host
organisms but also parasites. For example, Ascaris
suum, a very large nematode that is thought to infect
1/4 of the human population of the earth and nearly
all of the pigs, secretes a pepsin inhibitor.
487
However,
the large 720-kDa serum -macroglobulins (Box 12-D)
inhibit all classes of proteases.
About 20 families of protein inhibitors of proteases
have been described.
488
The egg white ovomucoids
comprise one family. Turkey ovomucoid is a three-
domain protein whose 56-residue third domain is a
potent inhibitor of most serine proteases.
488,489
The 58-
residue pancreatic trypsin inhibitor
490
is a member
of another family of small proteins. A 36-residue
insect (locust) protease inhibitor is even smaller.
491
Inhibitors that block the action of trypsin and other
proteases are found in many plants. The inhibitor
activity is usually highest in seeds and tubers, but
synthesis of inhibitors can be induced in other parts
of plants e.g. tomato and potato by wounding.
492
See also Chapter 31. Legumes contain small 60- to
76-residue inhibitors (BowmanBirk inhibitors) each
containing seven disulfide linkages, and they are
relatively stable toward cooking and toward acid
denaturation in the stomach.
493,494
Although they
interfere with protein digestion they seem to have an
anticarcinogen effect.
493
Soybeans also contain a larger
181-residue trypsin inhibitor (Kunitz type).
495
Several inhibitorprotease complexes have been
crystallized and details of their interactions are known.
For example, the pancreatic trypsin inhibitor binds at
the active site of trypsin with K
f
>10
13
M
1
at neutral
pH.
496
The two molecules fit snugly together,
490,497
the
inhibitor being bound as if it were a peptide substrate
with one edge of the inhibitor molecule forming an
antiparallel structure with a peptide chain in the
enzyme. Lysine 15, which forms part of this structure,
enters the specific P
1
binding site for a basic amino
acid in a substrate. Thus, the protease inhibitor is a
modified substrate which may actually undergo attack
at the active site. However, the fit between the two
molecules is so tight that it is hard for a water molecule
to enter and complete the catalytic act. The complex
reacts very slowly, keeping the enzyme inactive. There
is not enough inhibitor to interfere with the large
amounts of trypsin formed from trypsinogen in the
small intestine so that trypsin can function there.
Most protease inhibitors act by mechanisms similar
to that of the pancreatic trypsin inhibitor. They are
very slow substrates with a reactive loop that carries
suitable P
1
, P
2
, and P
1
' residues that meet the specificity
requirements of the enzyme. Additional noncovalent
interactions prevent dissociation and make the energy
barrier for hydrolysis so high that the reaction is ex-
tremely slow.
488,494,495
The serpins are larger ~ 400-residue proteins.
498 500
They also form complexes in which hydrolysis and
release of the serpin occurs very slowly. However,
structural analysis before reaction and after release
showed that a major rearrangement occurs in the
serpin structure. The P
1
through ~ P
15
residues of the
cleaved reactive loop are inserted into the center of the
main sheet of the serpin, leaving the P
1
and P
1
' residues
~ 7 nm apart.
499,500
It seems likely that the rearrange-
ment begins during formation of the tight inhibited
complex, which cannot be dissociated by boiling in a
sodium dodecyl sulfate (SDS) solution and which may
be an acyl-enzyme.
501,502
A serpin molecule can act
only once.
Blood contains several serpins. They are abundant,
accounting for about 10% of the total protein of human
plasma.
478,500
The most abundant is the 1-protease
inhibitor or
1
-antitrypsin, a 394-residue glycoprotein
component of the -globulin fraction of blood serum.
500a
There is no trypsin in tissues, but this inhibitor blocks
the action of other serine proteases, including cathepsin
G and leukocyte elastase.
503
Hereditary absence of

1
-protease inhibitor often leads to severe pulmonary
emphysema at an early age. Elastase released by
neutrophils at sites of inflammation degrades many
components of connective tissue including elastin,
collagen, and proteoglycans. Without the presence
of protease inhibitor too much damage is done to
surrounding tissue. A lack of this inhibitor is one of
the commonest genetic defects among persons of
European ancestry, affecting 1 in 750 persons born.
504 506
The most serious known mutation is a replacement of
guanine by adenine at a specific point in the DNA and
a resultant replacement of Glu 342 with lysine in the
protease inhibitor. This in some manner adversely
affected the processing and secretion of the protein.
At its reactive site the
1
-protease inhibitor has the
sequence
Ala-Ile-Pro-Met*-Ser-Ile-Pro-Pro,
the Met-Ser pair marked by the asterisk fitting into the
P
1
-P
1
' sites (Fig. 12-14). The methionine (Met 358) in
C. Displacement Reactions on Carbonyl Groups
630 Chapter 12. Transferring Groups by Displacement Reactions
BOX 12-D MOLECULAR MOUSETRAPS
The large 720-kD
2
-macroglobulin of human
serum, as well as related proteins of vertebrate and
invertebrate circulatory systems and of egg whites
of birds and reptiles, is a trap for proteases.
af
Human
macroglobulin is a homotetramer consisting of two
pairs of identical 180-kDa subunits, each pair being
held together in an antiparallel configuration by
two disulfide bridges. Each subunit contains a
bait region with cleavage sites appropriate for
nearly all known endoproteases
f
and also a thioester
linkage as explained later. Electron microscopic
reconstructions of the native protein and its com-
plexes with proteases show that a major structural
transformation occurs.
e,f
The macroglobulin traps
two protease molecules of the size of trypsin, or one
larger one such as plasmin, in an internal cavity.
The internal thioesters, which are formed between
Cys 949 and Gln 952 (with loss of NH
3
) in each
subunit, become reactive
g
and form covalent bonds
with -amino groups of various lysine side chains of
the trapped proteases.
The serum proteins C3 and C4, members of the
complement system (Chapter 31), are converted
into their active forms, C3b and C4b, by proteolytic
removal of short N-terminal peptide fragments.
Both C3b and C4b bind tightly to cell surfaces, a
feature that helps to direct the complement systems
attack to the surfaces of invading organisms. This
tight binding also involves covalent attachment of
macromolecules by reaction with a preformed thio-
ester just as with
2
-macroglobulin.
h j
In fact, the
thioester linkage was first discovered in the comple-
ment proteins. Both C3 and C4 contain the thioester
CH
2
N N
O
CH
2
HN
O
H
H
H
COO
O
N
O
H
O
S
NH
2
CH
3
Protease
N
NH
H
O
H
949
H
952

N
O
H
Lys
H
H
949
or
SH
952
H
2
N
Protease
within the following sequence, which is the same as
that in
2
-macroglobulin:
Here the side chains have been added for the thioester-
forming cysteine and glutamate and the sequence
numbers are for C3. The thioester-forming glutamate
is labeled E* because it is not encoded as glutamate
but as glutamine, suggesting a mechanism of thio-
ester formation. Protein C4 exists as two subforms,
C4A and C4B. Both C3 and C4A react predominately
with lysine amino groups, but C4B reacts with OH
groups of cell surface polysaccharides.
h
It has a
histidine at position 1106. There is good evidence
that it is adjacent to the C = O group of the thioester
and reacts to form an acyl-imidazole which is more
reactive with hydroxyl groups than is a thioester:
Activation of C3 and C4 apparently allows the
preformed thioester, which is buried in the interior
R
SH
N
NH
O
O
R
H
S
N
NH
O

+
1106
N
NH
O
S
O
G C G E E* N(T) M
H
2
C (CH
2
)
2
S C
O
1010 1015
E*, encoded as Q
631
BOX 12-D (continued)
of these proteins, to become exposed on the external
surface where it can react. The high group transfer
potential of the thioester ensures that the reaction
will go to completion.
a
Sottrup-Jensen, L. (1989) J. Biol. Chem. 264, 11539 11542
b
Fothergill, J. (1982) Nature (London) 298, 705 706
c
Jacobsen, L., and Sottrup-Jensen, L. (1993) Biochemistry 32,
120 126
d
Andersen, G. R., Koch, T. J., Dolmer, K., Sottrup-Jensen, L., and
Nyborg, J. (1995) J. Biol. Chem. 270, 25133 25141
e
Boisset, N., Taveau, J.-C., Pochon, F., and Lamy, J. (1996) J. Biol.
Chem. 271, 25762 25769
f
Qazi, U., Gettins, P. G. W., Strickland, D. K., and Stoops, J. K.
(1999) J. Biol. Chem. 274, 8137 8142
g
Gettins, P. G. W. (1995) Biochemistry 34, 12233 12240
h
Law, S. K. A., and Dodds, A. W. (1997) Protein Sci. 6, 263 274
i
Khan, S. A., Sekulski, J. M., and Erickson, B. W. (1986) Biochem-
istry 25, 5165 5171
j
Dodds, A. W., Ren, X.-D., Willis, A. C., and Law, S. K. A. (1996)
Nature (London) 379, 177 179
this sequence, as well as another nearby methionine
residue, is very susceptible to oxidation to sulfoxides:
The oxidation, which may be caused by such agents
as myeloperoxidase (Chapter 16) released from leuko-
cytes,
507
inactivates the inhibitor. This may be physio-
logically important in permitting the proteases to be
uninhibited in the immediate vicinity of the leukocyte.
Cigarette smoke also inactivates
1
-protease inhibitor
by oxidation of the same methionine residues and the
lungs of smokers contain the oxidized inhibitor.
508
However, the major cause of emphysema among
smokers appears to be an increase in released neutro-
phil elastase.
509
One approach to the treatment of
emphysema involves weekly intravenous injection of

1
-antitrypsin.
510
This treatment may be improved
by use of genetically engineered oxidation-resistant
variants of the antitrypsin such as Met 385Val.
504,511
Efforts are also being made to introduce an
1
-anti-
trypsin gene into lung epithelial cells.
510,512
Blood plasma also contains at least nine other pro-
tease inhibitors. One of these, the thrombin inhibitor
antithrombin III (Section 9), contains the sequence
Arg-Ser-Leu at the P
1
, P
1
', and P
2
' sites. A tragic case
of a person born with a Met 385 Arg mutation in

1
-antitrypsin has been reported.
513
This converted
the antitrypsin to an antithrombin causing a bleeding
disorder that was eventually fatal.
9. Coagulation of Blood
The clotting of blood following injury and the subse-
quent dissolving of the clot are familiar phenomena
that involve several cascades of proteolytic enzymes
together with a number of accessory cofactors.
514 516
The first step in clotting results from activation of
blood platelets which aggregate to form a platelet plug
that slows bleeding.
514,517
The clot, which is formed by
the insoluble fibrin, grows on the platelet surfaces and
strengthens the plug. The initial rapid formation of a
clot occurs via the tissue factor pathway (or extrinsic
pathway; right side of Fig. 12-17) which is triggered by
the exposure in injured tissues of tissue factor (TF),
a transmembrane glycoprotein
518 522
and a member of
the cytokine receptor superfamily.
518
Human TF is a
263-residue protein with a single membrane-spanning
region and a small 20-residue C-terminal cytoplasmic
domain. The 219-residue extracellular domain consists
largely of two IgG-like domains (Fig. 12-18).
519,523
This
protein stimulates the conversion of fibrinogen
524 526
into the insoluble fibrin through the action of three
proteasesfactor VIIa, factor Xa, and thrombin. These
enzymes are generated from proenzymes VII, X, and
prothrombin, respectively in a cascade mechanism.
Factor VII binds tightly to TF,
527,527a,b
which also
binds Ca
2+
and phospholipid of the cell membranes.
Within this complex a plasma protease, such as throm-
bin or factor VIIa or Xa, cleaves a single Arg-Ile bond
in VII to form active VIIa.
528 530
The TFVIIa complex
is a very active protease which cleaves a specific pep-
tide bond in factor X to form Xa
531 533
which continues
the cascade. Notice that there are autocatalytic features:
VII can be converted to the active VIIa by the action of
Xa and the accessory factor Va is generated from the
precursor, factor V, in part through the action of throm-
bin.
514,534 535
Factors Xa and Va together with Ca
2+
and
phospholipid form the active prothrombinase complex
which attacks prothrombin to form the active enzyme
thrombin.
536,537
The roles of factor Va and Ca
2+
appear
to be to hold the prothrombin and the activated pro-
tease Xa together on the phospholipid surface.
538
This
localizes the clotting. Factor Xa is unusually specific,
cleaving only after arginine in the sequence. Its activa-
tion of prothrombin results from cleavage of two
OOC S
H NH
3
O
CH
3
O
Methionine sulfoxide
+

C. Displacement Reactions on Carbonyl Groups


632 Chapter 12. Transferring Groups by Displacement Reactions
Figure 12-17 Major components of
the human blood clotting cascades.
The site of action of the leech anti-
coagulant protein hirudin is also
indicated.
peptide bonds, which releases the 39-kDa thrombin
from the much larger, immobilized 72-kDa proen-
zyme.
236,531
Thrombin, like most other clotting factors, is also a
serine protease. However, the clotting factors are multi-
domain proteins that are more elaborate and more
specific
539
than the digestive enzymes. Prothrombin,
as well as factors VII, IX, and X and the anticoagulant
protein C, contain at their N-terminal ends several
residues of -carboxyglutamate (Gla), an amino acid
generated in a posttranslational modification that
depends upon vitamin K (Chapter 15). In human
prothrombin there are ten of these in the following
N-terminal sequence, where E* represents Gla. Since
many of these enzymes are also dependent upon
activation by calcium ions (see Fig. 12-17), it is thought
that the function of Gla is to assist in the binding of
+
+
+
High MW
kininogen (150 kDa)
Surface
contact
XII (Hageman factor,
80 kDa)
XIIa
Thrombin
XI proenzyme (160 kDa)
surface
IX (Christmas factor,
proenzyme, 57 kDa)
XIa
IXa VIIa
VIIIa
Thrombin
VIII (Antihemophilic
factor, 330 kDa)
Phospholipid
X (Stuart factor, 56 kDa)
II (Prothrombin, 72 kDa)
I (Fibrinogen, 340 kDa)
XIII XIIIa (Transamidase)
(Fibrin stabilizing
factor, 320 kDa)
Firm clot
Hirudin
Fibrin (loose clot)
Fibrinopeptides
Prothrombinase complex
Va (170 kDa) V (330 kDa)
Thrombin (39 kDa)
TF (Tissue Factor, 30 kDa),
a transmembrane
glycoprotein
VII (45.5 kDa)
TFVII complex
plasma
proteases,
e.g. Xa or
thrombin
TFVIIa
TF
The tissue factor pathway The intrinsic pathway
+
+ +
Ca
2+
+
+ +
Xa
H
2
O
Ca
2+
+
As complex
with vWF
VIIIi (inactive)
Kallikrein (88 kDa) Prekallikrein

Ca
2+
which helps to tie these proteins to the phospho-
lipids of platelet surfaces. In factors VII, IX, X, and
protein C this Ca
2+
-binding domain is followed by two
epidermal growth factor (EGF)-like domains, each
containing one residue of erythro--hydroxyaspartate
or hydroxyasparagine formed by hydroxylation of an
aspartate or asparagine residue in the first EGF-like
domain.
540,540a,b
The C-terminal catalytic domain of
each enzyme contains the protease active site.
Fibrinogen is an elongated molecule with an ()
2
structure.
524,541,541a
Thrombin cleaves specific Arg-Gly
bonds in the and chains releasing short (14- to16-
residue) fibrinopeptides from the N termini of the
peptide chains. This leaves Gly-Pro-Arg knobs at
ANTFLE*E*VRKGNLE*RE*CVE*E*TCSYE*E*AAFE*ALE*SS
10 20 30
633
Figure 12-18 A ribbon drawing of the three-dimensional
structure of the extracellular region (residues 3 to 213) of
human tissue factor. Strands of domain 1 and 2 are labeled
A to G. Two helices are labeled
1
, and
2
and the three
asparagines that provide the glycosylation sites are also
marked. MolScript drawing from Harlos et al.
520
C. Displacement Reactions on Carbonyl Groups
the N termini of the -chains and these fit into com-
plementary holes in the chains to form nonco-
valently linked aggregates.
542 544
The clot that forms
is unstable, but it is soon crosslinked by the action of
the transamidase (transglutaminase or factor XIIIa;
Eq. 2-23).
545,546
The fibrin monomers, von Willebrand
factor (discussed in following paragraphs), fibronectin,
collagen, and other proteins all become crosslinked.
The slower intrinsic mechanism first described
in 1964
547,548
consists of a cascade involving six proteases
(Fig. 12-17, left side). Again, autocatalytic cycles are
present in the activation by XIIa of both prekallikrein
549
and XII and in activation by thrombin of factors XI and
VIII. This intrinsic pathway is initiated by the serine
protease proenzymes prekallikrein and factor XII
together with the accessory protein high-molecular-
mass-H- kininigen.
549
Activation occurs when blood
contacts surfaces such as glass or kaolin (a clay).
547,548,550
Factor XI can also be activated by thrombin. Heredity
absence of factor XI leads to bleeding problems, especially
after surgery, but absence of XII does not. This fact
suggests that direct activation of XI by thrombin is
important and that the kallikreinfactor XII pathway
is usually less important.
Factor IX (Christmas factor) is next in the intrinsic
mechanism cascade. It can be activated either by XIa
or by VIIa of the tissue factor pathway. The absence of
a functional factor IX leads to the inherited X-linked
bleeding disorder hemophilia B which affects 1 in
30,000 males. The condition can be mild or very seri-
ous
551,552
and may be caused by a variety of mutations
or by incorrect splicing of the messenger RNA for the
416-residue factor IX. The level of factor IX in blood
increases with age, almost doubling by old age.
552a
Factor IXa causes a rapid activation of factor X
only if Ca
2+
, phospholipid,
553,554
and the accessory
factor VIIIa
555
are present. The IXaVIIIa complex acts
on X about 2 x 10
5
times faster than does IXa alone.
This complex cleaves the same bonds in X as does the
VIIaVa complex formed in the tissue factor pathway.
514
The 2332-residue factor VIII and factor V have similar
structures that include three repeats of a domain homo-
logous to the blue copper-containing plasma protein
ceruloplasmin (Chapter 16).
556 559
Tyrosine 1680 of
VIII apparently must be converted to a sulfate ester for
full activity.
560
The absence of factor VIII in about 1/ 10,000 males
born, leads to the severe X-linked bleeding disorder
hemophilia A. Human factor VIII is encoded by a
186-kb gene containing 26 coding exons. Severe cases
of hemophilia are usually a result of point mutations
that produce stop codons in this gene and therefore a
shortened protein. Milder cases may result from an
amino acid substitution.
561
Factor VIII circulates in the
plasma as a complex with the von Willebrand factor,
(vWF), a large multimeric protein derived by proteo-
lytic cleavage, glycosylation, and sulfation of a 2813-
residue precursor.
562 566
The mature 2050-residue vWF
stabilizes factor VIII in the blood. Another important
function of vWF is to bind platelets to damaged endo-
thelial surfaces.
567,568
Like fibrinogen, vWF contains
RGD sequences specific for binding to adhesion recep-
tors.
569,570
It also binds to collagen. A serious inherited
bleeding disorder caused by deficiency of vWF was
first discovered among inhabitants of islands in the Gulf
of Bothnia, Finland, in 1926 by von Willebrand.
563,571
If mild forms of the disease are included, vWD defi-
ciency is the commonest bleeding problem. However,
abnormalities have been identified in almost every one
of the proteins in the coagulation cascades.
514
Until recently, these bleeding diseases were treated
by regular injections of the appropriate factors isolated
from human plasma. This was both costly and carried
a high risk of infection by HIV, hepatitis, or other
contaminating viruses. Now, cloned genes are used
for commercial production of the factors.
572,573
Experi-
ments in animals, designed to lead to eventual gene
634 Chapter 12. Transferring Groups by Displacement Reactions
replacement therapy, have been somewhat success-
ful.
573,574
It is of interest to compare the relative con-
centrations of several of the clotting factors in plasma
and to compare these with their positions in the cas-
cade of the tissue factor (extrinsic) pathway.
515
mg / L Mass kDa M
VII 0.5 45.5 0.01
V 10.0 330 0.03
Prothrombin 150 72 2.1
Fibrinogen 3,000 340 8.8
Why do we have the intrinsic pathway when the
tissue factor pathway provides rapid clot formation?
The answer seems to be that the tissue factor pathway
is needed immediately after injury but that it is turned
off quickly by the anticoagulation systems of the
body. As a result the protease plasmin begins to dis-
solve (lyse) the clot within a few hours. The intrinsic
pathway is apparently needed to maintain the clot for
a longer period.
514
What prevents the clotting mechanism, with its
autocatalytic cycles, from running out of control? In
part the answer lies in the localization of the enzymatic
activation to tissue surfaces near a wound. The flow
of blood rapidly dilutes components that escape into
the general circulation and liver cells take up and
destroy the active proteases. A variety of circulating
antiproteases including the tissue factor pathway
inhibitor
575
act on these escaped enzymes. Two anti-
coagulation systems that are localized on vessel walls
576
also come into action very quickly. The circulating
polysaccharide heparin (Chapter 4) forms a complex
with the serpin antithrombin. Antithrombin traps
thrombin as an inactive complex or compound
577,578a
and heparin greatly accelerates the inactivation.
578
Kallikrein and factors IXa, Xa, XIa, and XIIa are also
inhibited.
Thrombin is an allosteric protein which exists as
a mixture, in nearly equal amounts, of a fast-acting
form that cleaves fibrinogen and is stabilized by Na
+
,
and a slow-acting form that initiates the second anti-
coagulant cascade.
579
The slow acting form, bound to
thrombomodulin,
527a,540b,580,580a,b,581
an endothelial
cell surface protein, attacks the proenzyme of another
serum protease called protein C.
582,582a
Activated
protein C (APC) inactivates the accessory clotting
factors Va and VIIIa. The accessory factor protein S
583
is also needed for rapid inactivation. A blood clot is
temporary and its dissolution begins as soon as it is
formed, largely through the action of plasmin, a
protease derived from the circulating 791-residue
plasminogen
584 588
through the action of yet other
proteases. Plasminogen often becomes crosslinked
to cell surface proteins by transglutaminase.
586
Plasminogen activators include urokinase,
589
a
protease present in kidney tissue and urine, and
tissue plasminogen activator (tPA),
590,591
a
527-residue protease which is now produced by
recombinant DNA technology. It is sometimes used to
dissolve blood clots in emergency situations, such as
myocardial infarction and pulmonary embolism,
592
but can also cause serious bleeding problems. Plasmin
is inhibited by the plasma
2
antiplasmin and is
inactivated by action of clotting factor XIIa. Likewise,
tPA is inhibited by several protease inhibitors present
in tissues and in plasma. One is a fast-acting serpin
called plasminogen activator inhibitor.
593
Another
anticoagulant compound of medical interest is hirudin,
a 65-residue peptide from the leech. It binds very tightly
to thrombin (K
d
= 1 pM) preventing its action.
594 596
Insects also produce antithrombin.
597,598
Ticks
599,600
and some insects
598
inject proteins that inhibit Xa
selectively.
599
Anticoagulants are of great medical
importance and much effort is being devoted to the
design of better inhibitors of thrombin,
601603
factor Xa,
604
and other components of the blood coagulation cascade.
Inherited deficiencies of the anticoagulant path-
ways with associated problems of thrombosis are
known. These include problems with protein C,
576,605
plasminogen,
606
and antithrombin.
607,608
10. Esterases and Lipases
A group of esterases hydrolyze simple oxygen esters.
Some of these are designed to hydrolyze a particular
ester or small group of esters, while others have a
more nonspecific action. Acetylcholinesterase
609 611a
is specific for acetylcholine (Eq. 12-25), a neurotrans-
mitter that is released at many nerve synapses and
neuromuscular junctions (Chapter 30). The acetylcho-
line, which is very toxic in excess, must be destroyed
rapidly to prepare the synapse for transmission of
another impulse:
The more widely distributed butyrylcholinesterase
612
is less specific but prefers butyrylcholine. Acetylcholinest-
erase is a very efficient catalyst:
613 615
k
cat
= 1.6 x 10
4
s
1
(12-25)
H
3
C C
O
O
CH
2
CH
2
N CH
3
CH
3
CH
3
+
Acetylcholine
H
3
C COO

HO CH
2
CH
2
N CH
3
CH
3
CH
3
+
H
2
O
Acetylcholine esterase
Acetylcholine
635
and k
cat
/K
m
= 2 x 10
8
M
1
s
1
at 25C. It exists as a series
of molecular forms containing varying numbers of
68-kDa catalytic subunits, 100-kDa structural subunits
and subunits with triple-helical ~120-kDa collagen-
like tails.
609,616
The subunits are joined by disulfide
bridges to give aggregates that range from simple
dimers of catalytic subunits to tailed forms containing
812 catalytic subunits and non-collagenous struc-
tural subunits as well. The tailed forms are secreted
from cells and may be designed to take up residence
in the basal lamina of synapses, whereas the dimers
are apparently attached to phosphatidylinositol an-
chors in the membranes (Fig. 8-13).
617
Human liver
carboxylesterases are relatively nonspecific enzymes
that hydrolyze ester groups of various drugs and toxins
including cocaine and heroin. Products are often ex-
creted in the urine.
618
Thioesterases function in
biosynthesis of fatty acids, polyketides (Chapter 21),
and many other substances.
619
Lipases
620,621
hydrolyze triacylglycerols. The pan-
creatic digestive lipase
622,623
acts faster on emulsified
fats than on glycerol esters in true solution but re-
quires the cooperation of a small 10-kDa colipase.
624
Cholesterol esterase hydrolyzes not only cholesterol
esters but also esters of fat-soluble vitamins, phospho-
lipids, and triacylglycerols.
625,626
Other lipases include
gastric
627
and hepatic lipases and a lysosomal acid
lipase
628
which also attacks neutral lipids. Plasma
lipoprotein lipase
629,630
digests fats in the chylomicrons
and from the very low-density lipoproteins of blood.
Hormone-sensitive lipase hydrolyzes stored triacyl-
glycerols in the cytosol in response to catecholamines,
ACTH, and other hormones.
631
The phospholipases
attack phospholipids, while cutinase,
632
produced by some fungi, cleaves the
ester linkages in the cutin (Chapter 21)
of plant surfaces. Fungal lipases are
important industrial commodities.
Numerous structural and mechanistic
studies have been made with
them.
620,629,633 635
The gene for a lipase
from Candida rugosa has been synthe-
sized using codons that maximize its
expression in Saccharomyces cerevisiae
and which allow for further genetic
engineering of the lipase.
636
All of these esterases appear to act
by mechanisms closely related to those
of proteases. Acetylcholinesterase
contains an active site serine that reacts
with organophosphorus compounds
(Box 12-E) and is part of an Asp-His-
Ser catalytic triad which lies in a deep
gorge as well as an oxyanion hole.
637
A surprise is the absence of an essential
carboxylate group that might bind the
positively charged trimethylammonium
group of acetylcholine. Instead, the lining of the
gourge is rich in aromatic side chains which may
interact with the methyl groups of the substrate and
by their polarizability stabilize the charge.
611,638
Most
lipases, including cutinase, also have an Asp-His-Ser
or Glu-His-Ser triad as well as some form of oxyanion
hole.
620,632,639
Like the serine proteases, the lipases
have bell-shaped pH optima.
Phospholipase A
2
cleaves the ester linkage at the
2 position in phospholipids.
640 642
One isoenzyme
form is secreted by the pancreas as a proenzyme
whose N-terminal seven residues are removed by
trypsin to give an active 125-residue enzyme. Phos-
pholipase A
2
of a similar type is abundant in venoms
of reptiles and bees. The venom and pancreatic en-
zymes have closely similar three-dimensional struc-
tures.
643 645
Although the folding pattern is different
from that of chymotrypsin, imidazole (His 48) and
carboxylate (Asp 99) groups are present in the active
site in an orientation resembling that of the catalytic
triad of serine protease. The enzyme requires calcium
ions, one of which binds at an appropriate point for
complexation with the substrate carbonyl as it is con-
verted to an oxyanion intermediate. The backbone
NH of Gly 30 may also serve as an oxyanion ligand.
However, in most phospholipases there is no active
site serine. Instead, a water molecule is positioned to
serve as the attacking nucleophile in formation of the
oxyanion as is indicated in the following scheme,
which shows a truncated phosphatidylethanolamine
as the substrate.
643
Phospholipase A
2
is up to 1000
or more times as active on phospholipids in micelles
as on dissolved substrates.
621,646,647
Apparently, the
D49
O
O
Ca
2+
O
O
H
N
C
O
O
H
N
N
H
O
H
O
P
O
O
O
Ethanolamine
D99
O
O
Y52
O
H
G30
H48
1
Y73
O
H
2
3

Ethanolamine
O
O
O
O
O
O
O
O
H P

C. Displacement Reactions on Carbonyl Groups


636 Chapter 12. Transferring Groups by Displacement Reactions
BOX 12-E INSECTICIDES
Over 200 organic insecticides, designed to kill
insects without excessive danger to humans and
animals, are presently in use.
a e
Many of these
compounds act by inhibiting cell respiration; others
uncouple ATP synthesis from electron transport.
The chlorinated hydrocarbons such as DDT act on
nerves in a manner that is still not fully understood.
One of the largest classes of insecticides acts on the
enzyme acetylcholinesterase of nerve synapses.
Like chymotrypsin, it contains an active site serine
residue that reacts with organophosphorus com-
pounds. The extreme toxicity of esters of pyrophos-
phate and of dialkylphosphonofluoridates was
recognized in the 1930s and led to their develop-
ment in Germany and in England as insecticides
and as nerve gases. Among the most notorious is
diisopropylphosphonofluoridate (diisopropylfluo-
rophosphate; DFP), for which the LD
50
(dose lethal
to 50% of the animals tested) is only 0.5 mg kg
1
intravenously. This exceedingly dangerous com-
pound can cause rapid death by absorption through
the skin.
The following structures are a few of the organ-
ophosphorus compounds and other acetylcholinest-
erase inhibitors that are selectively toxic to insects.
The characteristic high group transfer potential of a
phospho group in pyrophosphate linkage, which
makes ATP so useful in cells, also permits tetraethyl
pyrophosphate to phosphorylate active sites of acetyl-
cholinesterases. While TEPP is very toxic, it is rapidly
hydrolyzed; all harmful residues are gone within a
few hours after use.
Two insecticides that have been used widely are
parathion and malathion. They are less toxic than
DFP or TEPP and do not become effective insecticides
until they undergo bioactivation during which
conversion from a P = S to a P = O compound occurs:
The desulfuration reaction involves microsomal
oxidases of the liver, the sulfur being oxidized
ultimately to sulfate.
f
The reactivity of parathion
with cholinesterases depends upon the high group
transfer potential imparted by the presence of the
excellent leaving group, the p-nitrophenolate anion.
If the P O linkage to this group is hydrolyzed before
the desulfuration takes place, the phosphorus com-
pound is rendered harmless. Thus, the design of an
effective insecticide involves finding a compound
which insects activate rapidly but which is quickly
degraded by higher animals. Other factors, such as
rate of penetration of the insect cuticle and rate of
excretion from the organism, are also important.
The phosphorylated esterases formed by the
action of organophosphorus inhibitors are very
stable, but some antidotes can reverse the inhibition.
The oxime of 2-formyl-1-methylpyridinium ion
(pralidoxime) is very effective.
g
Its positive charge
permits it to bind to the site normally occupied by
the quarternary nitrogen of acetylcholine and to
displace the dialkylphospho group:
Carbaryl, a widely used methyl carbamate, is a
pseudosubstrate of acetylcholinesterase that reacts
10
5
to 10
6
times more slowly than do normal sub-
strates. The carbamoylated enzyme formed is not
as stable as the phosphorylated enzymes and the
inhibition is reversible.
A basic problem is that most insecticides are
designed to attack the central nervous system of the
insect, the system that depends heavily upon acetyl-
choline. However, in human beings the readily
accessible peripheral nervous system also depends
upon acetylcholine, e.g., in neuronuscular junctions.
The danger of poisoning is great. Another approach
is to attack the glutamate-dependent peripheral
system in insects, e.g., with inhibitors of glutamate
decarboxylase. Glutamate functions as a neuro-
transmitter in the human body but only in the well-
O NO
2
P
Et O S
Et O
O
C N
CH
3
O
S
P
O S
O
H
3
C
H
3
C
C O Et
C O Et
O
O
H
P O P
O O
O Et Et O
Et O O Et
Parathion
Carbaryl
H
Malathion
Tetraethyl pyrophosphate
(TEPP)
N
CH
3
C N O P OR
O
Seryl (enzyme)
OR O
H

+
Pralidoxime
R O
P
X
S
O R
R O
P
X
O
O R
Highly toxic
637
BOX 12-E (continued)
protected central nervous system.
Another important problem is the development
of insects resistant to insecticides. This often arises
as a result of increased levels of carboxylesterases
which hydrolyze both organophosphates and car-
baryl.
h,i
A mutation that changed a single active site
glycine to aspartate in a carboxylesterase of a blow-
fly changed the esterase to an organophosphorus
hydrolase which protected the fly against insecticides.
j
a
Bchel, K. H., ed. (1983) Chemistry of Pesticides, Wiley, New
York
b
Hassall, K. A. (1990) The Biochemistry and Uses of Pesticides, 2nd
ed., VCH Publ., Weinheim
c
Kamrin, M. A., ed. (1997) Pesticide Profiles, CRC Press, Boca
Raton, Florida
d
Casida, J. E. (1973) Ann. Rev. Biochem. 42, 259 278
e
Wilkinson, C. F., ed. (1976) Insect Biochemistry and Physiology,
Plenum, New York
f
Nakatsugawa, T., Tolman, N. M., and Dahm, P. A. (1969)
Biochem. Pharmacol. 18, 1103 1114
g
Wilson, I. B., and Ginsburg, S. (1955) Biochim. Biophys. Acta. 18,
168 170
h
Raymond, M., Callaghan, A., Fort, P., and Pasteur, N. (1991)
Nature (London) 350, 151 153
i
Karunaratne, S. H. P. P., Hemingway, J., Jayawardena, K. G. I.,
Dassanayaka, V., and Vaughan, A. (1995) J. Biol. Chem. 270,
31124 31128
j
Newcomb, R. D., Campbell, P. M., Ollis, D. L., Cheah, E.,
Russell, R. J., and Oakeshott, J. G. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 7464 7468
substrate-binding cavity of the protein is designed to
accommodate phospholipid molecules in the preferred
conformation found in the micelles.
643
Most of the
other lipases have lids which close over the active sites
and impede access of substrates. Binding to a phospho-
lipid surface apparently induces a conformational
change that opens the lid. This allows substrate to enter
from the lipid surface.
620,621,646
Cutinases, which do
not display interfacial activation, do not have lids. Phos-
pholipase A
2
activity is stimulated by an applied elec-
trical field, a result that suggests that its activity in vivo
may be regulated in part by the membrane potential.
648
11. Other Acyltransferases
Acyl groups are frequently transferred from
amides or esters to various acceptors in biosynthetic
reactions. Among the many known acyltransferases
are the ribosomal peptidyl transferases (Chapter 29),
a transacylase involved in bacterial peptidoglycan
synthesis (Chapter 20), transglutaminase (Eq. 2-23),
649,650
-glutamylcyclotransferase (Box 11-B), and transacyla-
tion reactions involving acyl-CoA derivatives. Exam-
ples of the latter are N-acetylation
651
or myristoylation
(Chapter 8) of proteins, the formation of acetylcholine
from choline
652
and of acetylcarnitine from carnitine
(Eq. 17-4), and acetylation of the antibiotic chloram-
phenicol. The high group transfer potential of thioesters
ensures that these reactions proceed to completion.
Chloramphenicol acetyltransferase (CAT),
653
the
enzyme that catalyzes acetylation and inactivation of
the antibiotic by bacteria, is much used in studies
of gene expression (Chapter 28). A catalytic histidine
removes the proton from the 3-OH group and a serine
hydroxyl provides an oxyanion hole to accommodate
the anticipated tetrahedral intermediate.
652,654
The
steroidal antibiotic fusidic acid (Chapter 22, Section G)
is a competitive inhibitor.
655
A related transferase is
aspartate carbamoyltransferase (Fig. 7-20; Chapter 24).
Penicillin and related antibiotics are inactivated by
-lactamases (Box 20-G), some of which resemble serine
proteases in forming acyl enzymes with active site
serine side chains.
656,657
Others are zinc metallogen-
zymes.
658,659
Amidohydrolases such as asparaginase
and glutaminase,
660,661
deacetylases,
662
and many
other hydrolases can also be described as acyltrans-
ferases.
D. Displacement on a Phosphorus Atom
Nucleophilic displacements on phosphorus (Table
10-1, reaction type 1C) are involved in virtually every
aspect of cellular energetics and in many aspects of
biosynthesis. One large group of such enzymes are
phosphotransferases, which transfer phospho (also
called phosphono or, traditionally in biochemistry,
phosphoryl) groups from one nucleophilic center to
another. When transfer is to water the enzymes are
called phosphatases, and when from one group in a
molecule to another in the same molecule, mutases.
O
2
N
N
Cl
H
H
OH H
CH
2
OH
O
H
Cl
Chloramphenicol, a broad-spectrum
Streptomyces antibiotic
Inactivated by acetylation here
1
3
D. Displacement on a Phosphorus Atom
638 Chapter 12. Transferring Groups by Displacement Reactions
Enzymes that transfer a phospho group from ATP to
water are ATPases and those that transfer the phos-
pho group from ATP to some other nucleophile are
kinases. Substituted phospho groups can also be
transferred. Thus, nucleases, members of a larger
class of phosphodiesterases, hydrolyze nucleic acids
by transfer of nucleotidyl groups to a hydroxyl group
of water. Polynucleotide polymerases transfer
nucleotidyl groups to growing polynucleotide chains.
An intramolecular nucleotidyl transferase is adenylate
cyclase. Topoisomerases carry out a sequence of
phosphotransferase reactions.
1. Questions about Mechanisms
Consider the following general equation for trans-
fer to nucleophile Y

of a phospho group attached in


an ester or an anhydride linkage to form ROH, which
could be an alcohol, carboxylic acid, or a phosphoric
acid such as ATP.
This equation could also represent a half-reaction in a
double-displacement process. As with displacements
on saturated carbon atoms, two basic mechanisms can
be imagined. The first is S
N
2-like or associative.
663 667
The transition state might be represented where the
bonds from the phosphorous atom to Y and to O are
approximately equally formed. In an in-line displace-
ment, where Y, P, and OR are colinear, this mecha-
nism leads to inversion if a chiral phospho group is
used. There is another possibility for the associative
mechanism. Whereas a carbon atom can form only
four stable covalent bonds, phosphorus is able to form
five. While nucleophilic attack on carbon leads to a
transient five-bonded transition state (Eq. 12-2), attack
on phosphorus could produce a relatively long-lived
pentacovalent intermediate (Eq. 12-27). Notice that
two transition states are involved. Remember that our
conventional way of drawing phosphate esters with a
double bond from phosphorus to one of the oxygens is
misleading. All of the P O bonds share some of the
double-bond character and the phosphate group has
many characteristics of a completely single-bonded
structure:
Y OR
O
H
O
O
H
Transition state
P
ROH
YPO
3
H

P O
R O
OH
HO

(12-26)
a
b
Y OR
O
H
O
O
H
Pentacovalent intermediate,
a trigonal bipyramid
P
YPO
3
H
2
P O
R O
OH
HO
Y

Y OR
O
H
O
O
H
Second transition state
P


RO

Apical
positions
Equatorial positions

(12-27)
Likewise, in the transition state structures of Eq. 12-27,
step b, the P O bonds, except for that to O R, are equiv-
alent and all have partial double-bond character.
Geometric complexities. The geometry of the
pentacovalent intermediate in Eq. 12-27 is that of a
trigonal bipyramid. In this structure the bond angles
in the equatorial plane are 120, whereas all of the
angles between any of those bonds and the two that
attach to the groups in the apical positions are 90.
This disparity arises naturally from the fact that it is
impossible to place five points on the surface of a
sphere all equidistant one from the other. The attack
of Y

from the side opposite O R (an in-line attack)


leads to a trigonal bipyramid in which O R and Y
occupy the two apical positions. However, if Y

attacks
a face opposite one of the other oxygens (adjacent
attack), O R will take an equatorial position.
The chemical reactivities of groups in the apical
and equatorial positions of pentacovalent intermediates
are different.
664
In particular, elimination of a nucleo-
philic group to form a tetrahedral phosphate is easier
from an apical position than from an equatorial position.
For the in-line displacement of Eq. 12-27 elimination of
RO

should be easy. However an adjacent attack would


leave OR in an equatorial position. Before it could be
eliminated, the intermediate would probably have to
undergo a permutational rearrangement by which
OR would be transferred from an equatorial to an
apical position.
One type of permutational rearrangement, known
as pseudorotation, can be visualized as in Eq. 12-28.
668,669
The axial groups a and b move back while equatorial
groups 2 and 3 move forward, still in the same equato-
HO
P
O O
O HO
P
O O
O

639
rial plane. Equatorial group 1 does not move. This
decreases the 120 bond angles between the equatorial
groups and increases the bond angles between group 1
and the axial groups until all four bond angles to group
1 are equivalent. The resulting square pyramid is a
high-energy transition state structure in the pseudo-
rotation process and can either revert to the original
structure or, by continued motion of the groups in the
same directions as before, to the structures shown at
the bottom of Eq. 12-28. In the final structure, groups
2 and 3 are axial and the original axial groups a and b
are equatorial. Pseudorotation is slow enough that it
could be rate limiting in enzymatic reactions. Stereo-
electronic effects could also affect these rates.
670,671
However, much evidence indicates that enzymes almost
always avoid these complexities by using in-line
mechanisms.
Metaphosphate ions. An alternative to an asso-
ciative mechanism is an S
N
1-like or dissociative
mechanism which can occur by elimination of a
metaphosphate ion (Eq. 12-29, step a). A nucleophilic
reagent can then add to the eliminated metaphosphate
in step b. The formation of metaphosphate is analogous
to formation of carbocation during the action of lysozyme
(Eq. 12-11). This dissociative mechanism could lead
either to racemization or to inversion of a chiral phos-
phate. Metaphosphate ions have been shown to exist.
They are generated in certain nonenzymatic elimination
reactions in aprotic solvents
672 674
and they are reactive
electrophiles able to react as in Eq. 12-29. However,
there is some doubt that they can exist free in aqueous
media.
675
Jencks and associates concluded from studies
of linear Gibbs energy relationships that in the transition
state for nonenzymatic phospho transfer reactions
there is a large amount of bond breaking and a small
amount of new bond formation in the transition
state
676
but no free metaphosphate. There is a large
dependence of the rate on the pK
a
of the leaving
group, but there is still a small dependence on the
pK
a
of the entering nucleophile. Thus, we have a
a
b
a b
3
P
2
1
a
1
b
3
2
P
a
1
b
3
2
P
(12-28)
a
b
O P
O
O
O
R

H
OH R
O
P
O O

Y O
H
O P Y O
O
OH

(12-29)
dissociative mechanism but without free metaphosphate.
The concept is supported by studies of kinetic isotope
effects.
677,678
Coping with negative charges. We visualize
phospho group transfers as involving attack by a
nucleophile bearing at least a partial negative charge.
However, phospho groups also carry one or two nega-
tive charges and that in ATP even more. Therefore, it
does not seem surprising that many phosphotrans-
ferases are metalloenzymes, sometimes containing
bound Zn
2+
which neutralizes some of the negative
charge. Furthermore, enzymes usually accept ATP as
a substrate only when it is accompanied by a divalent
metal ion,
679
usually Mg
2+
. Another way in which
enzymes deal with the negative charges on phospho
groups is to have arginine side chains in appropriate
positions to interact by forming strong ion paired-
hydrogen bonds. It is often assumed that it is essential
to neutralize the charge on the phospho group to avoid
electrostatic repulsion from a partial or complete
negative charge on the attacking nucleophilic center.
However, with a dissociative mechanism and a meta-
phosphate-like intermediate the transition state may
be reached without charge neutralization.
679,680
Inter-
actions of Mg
2+
with phosphorus-containing substrates,
like those of fixed positive charges in the protein may
also be essential for binding the substrate correctly to
an enzyme.
2. Magnetic Resonance Studies
There have been many investigations of phospho-
transferases by NMR and EPR methods.
681,682
One
approach is to use paramagnetic ions such as Mn
2+
,
Cu
2+
, or Cr
3+
to induce nuclear relaxation in substrate
and coenzyme molecules at active sites of enzymes.
Flavin radicals and specifically introduced nitroxide
spin labels can serve as well. Paramagnetic ions greatly
increase the rate of magnetic relaxation of nearby
nuclei (Chapter 3, Section I). Thus, small amounts of
D. Displacement on a Phosphorus Atom
640 Chapter 12. Transferring Groups by Displacement Reactions
(12-33)
(12-32)
Mn
2+
in a sample lead to broadening of lines in ordinary
1
H,
13
C, and
31
P NMR spectra.
Useful information about enzymes can sometimes
be obtained by observing effects of paramagnetic ions
on the NMR signal of protons in the solvent water.
The relaxation time of solvent protons is usually greater
than 1 s. However, in the ion Mn(H
2
O)
6
2+
the protons
of the coordinated water molecules relax much more
rapidly, both T
1
and T
2
being ~ 10
1
s. Since the coordi-
nated water molecules usually exchange rapidly with
the bulk solvent, a small number of manganese ions
can cause a significant increase in relaxation rate for all
of the water protons. Broadening of the proton band is
observed and differences in T
1
and T
2
can be measured
by appropriate methods. Paramagnetic relaxation
effects usually increase as the inverse sixth power of
the internuclear distance. Knowing the Mn
2+
H
distance to be 0.287 0.005 nm for hydrated Mn
2+
, it
has been possible to relate the effects on T
1
and T
2
to
the number of H
2
O molecules coordinated at any one
time to a protein-bound metal ion and to their rate of
exchange with the bulk solvent.
Relaxation effects on
1
H,
13
C, and
31
P, while more
difficult to observe, can provide geometric information
about active sites. The theory is complex, but under
some conditions the paramagnetically induced relax-
ation can be described adequately by Eq. 12-30.
Here r is the internuclear distance, C is a combina-
tion of physical constants, and T
1M
is the longitudinal
relaxation time. The complex function f(
c
) depends
upon the correlation time
c
, the resonance frequency
of the nucleus being observed, and the frequency of
precession of the electron spins at the paramagnetic
centers. The value of
c
can be estimated (Chapter 3)
and, in turn, the distance r according to Eq. 12-30.
Such studies on creatine kinase (Eq. 12-31) utilized
both a bound Mn
2+
ion and a nitroxide spin label to
estimate distances of various protons from the nitrox-
ide.
683
Together with EPR measurements (Box 8-C),
which gave the Mn
2+
nitroxide distance, a model of the
ATPMn
2+
complex in the active site was constructed.
Additional EPR experiments on Mn
2+
complexes with
ATP and ADP containing
17
O in the , , or phospho
groups showed that in the enzymeATP creatine
complex the metal ion is bound to all three phospho
groups of ATP. It remained coordinated with the two
phospho groups of ADP and also that of the phospho-
creatine product in the enzymeADPcreatine-P com-
plex as well as in the transition state, which is pictured
occurring via a metaphosphate ion.
684
Phosphorus-31 NMR has been used to measure
internal equilibrium constants within enzyme-
substrate (ES) complexes.
663,685 687
By having both
substrate and product concentrations high enough to
saturate the enzyme, all of the enzyme exists as ES and
enzymeproduct (EP) complexes in equilibrium with
each other. For a phosphotransferase at least one sub-
strate and one product contain phosphorus. Although
the NMR resonances are broadened by binding to the
large, slowly tumbling protein, their areas can be mea-
sured satisfactorily and can be used to calculate an
equilibrium constant such as that for Eq. 12-32:
An example is illustrated in Fig. 12-20. In this
experiment
685
the relative areas of the
31
P signals of
ADP (one for free ADP and one, slightly more intense,
for MgADP) and of the signal for phosphoenolpyruvate
(PEP) were measured in the absence of enzyme and in
the presence of a catalytic amount of pyruvate kinase
(Fig. 12-20A). The results verified that the equilibrium
constant for the overall reaction (Eq. 12-33) is very
high (3300).
Phosphoenolpyruvate (PEP) + ADP ATP + pyruvate
However, with an excess of enzyme (Fig. 12-20B)
the internal constant was estimated as
(12-30)
(12-31)
r = C [ T
1M
f (
c
) ]
1/6
Figure 12-19 Proposed transition state structure formed
from Mn
2+,
ATP, and creatine bound in the active site of
muscle creatine kinase. Based on EPR spectroscopy with
regiospecifically
17
O-labeled substrates. The electrical charg-
es have been added in one possible constellation. However,
hydrogen atoms bound to phospho groups are not shown.
After Leyh et al.
684
C
C
N
P
Mn
2+
Adenosine
Creatine
P
P
N
C
N
C

Oxygen
Hydrogen
Creatine-P + ADP
k
f
creatine + ATP
k
r
k
f
k
r
ES EP
K = [EP]/[ES]
641
This is a difficult measurement
and reinvestigation by another group
686
indicated that the amount of the
PEP-containing complex had been
overestimated and that K=10.
If the rates of the forward and
backward reactions in Eq. 12-34 are
of the same order as the spin-lattice
relaxation times (T
1
) of the
31
P in the
bound substrate and product, the
rate constants k
f
and k
r
can be evalu-
ated by saturation-transfer NMR.
688
This is done by irradiating one
resonance, e.g., that of the -P of
ATP, and observing whether this
causes a loss of intensity of the
resonance for the product which is
receiving its phospho group from
ATP. This technique was used to
observe the creatine kinase reaction
(Eq. 12-31) in living muscle in both
relaxed and contracting states. For
resting muscle the observed forward
flux was 1.7 x 10
3
M s
1
and the
backward flux 1.2 x 10
3
M s
1
.
Thus, this reaction, which supplies
ATP for contraction of the muscle
from stored phosphocreatine,
appears to be operating at or near
equilibrium. This had been assumed
but had previously been difficult to
prove. Two-dimensional NMR tech-
niques can now be used for this kind
of measurement.
689
When
31
P is bonded to
18
O the
chemical shift of the
31
P is altered
by 0.0206 ppm from that when the
phosphorus is bonded to
16
O. This
allows
18
O labels introduced into
phospho groups to serve as tracers
which can be followed continuously
during reactions.
683
The technique is
useful in studies of stereochemistry
(see Section 2) and for examination
of positional isotope exchange.
690
This latter technique is often used
with ATP containing
18
O in the
,-bridge position. If an enzyme
transfers the terminal () phospho
group to an acceptor via a phosphoenzyme but without
loss of the ADP, we may expect positional isomerization.
The
18
O will move between the ,-bridge position and
8 4 0 4 8 12 16 20 24
A
B
C
D
E
PEP
-P (ADP)
-P (ADP)
Initial (no E)
-P (ATP)
-P (ADP)
-P (ATP)
Equilibrium mixture
(Catalytic E)
Equilibrium mixture
(Catalytic E)
[Pyruvate] / [ATP]15
Equilibrium mixture
Excess enzyme
+ EDTA
Chemical shift (ppm)
PEP(?)
P
i
PEP + P
i
(+ AMP?)
-P (ATP) +
-P (ADP)
-Ps
-P (ATP)
-Ps
Figure 12-20 Equilibria in pyruvate kinase reaction as studied by
31
P NMR
at 40.3 MHz, pH 8.0, 15C. (A C) Equilibria with low enzyme in levels ~ 15%
2
H
2
O. (A)
31
P NMR spectrum of 1.5 ml of reaction mixture; PEP, 13.3 mM;
ADP, 14.1 mM; MgCl
2
, 20 mM; potassium Hepes buffer, 100 mM; KC, 50 mM
without enzyme. (B) Equilibrium mixture after the addition of ~ 1 mg of
pyruvate kinase to the reaction mixture. (C) Equilibrium after the addition
of potassium pyruvate (final concentration of 200 mM) to the sample of the
spectrum in (B). (D,E) Equilibrium with enzyme concentrations in excess of
the substrates. Sample volumes ~ 1.1 ml with 10%
2
H
2
O. (D) Equilibrium
mixture set up with enzyme (2.8 mM active sites); 2.8 mM PEP; 2.4 mM ADP;
5.7 mM MgCl
2
; 100 mM potassium Hepes; 100 mM KCl. (E) Spectrum after
the addition of 50 l of 400 mM EDTA (pH readjusted to 8.0) to the sample of
spectrum D. The EDTA removes metal ions, stopping the catalytic reactions
and sharpening the resonances. From Nageswara Rao et al.
685
(12-34)
D. Displacement on a Phosphorus Atom
a nonbonding position as the phospho group is repeat-
edly transferred back and forth between ATP and the
acceptor and as the phospho group rotates.
682,690
= 0.5 1.0
[E MgATP pyruvate]
K =
[E MgADP PEP]
642 Chapter 12. Transferring Groups by Displacement Reactions
Equation 12-35 shows one part of this isomerization.
The negative oxygens have been omitted here to avoid
implying a known state of protonation or a localization
of charge.
Figure 12-21 illustrates the use of the technique in
investigating the possible participation of metaphos-
phate in a nonenzymatic reaction.
3. Stereochemistry
Evidence for an in-line S
N
2-like mechanism of most
enzymatic phospho group transfer reactions comes
largely from study of chiral phospho groups.
663,692 695
A chiral phosphate can be introduced at either the
or phosphorus of ATP by substitution of one of the
oxygen atoms by sulfur. A chiral phospho group in
the position can be formed by substituting one oxygen
by S and a second by
18
O.
Notice that the negative charge is largely localized on
sulfur in these phosphorothioate compounds.
696
More
general is the use of
17
O and
18
O to form a chiral phos-
pho group:
O
O
O
O
a
ATP
2
Enzyme-product complex
P P O P O Adenosine
O
O
O
O
O
18

Acceptor
P P O AMP

18
Acceptor

O
O O
P O P O AMP
18
Acceptor
+
O
O
O
O
O

(12-35)
Figure 12-21 The
31
P NMR spectrum at 101.2 MHz of P

of
isotopically labeled ADP. This was recovered from an exper-
iment in which ADP containing 87 atom % of
18
O in all four
oxygens around P

was allowed to undergo partial (~ 20%)


nonenzymatic hydrolysis to AMP and P
i
. Peaks 1 represent
the species containing no
18
O bonded to P

. Peaks 2 repre-
sent all species with
18
O in the P

OP

bridge, and peaks 3


represent species with
18
O in nonbridging positions at
P

.
These have undergone positional isotope exchange. From
Lowe and Tuck.
691
13.5 13.6 13.7 13.8
ppm relative to trimethyl phosphate
1
3
2
1
3
2
O
3
P O
P O
P
O Adenosine
S
O
O
O

ATP S (R configuration at P)

O
3
P O
P O
P
O Adenosine
O
S
O
O
ATP S (S configuration at P)
2

Considerable ingenuity was required in both the


synthesis of these chiral compounds
695,697
and the
stereochemical analysis of the products formed from
them by enzymes.
698 700
In one experiment the phospho
group was transferred from chiral phenyl phosphate
to a diol acceptor using E. coli alkaline phosphatase as
a catalyst (Eq. 12-36). In this reaction transfer of the
phospho group occurred without inversion. The chirality
of the product was determined as follows. It was
cyclized by a nonenzymatic in-line displacement to
give equimolar ratios of three isomeric cyclic diesters.
These were methylated with diazomethane to a mixture
of three pairs of diastereoisomers triesters. These dia-
stereoisomers were separated and the chirality was
determined by a sophisticated mass spectrometric
analysis.
692
A simpler analysis employs
31
P NMR
spectroscopy and is illustrated in Fig. 12-22. Since
alkaline phosphatase is relatively nonspecific, most
phosphate esters produced by the action of phospho-
transferases can have their phospho groups transferred
without inversion to 1,2-propanediol and the chirality
can be determined by this method.
O P
R
O
O
O
16
18
17
16
An ester chiral at the phosphorus atom; R configuration
643
Although inversion was not observed with the
E. coli alkaline phosphatase, it has been observed for
ribonucleases and many other hydrolytic enzymes and
for most kinases transferring phospho groups from
ATP. The difference lies in the existence of a phospho-
enzyme intermediate in the action of alkaline phospha-
tase (see Eq. 12-38). Each of the two phosphotransferase
steps in the phosphatase action apparently occurs with
inversion. The simplest interpretation of all the exper-
imental results is that phosphotransferases usually act by
in-line S
N
2-like mechanisms which may involve metaphos-
phate-ion-like transition states that are constrained to react
with an incoming nucleophile to give inversion. An adjacent
attack with pseudorotation would probably retain the
original configuration and is therefore excluded.
The substrate for many phosphotransferases is
MgATP. Which of the possible isomers of this chelate
complex is utilized by these enzymes? Since Mg
2+
associates and dissociates rapidly from the complexes
there are several possibilities: a tridentate complex with
oxygens from , , and phospho groups coordinated
with the metal ion, an ,-bidentate, a ,-bidentate, or
a monodentate complex. Most evidence suggests that
,-bidentate complexes of the following types are the
true substrates.
(12-36)
The first structure drawn is for a MgATP S
complex with a chiral -phosphothioate group.
The Mg
2+
is expected to bond to oxygen. How-
ever, in the second complex, in which Cd
2+
has
been substituted for Mg
2+
, it is expected that the
Cd
2+
will bond to sulfur. Therefore, the stereo-
chemically equivalent structure will be obtained
only when the compound has the S configuration
at the phosphorus. In the third of the foregoing
complexes, Mg
2+
has been replaced by Cr
3+
to
give an exchange-inert complex of ATP in
which the Cr
3+
will remain attached firmly to
the same two oxygen atoms under most experimental
conditions. Notice also that in this complex, the AMP
portion occupies a different position than in the first two
complexes. It is called the screw-sense isomer,
702
while the upper two complexes are screw-sense
isomers. We may reasonably expect that an enzyme,
which may provide additional ligands to the metal
ions, will prefer one or the other of these screw-sense
isomers. For ,-bidentate ATP complexes both and
phosphorous atoms become chiral centers and even
more isomers are possible.
The use of exchange inert Cr
3+
and Co
3+
complexes
of ATP has been developed by Cleland and associ-
ates.
702 705
The ,-bidentate chromium complexes
were separated into the and isomers.
704
Each was
separated further into a pair of ring-puckering isomers.
These metal complexes are all competitive inhibitors of
MgATP and both ring puckering isomers of the screw
sense are very slow substrates of various kinases. The
isomers serve as very slow substrates for pyruvate
kinase, adenylate kinase, and fructose-6-phosphate
kinase.
703
The exo isomer is the strongest inhibitor
of creatine kinase, suggesting this same conformation
for the MgATP substrate. The disastereoisomers of
ATPS and ATPS have also been tested with kinases
in the presence of either Mg
2+
or Cd
2+
. With creatine
kinase
695,706
the isomers with the R configuration at
phosphorus are preferred in the presence of Mg
2+
but
those with the S configuration are preferred in the pres-
ence of Cd
2+
. This might be related to the previously
mentioned preferences of the metals for O vs. S ligands,
and would suggest that this enzyme prefers the isomer
(see foregoing structures). However, EPR studies with
the corresponding Mn
2+
complexes suggest that the
isomer is preferred by the enzyme.
693
There are 12
isomers of monoammine Cr(III)ATP. Their use has
provided information about the location of water
molecules in metal complexes of kinases.
707
In the following sections we will consider several
individual phosphotransferases.
O
P O
P
O Mg
O
O
O AMP
S
O
P O
P
S Cd
O
O
O AMP
O
O
P O
P
O Cr
O
O
O
O
CdAT S complex
with screw sense,
S configuration at P
CrATP complex
with screw sense
AMP
MgATP S complex
with screw sense,
R configuration at P
+
D. Displacement on a Phosphorus Atom
O P
O
O
O
OH
CH
2
C
OH
CH
3
H
HO
CH
2
C
OH
CH
3
H
O
P O
O
O
16
17
18
S-propane-1,2-diol
(R)
16
17
18
Phenyl[(R)-
16
O,
17
O,
18
O] phosphate

644 Chapter 12. Transferring Groups by Displacement Reactions


The product 1-phospho-(S)-propane-1,2-
diol 1 (here shown as R at phosphorus) is
converted by in-line ring closure to an
equimolar mixture of three cyclic diesters.
These are methylated to give six cyclic
triesters. Of these, only 3 and 6 give sharp
31
P resonance because the
17
O in the others
broadens the lines.
Two cyclic triesters 3 and 6 from an R-
phospho compound and two others 8 and
9 from an S-phospho compound will give
sharp
31
P NMR resonances.
Predicted and observed
31
P NMR spectra of
the mixtures of syn and anti cyclic triesters
derived from labeled samples of 1 phospho-
(S)-propane-1,2-diols that are R and S at
phosphorus.
H
3
C
H
OH
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
O
O
P
CH
3
OCH
3
CH
3
OCH
3
CH
3
CH
3
2 5
3 6
4 7
Syn cyclic
triesters
Anti cyclic
triesters
Set of cyclic
diesters
(R) 1
O
O
O
O
R-phospho-compound
O
O
P
OCH
3
O
O
P
O
CH
3
O
O
P
CH
3
O
O
P
O
OCH
3
3
6
8
9
Syn
Anti
+ +
S-phospho-compound
1 Hz/division
1 Hz/division
Predicted Observed
R
S
O =
16
O
=
17
O
=
18
O
Figure 12-22 Method for determining
chirality of phospho groups containing
16
O,
17
O, and
18
O. From Buchwald and
Knowles.
701
645
4. Phosphatases
The phosphatases catalyze hydrolysis of phosphate
esters to produce inorganic phosphate:
667
The acid phosphatases and alkaline phosphatases
are nonspecific and cleave many different phosphate
esters, whereas glucose-6-phosphatase, fructose-
1,6-bisphosphatase, and many others are specific for
single substrates. The nonspecific phosphatases may
provide inorganic phosphate ions in places where they
are needed, e.g., in mineralizing bone. All phosphatases
help to drive metabolic cycles (Chapter 17).
The alkaline phosphatases are found in bacteria,
fungi, and higher animals but not in higher plants. In
E. coli alkaline phosphatase is concentrated in the peri-
plasmic space. In animals it is found in the brush border
of kidney cells, in cells of the intestinal mucosa, and in
the osteocytes and osteoblasts of bone. It is almost absent
from red blood cells, muscle, and other tissues which
are not involved extensively in transport of nutrients.
The alkaline phosphatase of E. coli is a dimer of
449-residue subunits which requires Zn
2+
, is allo-
sterically activated by Mg
2+
, and has a pH optimum
above 8.
667,708 711
At a pH of ~ 4, incubation of the
enzyme with inorganic phosphate leads to formation
of a phosphoenzyme. Using
32
P-labeled phosphate, it
was established that the phosphate becomes attached
in ester linkages to serine 102. The same active site
sequence Asp-Ser-Ala is found in mammalian alkaline
phosphatases. These results, as well as the stereo-
chemical arguments given in Section 2, suggest a
double-displacement mechanism of Eq. 12-38:
The active site contains two Zn
2+
ions and one Mg
2+
ion
which are held by imidazole and carboxylate groups.
The inorganic phosphate in an enzymeproduct com-
plex is bound to both zinc ions (Fig. 12-23). The Ser
102 side chain is above one Zn. In the enzyme-P inter-
mediate it would be linked to the phospho group as
an ester which would then be hydrolyzed, reversibly,
by a water molecule bound to Zn.
712713a
This water
presumably dissociates to Zn
+
OH and its bound
hydroxyl ion carries out the displacement. This reaction
may be preceded by a proton transfer to an oxygen atom
of the phospho group.
714
P O O R
O
O
OH R

+ H
2
O + HPO
4
2
(12-37)
Ser 102OH
(Phosphatase)
Ser 102OP
(EnzymeP)
P
i
H
2
O
ROH
ROP
Substrate
Acid phosphatases, which have pH optima of ~ 5
and are inhibited by fluoride ion, occur in bacteria,
fungi, plants, and animals. In bone, the acid phosphatase
content is high in the osteoclasts which function in the
resorption of calcium from bone. The highest content
of acid phosphatase in humans is in the prostate and
an elevated serum level has long been used as a diag-
nostic indicator of prostatic cancer.
715
Acid phosphatase
is a periplasmic enzyme in E. coli.
716
Phosphoenzymes
have been trapped from both plant and animal acid
phosphatases.
717
For example, a brief incubation with
32
P-labeled p-nitrophenyl phosphate followed by rapid
denaturation in an alkaline medium gave a covalently
labeled protein from which
32
P-containing N -phos-
phohistidine was isolated. In agreement with kinetic
evidence that a phosphoenzyme is a true intermediate,
there is no inversion of chiral phospho groups by liver
or prostate acid phosphatases.
718
Some acid phosphatases from animals and plants
are violet in color and contain iron (Chapter 16) and an
Mn
3+
-containing acid phosphatase has been isolated
from sweet potatoes.
720
These enzymes have dimetal
centers, often containing one Zn
2+
and one Fe
3+
with
bridging carboxylate and hydroxide ions between the
metals. Imidazole, tyrosinate, and carboxylate side
chains hold the metals as in Fig. 16-20. A water mole-
cule bound to the Fe
3+
is thought to dissociate with
a low pK
a
of 4.8 to give an Fe
3+

OH complex. The
hydroxyl ion can then attack the phospho groups, one
(12-38)
D. Displacement on a Phosphorus Atom
R166
N
H
N
H
H
N
H
H
O
O
P
O
O
H
Zn
2+
D327
O
O
Zn
2+
O
Ser 102
H
H331
N
N
H N
NH
H412
O
D369
O

D51
O
N
N
H
H370
+
2
Mg
2+
O

Figure 12-23 Schematic drawing of the product inorganic


phosphate bound in the active site of E. coli alkaline phos-
phatase. See Ma and Kantrowitz.
719
646 Chapter 12. Transferring Groups by Displacement Reactions
of whose oxygen atoms is coordinated with the Zn
2+
.
The mechanism resembles that proposed for a
phosphotriesterase (Fig. 12-24). The triesterase cata-
lyzes detoxification of organophosphorus toxins such
as parathion (Box 12-E) and seems to have evolved
rapidly from a homologous protein of unknown func-
tion.
721
The phosphotriesterase contains two Zn
2+
ions
in a dimetal center. An unusual structural feature is a
carbamate group, formed from Lys 169 and CO
2
, which
provides a bridging ligand for the metal pair.
721 725
A
carbamylated lysine also functions in ribulose bisphos-
phate carboxylase (Fig. 13-11).
Phosphatases specific for such substrates as glucose-
6-phosphate, fructose-1,6-bisphosphate, and phospho-
glycolate help to drive metabolic cycles (Chapter 17).
The 335-residue fructose-1,6-bisphosphatase associ-
ates to form a tetramer with D
2
symmetry.
726 730
The
allosteric enzyme exists in two conformational states
(see Chapter 11). Activity is dependent upon Mg
2+
or
other suitable divalent cation, e.g., Mn
2+
or Zn
2+
, and is
further enhanced by K
+
or NH
3
+
. While the dimetal
sites depicted in Figs. 12-23 and 12-24 are quite rigid and
undergo little change upon formation of complexes
with substrates or products, the active site of fructose-
1,6-bisphosphatase is more flexible. There are three
metal-binding sites but they contain no histidine side
chains and have been seen clearly only in a product
complex.
727,728
Perhaps because of the need for
flexibility involved in allosteric changes, the active
site is not fully formed until the substrate binds.
Fructose-2,6-bisphosphatase forms one domain
of a bifunctional kinase-phosphatase (Chapter 11).
It has two imidazole rings, as well as side chains from
a glutamate and two arginine residues at the catalytic
and substrate-binding site.
728a
The 357-residue mammalian glucose-6-phospha-
tase plays an important role in metabolism (Chapter
17). Defects in the enzyme cause a glycogen storage
disease (Box 20-D) and severe disruption of metabo-
lism.
731
However, the molecular basis of its action is
not well-known. Furthermore, the active site of the
enzyme is located in the lumen of the endoplasmic
reticulum
732
and glucose-6-phosphate must pass in
through the plasma membrane. An additional glucose-
6-phosphate transporter subunit may be required to
allow the substrate to leave the cytoplasm.
73
Pyrophosphatases, which are present in all cells,
and catalyze hydrolysis of inorganic pyrophosphate
(PP
1
) to orthophosphate (P
i
) (see Chapter 6, Section D),
also drive metabolic sequences. The very active pyro-
phosphatase of E. coli has a turnover number of over
2 x 10
4
s
1
at 37C. The 1000 molecules per cell are
sufficient to immediately hydrolyze any pyrophosphate
produced by bacterial metabolism.
733
The much studied
soluble pyrophosphatases of E. coli,
734,735
yeast,
736
and
other organisms
736a,b
are metalloenzymes that are most
active with Mg
2+
. Two Mg
2+
ions are held, mostly by
carboxylate side chains, while a third apparently enters
the active site as magnesium pyrophosphate, perhaps
MgP
2
O
7

. As with other metallohydrolases, a metal-


bound hydroxyl ion may serve as the attacking nucleo-
phile.
At least three distinct families of protein phospha-
tases remove phosphate groups from serine, threonine,
and tyrosine side chains in proteins.
737
Their role in
control of numerous biochemical processes has been
discussed in Chapter 11, Section C,2. The catalytic
domains or subunits of the protein phosphatases act
together with regulatory domains or separate regula-
tory subunits to control thousands of reactions. For
example, protein phosphatase 1 (PP1) together with a
glycogen-targeting subunit dephosphorylates inactive
glycogen kinase (see Fig. 11-4). Belonging to the same
family is calcineurin (PP2B), a phosphatase activated
by Ca
2+
through binding to calmodulin (Box 6-D). There
are two families of Ser/Thr phosphatases. Their poly-
peptide folding patterns differ, but the active sites have
similar dimetal centers resembling those in Figs. 12-24
and 16-20 with Mn
2+
+ Fe
2+
, Zn
2+
+ Fe
3+
, and probably
other pairs of metals.
737 740
The family containing PP1
has weak sequence homology with the purple acid
phosphatases.
740
Another common feature of these
enzymes is a conserved His-Asp dyad (His 125 and
Asp 95 in PP1) which is thought to be a proton donor,
protonating the leaving group ( O

) in a manner
Figure 12-24 Hypothetical event in the action of a phos-
photriesterase. A carbamylated lysine (lower center), as well
as a water molecule, bridge the two Zn
2+
ions, which are
held by imidazole and aspartate carboxylate groups. The
bound H
2
O can be deprotonated to give the HO

complex
shown. The substrate may displace the HO

ion from the


right-hand zinc and thereby move close to the bound HO

which attacks as indicated. Based on Cd


2 +
-containing
structure and discussion by Benning et al.
722
D301
O
HN
N
Zn
2+
O

C
NH
+
K169

OH
2
Zn
2+
OH

OH
2
H55
N
H
N
R
O
P
OEt
OEt
O
N
H
N
N
H57
H201
H230
N
H
647
reminiscent of the serine proteases. The reaction is
evidently initiated by attack of an

OH ion held by the


dimetal center. This would resemble the mechanism
pictured in Fig. 12-24 except that the phospho group
would carry two negative charges.
The protein tyrosine phosphatases also exist as
several families with numerous functions in control
of transcription, growth, differentiation, and metabo-
lism.
741 743
These enzymes function by a double-
displacement mechanism, as in Eq. 12-38, but with a
cysteine side chain rather than serine. The cysteine is
present in the conserved sequence (H/V)CX
5
R(S/T).
The arginine binds the phospho group and helps to
stabilize the transition state, which probably is meta-
phosphate-like.
742
5. Ribonucleases (RNases)
Many hydrolases act on phosphodiester linkages,
which abound in nature.
725,744
Some are digestive
enzymes but others serve more specific metabolic
functions. Ribonuclease A (RNase A), the pancreatic
digestive enzyme responsible for breakdown of RNA,
was one of the first enzymes for which a structure was
deduced. By 1963 Moore and Stein and their associates,
who had earlier developed ion exchange methods for
separating amino acids and peptides (Fig. 3-6), had
determined the sequence of the 124-residue bovine
enzyme.
745
They observed that Lys 41 was unusually
reactive with dinitrofluorobenzene and that photo-
oxidation of His 12 and His 119, which are almost at
opposite ends of the peptide chain, inactivates the
enzyme. They concluded that both histidines are at
the active site, a conclusion that was later substantiated
by X-ray crystallography.
746 748
A segment 12 nucleo-
tides in length can fit into the cleft in the enzyme that
contains the active site. The negatively charged phos-
phates of the RNA backbone form 8 9 electrostatic bonds
to lysine and arginine side chains of the enzyme.
749,750
However, the only close interactions of the nucleic acid
bases with the enzyme occur at the site of cleavage as
shown in Fig. 12-25. The four residues His 12, Lys 41,
Thr 45, and His 119 are strictly conserved in the RNAse
A superfamily.
749
The carboxylate of Asp 121 apparently
helps to orient the proper tautomer of His 119 for
catalysis.
751
Ribonuclease A was the first enzyme to be synthe-
sized in the laboratory. Fully active ribonuclease has
been synthesized,
752
as have new modified enzymes.
For example a 63-residue peptide made up of five
segments of the native RNase sequence retained mea-
surable catalytic activity.
753
Using total synthesis,
unnatural amino acids, such as 4-fluorohistidine, have
been incorporated at specific positions in RNAse.
752
Cleavage of a phosphodiester linkage in the sub-
strate chain occurs in two steps. In the first or trans-
esterification step, the hydroxyl group on the 2' position
of the ribose ring is thought to be deprotonated by
attack of either the imidazole of His 12, as shown in
Fig. 12-25, or by the adjacent amino group of Lys 41.
(In the latter case His 12 would have to remove a
proton from the NH
3
+
group of Lys 41 before it could
attack.) In either case the positive charge of Lys 41
would help to neutralize the negative charge on the
phosphate. The deprotonation of the 2' OH may
occur synchronously with its attack on the adjacent 3'
phospho group. An in-line displacement of the oxygen
attached to the 5' carbon of the next nucleotide unit is
thought to be assisted by His 119. Its protonated imida-
zolium group may transfer a proton to a phosphate
oxygen atom prior to or synchronously with formation
of the new P O bond in step a (Fig. 12-25).
754
The
intermediate formed in step a is a cyclic 2', 3'-diphos-
phate which then undergoes hydrolysis by attack of
a water molecule in step b to give the free nucleoside
3'-phosphate. The overall reaction is a two-step double-
displacement, analogous to that with chymotrypsin,
except that a neighboring group in the substrate rather
than an amino acid side chain is the nucleophilic catalyst.
The pH dependence of the enzyme is in agreement
with this mechanism because there are two pK
a
values
of ~ 5.4 and ~ 6.4 which regulate the catalytic activity.
Microscopic pK
a
values of His 12 and His 119 have
been measured by NMR spectroscopy as ~ 6.1 and ~ 6.3
and are shifted somewhat by binding of nucleotides.
A bacterial peptidase splits a 20-residue fragment
containing His 12 from the N-terminal end of RNase
A. This S-peptide can be recombined with the rest
of the molecule, which is inactive, to give a functional
enzyme called ribonuclease S. In a similar way, resi-
dues 119 124 of RNase A can be removed by digestion
with carboxypeptidase to give an inactive protein
which lacks His 119. In this case, a synthetic peptide
with the sequence of residues 111 124 of RNase A forms
a complex with the shortened enzyme restoring full
activity.
755
Stereochemical studies support in-line mechanisms
for both the transesterification and hydrolysis steps
of ribonuclease catalysis. For example, chiral uridine
2',3'-cyclic phosphorothioates are hydrolyzed with
inversion of configuration, with the diastereoisomer
shown yielding a 2'-monophosphothioate of the R
configuration at phosphorus.
D. Displacement on a Phosphorus Atom
O
CH
2
Uracil
O O
P
O S

2'
3'
HO
H
2
O
Uridine 2',3'-cyclic phosphorothioate
648 Chapter 12. Transferring Groups by Displacement Reactions
Transesterification step is also in-line
757
as it is shown
in Fig. 12-25. Study of kinetic isotope effects in H
2
O
D
2
O mixtures suggested that two protons may move
synchronously as the enzymesubstrate complex
passes through the transition state.
758
Although
RNase A is one of the most studied of all enzymes,
there are still uncertainties about the mechanism. Is a
proton removed first by His 12 (Fig. 12-25), as has long
been assumed, or is a proton transferred first from His
119 to the oxygen of the phospho group?
759,760
Is the
reaction concerted, as suggested by kinetic isotope
effects,
761
or is there a pentacovalent intermediate?
The specificity of RNase A for a pyrimidine on the
3' side of the phosphodiester bond that is cleaved is
evidently ensured by the pair of hydrogen bonds from
O-2' of the pyrimidine to the backbone NH of Thr 45
and a second from the N-4' proton to the side chain
OH of the same threonine (Fig. 12-25). Other nucleases,
such as ribonuclease T
2
,
762
with different specificities
also make use of hydrogen bonding of the base at the
3' side of the cleavage point with backbone amide
groupings.
Various bacterial ribonucleases as well as the fungal
ribonucleases T
1
, U
1
, and U
2
(see also Fig. 5-43) have
amino acid sequences related to that of RNase A
763,764,764a
but with distinctly different three-dimensional struc-
tures. The active sites contain Glu, His, and Arg side
chains. For RNase T
1
, Glu 58 and His 92 appear to
provide acidbase catalysis with assistance from Tyr
38, Arg 77, and His 40.
763,765
A glutamate carboxylate
also appears to be the catalytic base in the related RNase,
called barnase, from Bacillus amyloliquefaciens.
766
In addition to extracellular digestive enzymes, the
RNase family contains many intracellular enzymes
that are involved in turnover of RNA.
767,768
RNase H
digests away RNA primers during DNA synthesis
(Chapter 27). RNase H activity is also present in a
domain of viral reverse transcriptases and is absolutely
essential for the replication of HIV and other retro-
viruses.
769
The structures of the reverse transcriptase
RNase H domain and of the E. coli enzyme are simi-
lar.
769 771
Unlike RNase A, the RNases H are metallo-
enzymes which apparently contain two Mg
2+
ions held
by carboxylate groups and utilize a metal bound HO

ion as in previously discussed phosphatases. Secreted


RNases sometimes have specific functions. For example,
the 123-residue angiogenin is homologous to pancreatic
RNases but acts to induce formation of new blood
vessels (angiogenesis).
772 774
This is essential to growth
of solid cancers as well as for normal growth. The
enzyme is a very poor catalyst but its RNase activity
appears essential for its biological function. Mutation
of any of the catalytic residues His 13, Lys 40, or His
114 abolishes all angiogenic activity. A neurotoxin
secreted by eosinophils
775
is one of a group of selec-
tively toxic RNases.
776,777
Intracellular RNases are often
found as complexes of specific inhibitor proteins.
778
O
Base
O
OH
P O
O
O
O
N
O
N
O
O
H
H
O
N
N
NH
NH
3
H
N
HN
H
CH
3
C
O
O
Base
HO
OH
P
O
O
O
O
N
O
N
NH
NH
3
His 12
N
HN
H
H
O
H
O
Base
O
OH
P O
O
O
O
N
O

N
NH
+
His 119
His 12
Thr 45
NH
3
H
+

H
Lys 41
N
HN
+
+
H

+
HO
+
H
a Transesterification
b Hydrolysis
Figure 12-25 Proposed two-step in-line reaction mechanism
for ribonuclease A. The hydrogen bonding that provides
recognition of the pyrimidine base at the 3' end created by
the cleavage is also shown. See Wladkowski et al.
756
649
6. Ribonuclease P, Ribozymes, and Peptidyl
Transferase
A very different ribonuclease participates in the
biosynthesis of all of the transfer RNAs of E. coli.
Ribonuclease P cuts a 5' leader sequence from pre-
cursor RNAs to form the final 5' termini of the tRNAs.
Sidney Altman and coworkers in 1980 showed that the
enzyme consists of a 13.7-kDa protein together with a
specific 377-nucleotide RNA component (designated
M1 RNA) that is about five times more massive than
the protein.
779
Amazingly, the M1 RNA alone is able
to catalyze the ribonuclease reaction with the proper
substrate specificity.
780 782a
The protein apparently
accelerates the reaction only about twofold for some
substrates but much more for certain natural substrates.
The catalytic center is in the RNA, which functions
well only in a high salt concentration. A major role of
the small protein subunit may be to provide counterions
to screen the negative charges on the RNA and permit
rapid binding of substrate and release of products.
783
Eukaryotes, as well as other prokaryotes, have enzymes
similar to the E. coli RNase P. However, the eukaryotic
enzymes require the protein part as well as the RNA
for activity.
784
Thomas Cech and associates independently dis-
covered another class of catalytic RNA molecules. These
are self-splicing RNAs that cut out intervening se-
quences from themselves to generate ribosomal RNA
precursors (see Chapter 28).
785 787
They act only once
and are therefore not enzymes. However, the introns
that are cut out during self-splicing are ribozymes
which, like the RNA from ribonuclease P, can act
catalytically and have properties similar to those of
protein enzymes. They exhibit the kinetic properties
of enzymes and are denatured by heat. The RNAs are
folded into compact structures resembling those of
globular proteins. Like tRNA, they contain loops and
hydrogen-bonded stems. Phylogenetic comparisons
(Chapter 29) of the M1 RNA of ribonuclease P iso-
lated from various species have allowed prediction
of precise secondary structures.
783,788,789
A simplified
M1 RNA consisting of 263 nucleotides from conserved
regions of the molecule is catalytically effective.
783
Tetrahymena ribozyme also has a complex structure
with a 247-nucleotide catalytic core formed by two
structural domains (Fig. 12-26).
790,791
The crystal struc-
ture of a third ribozyme, one found in the RNA of the
human pathogen hepatitis delta virus (HDV), has also
been determined.
792
It is a smaller 72-nucleotide self-
cleaved molecule with a very different structure from
that in Fig. 12-26. It makes use of a double pseudo-
knot (see Fig. 5-29) to bind the RNA into a compact,
tightly hydrogen-bonded structure with a deep active
site cleft. It is the fastest known naturally occurring
self-cleaving RNA and is able to react at a rate of more
than 1 s
1
at its optimum temperature of 65C.
Smaller self-cleaving RNAs have been found
among plant viruses and viroids. Many of them have
a common catalytic core which can be converted into
30- to 40-nucleotide ribozymes. Only 17 nucleotides
and three hydrogen-bonded helical stems are required
to form the self-cleaving hammerhead domain, which
has a structural similarity to the catalytic core of the
Tetrahymena ribozyme. The hammerhead ribozymes
(Fig. 12-27) represent one form of small ribozyme.
793 797
Another is the hairpin ribozyme shown in Fig.
12-28,
798,801
which also shows the even smaller lead-
dependent leadzyme, a ribozyme that doesnt
occur in nature.
In an intact viral self-cleaving RNA the entire
catalytic center is formed from a single strand. Stems
II and III of the hammerhead ribozyme (Fig. 12-27C)
are closed by large loops. In the ribozyme shown, loop
III has been cut off and stem II has been closed by a
tight loop to form a compact catalytic RNA that will
cut a substrate having a suitable nucleotide sequence
for binding to the ribozyme. Only 12 bases in this ribo-
zyme are highly conserved. By varying the sequences
in the ribozyme half of stems I and III, catalysts that
cleave after any sequence GUX, where X=A, C, or V,
can be designed. Such catalysts are useful in the labo-
ratory and potentially also in medicine.
What groups of a ribozyme bind to substrates and
what groups participate in catalysis? Like peptides,
RNAs have amide groups that can hydrogen bond to
substrates. Adenine and cytosine can supply protonated
amino groups which could participate in acidbase
catalysis. This is evidently the case in the peptidyl
transferase centers of ribosomes. The RNA in these
centers catalyzes a transesterification in which an
aminoacyl group is transferred from an aminoacyl-tRNA
anto the growing polypeptide chain attached to a
second tRNA molecule. The reaction is evidently
catalyzed by a universally conserved adenine ring
located at position 2486 in the Haloarcula marismortii
23S RNA (position 2451 in E. coli). There are no protein
groups within 1.8 nm of the location of peptide bond
synthesis.
798a
The active site adenine appears to be
much more basic than normal. A high pK
a
of 7.6 controls
the peptidyl transferase, and also controls the methy-
lation of the active site adenine by dimethylsulfate.
798b
The site of protonation is thought to be largely N3 of
adenine 2486, which is probably the basic center in-
volved in catalysis. The peptidyltransferase reaction
may be initiated as follows.
798a
D. Displacement on a Phosphorus Atom
650 Chapter 12. Transferring Groups by Displacement Reactions
Figure 12-26 (A,B) Stereoscopic views of
the Tetrahymena ribozyme. (A) Ribbon
tracing of the phosphodiester backbone.
The successive conserved structural ele-
ments, from the 5' and 3' ends are designat-
ed P1 to P9. The P4 P6 region forms one
major domain, while the P3 and P5 P9
regions form a second domain which folds
around the first. From Golden et al.
790
(B)
Structure of the P4 P6 domain viewed
from the back side as pictured in (A). From
Cate et al.
791
The active site lies between
this domain and the larger one which folds
over it leaving the active site in a deep
crevice.
790
The active site is centered above
the tetraloop GAAA (residues G150 to
A153), which is shown with lighter shading
(in B) near the upper center of the P4 P6
domain. (C). Details of one of three layers
of hydrogen-bonded interactions between
purine and pyrimidine bases involved in
interaction of the tetraloop with the adja-
cent helix in the P5 region. Cleavage occurs
in the P1 domain, which folds into the
active site and is not shown in these draw-
ings. Courtesy of Thomas Cech.
A
B
C
A similar catalytic mechanism is probably used by the
small 85-nucleotide hepatitis delta virus ribozyme
whose catalytic base is thought to be N3 of cytosine 75,
which is associated with a pK
a
of ~ 6.1.
798c
Both this
HDV ribozyme and the ribosomal RNA resemble serine
proteases with histidine as the catalytic base. Howev-
er, the self-splicing RNA of Tetrahymena initiates a
nucleophilic attack with the 3' OH group of a gua-
nosine molecule that is bound to a site in the P7 region
(Fig. 12-16A) and which acts as a cofactor (see Fig. 28-
18). Ribonuclease P and all group I and II self-cleaving
introns also use an external nucleophile such as a
guanosine OH and form 3'-OH and O-phosphate or
H
N
H
N
N
N
N
H
N
H
H
C
O
tRNA
R
n+1
O
R
n
O
C
O
H
HN
Peptide
tRNA
3
Aminoacyl tRNA
Adenine 2486
651
Figure 12-27 (A) Structure of a hammerhead ribozyme. The cleavage
site region is drawn with dark lines. (B) The cleavage region showing
the cleavage site in the substrate strand. From Baidya and Uhlenback.
793
(C) Diagram of a hammerhead ribozyme with standard numbering of
nucleotides. The three helical stems are labeled I, II, and III. From Bevers
et al.
796
(D) Simplified cleavage mechanism which resembles step a of
Fig. 12-25 and ignores the known participation of metal ions.
O
Cytosine
O
O O
O
P
O
O
H
17
5'
3'
G G C C
C C G G
U
U
A
G
A G G A
U C C U
U
G
A
G
U
C
U
G
A
G
A
A
A U
C G
U A
C G
C
I
III
II
Substrate
Point of
cleavage
3 5
16.2
16.1
17
1.1
3
15.2
15.1
14
13
12
5
2.1
3
4
5
6 7
8
9
10.1
11
Substrate
Ribozyme
U
A G
C
U U GU C
A A A C A G
5
3
+5
I
G A
A
C A GU
G U C A A
U G
A
II
10
5
C
C
C
C
C
50
U A
C G
C G
A U
G C
III
A C
G A
A U
A A
20
40
C G
A U
C G
A
A
G
U
C
A
U
G
U
U 30
IV
Hairpin ribozyme
U
A
B
Cleavage site
Figure 12-28 (A) A hairpin ribozyme formed from the minus strand of a satellite RNA associated with tobacco ringspot
virus. On the basis of hydroxyl radical footprinting (see Fig. 5-50), to identify protected areas a folding pattern that brings
domains A and B together to form a compact catalytic core has been proposed.
798
(B) A leadzyme, a ribozyme dependent
upon Pb
2+
for cleavage of RNA.
802,803
(C) An RNA-cleaving DNA enzyme.
804
U
G G A C
C C U G
Cleavage site
3
5
C
G A
G
C C A G
G G U C
A G
3
5
9
8 7
6
5
4
3 2
1
10
Substrate
Ribozyme
(leadzyme)
RNA substrate
DNA enzyme
GC G U GG
C G C A C C 5
3
A
G U
C A
GA G A GA
C T C T C T
G G
C C
G A
G G
C C
T A
A A
G C
C A
T
5
3
Cleavage site
D. Displacement on a Phosphorus Atom
C2.1
G1.1
C3
Ribozyme
3
U4
C17
OH
5
Substrate
Cleavage site
A B
C
D
A B
C
652 Chapter 12. Transferring Groups by Displacement Reactions
phosphodiester ends at the cleavage points. Hammer-
head ribozymes utilize the 2'-OH of the ribose at the
cleavage site to form a 2',3'-cyclic phosphate ester as in
step a of the ribonuclease A reaction (Fig. 12-25) and as
indicated in Fig. 12-27D. Ribozymes act by in-line
mechanisms, causing inversion of the configuration at
the phosphorus.
799,800
Most ribozymes, as well as RNase P,
805,806
require
one or two metal ions for activity.
807
Magnesium ions
predominate and many Mg
2+
ions are bound at distinct
sites in crystalline ribozymes. Hammerhead and hair-
pin ribozymes work reasonably well with monovalent
ions. One proposed mechanism is for an HO

ion
bound to the Mg
2+
to remove H
+
from the 2' OH of the
ribose ring as follows:
When the pro-R oxygen of the phospho group (labeled
R in the accompanying structure) was replaced by sulfur
the rate of cleavage decreased 10
3
-fold. However, the
rate was restored fully when Cd
2+
was added, presum-
ably because of the high affinity of cadmium ions for
sulfur ligands.
808
This supports the possibility of a
dual role for Mg
2+
in activating a water molecule to
provide HO

and in stabilizing negative charges on


the phospho group by interaction with the pro-R oxygen
in the transition state. Several investigators have sug-
gested that two metal ions may be needed. One possi-
bility is for a mechanism similar to that proposed for
alkaline phosphatase (Fig. 12-25) and other phospho-
transferases.
809,810
The Tetrahymena ribozyme functions
best if both Mg
2+
and Mn
2+
are present.
811
A second
metal may act as a Lewis acid facilitating loss of the
5'-OH as indicated by M
2+
( in gray) in the preceding
diagram. Metal ions may also participate in conforma-
tional changes as well as have structural functions in
ribozymes.
812,813
Studies of the kinetics of action of
hammerhead ribozymes have suggested that the intrin-
sic ribozyme-substrate binding energy is utilized for
catalysis.
814
This may be possible because the ribozyme
is only partially folded in the ground state but it folds
into a tighter conformation in the transition state.
815
Methods have been devised for generating enor-
mous numbers of RNA molecules with random se-
quences and for selecting those with unusual catalytic
activities.
816 819
Among the new catalysts produced in
this way are very small ribozymes that cleave RNA
specifically in the presence of Pb
2+
(Fig. 12-28B).
802,803
The leadzyme is more active with neodymium (Nd
3+
)
+ Pb
2+
than with lead alone, suggesting a two-metal
mechanism.
820
Other artificial ribozymes include RNA
ligases,
817
acyltransferases,
821
and DNA hydrolases.
822,823
Is it possible to find a DNA enzyme? Without the 2' OH
of ribose to form hydrogen bonds it seemed doubtful,
but an RNA-cleaving DNA enzyme has been selected
from a population of ~ 10
14
different small DNA mole-
cules. The DNA enzyme (Fig. 12-28C) will cleave RNA,
whose sequence fulfills the base pairing requirements
of two 8-deoxynucleotide recognition domains. Cleav-
age occurs between an unpaired purine and a paired
pyrimidine using a metal-dependent mechanism that
gives a 2',3'-cyclic phosphate as in ribonuclease A
cleavage.
804
7. Deoxyribonucleases (DNases)
A multitude of nucleases cleave DNA, single- or
double-stranded. They range from the pancreatic
digestive enzyme DNase I through specialized nucleases
that function during DNA repair and the hundreds of
restriction endonucleases that have become so valuable
in modern laboratory work. Some nucleases leave a
3'-phosphate ester at a cut end in a DNA chain, while
others leave a 5'-phosphate end.
824
Many nucleases
are dealt with in later chapters. Only a few will be
mentioned here.
One of the most studied enzymes of this group is
the 149-residue micrococcal (staphylcoccal) nuclease
from Micrococcus which cleaves either RNA or single-
or double-stranded DNA. The relatively nonspecific
enzyme cuts nearly randomly at the 5' side of the
phosphodiester linkages, leaving 3'-phosphate groups.
It enhances the uncatalyzed hydrolysis rate at least
10
16
-fold.
825 827
The crystal structure showed that the
majority of the acidic and basic side chains of the
protein interact with each other through clusters of
hydrogen bonds. At the active site the side chains of
both Arg 35 and Arg 87 form pairs of hydrogen bonds
to the 5'-phosphate group of the specific inhibitor
deoxythymidine 3',5'-diphosphate (Fig. 12-29). While
Arg 87 appears to be in a position to protonate the
leaving group O

,
13
C NMR experiments showed that
all of the arginine side chains had pK
a
values above
11.6. However, Tyr 85 has a pK
a
of 9.5, which appears
to control k
cat
/K
m
.
828
A Y85F mutant lacks this pK
a
.
The X-ray structure also suggests that Glu 43 may be
the attacking nucleophile and that it may deprotonate
a water molecule bound to the Glu 43 carboxylate.
The resulting HO

probably carries out a direct in-line


attack as shown in Fig. 12-29. Mutants such as E43D,
E43Q, and E43S have greatly decreased activity,
829
in
O
Base
O
O O
H
P
O O
O
M
2
Mg
O H
+
+

+
(S) (R)
653
Figure 12-29 Drawing showing the hydrogen-bonding
interactions between the guanidinium ions of arginines 35
and 87 of the micrococcal (staphylococcal) nuclease with the
5'-phosphate of the inhibitor thymidine 3',5'-diphosphate in
the complex of E + I + Ca
2+
. A possible mechanism is illus-
trated. A hydroxyl ion bound to Ca
2+
carries out an in-line
attack on the phosphorus. See Libson et al.
826
O
H
N
O
O
O
N
H
H
3
C
N
O
O
O
P
O

H
N
H
H
O R35
+
R87
O
P
H
2
N
N
N
E43
C
O
O
H
O
H O
O H
H
O
H
Ca
2+
T41
H
O
O
H
D21
C
O
O
H
O
H
H
H
N H
+

D40

O
agreement with this mechanism. The nearby Ca
2+
is
essential. For this nonspecific nuclease there is no
hydrogen bonding of a purine or pyrimidine base of
the substrate to the enzyme.
The digestive enzyme, pancreatic DNAse I, makes
single-stranded cuts in double-stranded (ds) DNA.
An exposed strand of peptide chain from the enzyme
binds into the minor groove of B-type DNA.
830
Because
this groove becomes too narrow in long (A+T) rich
sequences, they are cleaved slowly. Certain hypersen-
sitive sites are cleaved very rapidly, perhaps because
the DNA at these regions is bent or is able to bend to
give a very good fit to the enzyme active site. A histi-
dine which is hydrogen bonded to a nearby carboxy-
late of a glutamate side chain appears to be a catalytic
base that acts upon a water molecule as in phospholi-
pase A (Section D,10), displacing the 3' oxygen of the
phosphodiester linkage. An imidazolium group from
a second histidine is hydrogen bonded to an aspartate
carboxylate and a tyrosine OH to form a catalytic
triad that can protonate the 3' O

as it is displaced.
831
Two Mg
2+
ions are also required. Both are held by
different carboxylate side chains and may also interact
with oxygen atoms of the phospho group to neutralize
charge and stabilize the transition state.
In contrast to DNAse I, the restriction endonu-
cleases, which are discussed in Chapter 5, Section H,2
and in Chapter 26, have precise substrate sequence
specificities. Three of the best known restriction endo-
nucleases are called EcoRI,
832 834
an enzyme which binds
to and cuts both strands of the palindromic sequence
5'-GAATTC; EcoRV,
835
which cuts both strands in the
center of the sequence 5'-GATATC; and BamH1, which
binds to the sequence 5'-GGATCC and cleaves after
the 5' G on each strand.
836
A high-resolution structure
is also known for Cfr10I, which recognizes the less strict
sequence 5'-PuCCGGPy and cleaves both strands after
the 5' Pu.
837
All of these enzymes require Mg
2+
and have
active sites containing carboxylate groups. Two-metal
mechanisms have been suggested.
Restriction endonuclease EcoRI is able to cut a
chain in dsDNA which has a chiral phophorothioate
group at the specific cleavage site.
838
The reaction
occurs with inversion of configuration at phosphorus,
suggesting direct in-line attack by a hydroxyl ion
generated from H
2
O.
Attempts are being made to design semisynthetic
restriction endonucleases specific for single-stranded
DNA or RNA. For example, an oligonucleotide with
a sequence complementary to a sequence adjacent the
linkage that is to be cut can be covalently linked to a
relatively nonspecific nuclease. Such an enzyme derived
from micrococal nuclease cuts a single-stranded chain
of either DNA or RNA adjacent to the double-stranded
region of the ES complex.
839
8. Mutases
Phosphotransferases that shift phospho groups from
one position within a substrate to another are often
called mutases. For example, phosphoglucomutase
catalyzes the interconversion of glucose 1-phosphate
and glucose 6-phosphate, an important reaction that
bridges glycogen metabolism and glycolysis (Fig. 11-2).
This 561-residue protein operates through formation of
an intermediate phosphoenzyme.
840842
The phospho
group becomes attached to the OH of Ser 116 and can
be transferred either to the 6 or the 1 position of a glucose
phosphate (step a and reverse of step b in Eq. 12-39).
The two-step reaction accomplishes the reversible
isomerization of glucose 1-phosphate and glucose 6-
phosphate via glucose 1,6-bisphosphate. Evidently,
the glucose bisphosphate, without leaving the enzyme
is reoriented to allow transfer of the phospho group to
either the 1- or 6-position.
841
The phospho enzyme is
relatively unstable and can undergo hydrolysis to free
enzyme and P
i
. To prevent loss of active enzyme in this
way, a separate reaction (catalyzed by a kinase; Eq. 12-39,
step c)
843
generates glucose 1,6-bisphosphate, which
rephosphorylates any free enzyme formed by hydroly-
sis of the phospho enzyme. The glucose 1,6-bisphos-
phate can be regarded as a cofactor or cosubstrate for
the reaction.
Phosphoglycerate mutase, which interconverts
2-phosphoglycerate and 3-phosphoglycerate in glyco-
lysis (Fig. 10-3, step c), functions by a similar mecha-
D. Displacement on a Phosphorus Atom
654 Chapter 12. Transferring Groups by Displacement Reactions
nism.
843 845
However, the enzyme-bound phospho
group is carried on an imidazole group. The essential
cosubstrate required by some phosphoglycerate mu-
tases is 2,3-bisphosphoglycerate.
845
It is formed from
the glycolytic intermediate 1,3-bisphosphoglycerate
(Fig. 10-3) by action of another mutase, bisphospho-
glycerate mutase.
843,846
1,3-bisphosphoglycerate 2,3-bisphosphoglycerate
This is also the pathway for synthesis of 2,3-biphos-
phoglycerate in red blood cells where it serves as an
important allosteric regulator (Chapter 7). Two human
phosphoglycerate mutase isoenzymes are known.
One is found in muscle and the other in brain and
other tissues.
844
A hereditary lack of the muscle type
enzyme is one of the known types of glycogen storage
diseases (Box 20-D).
Human phosphomannomutase, which catalyzes
the interconversion of mannose 1- and 6- phosphates,
appears to carry the phospho group on an aspartate
side chain in the sequence DXDX (T/V), which is
conserved in a family of phosphomutases and phos-
phatases.
847
The first aspartate in the sequence is
phosphorylated during the enzymatic reaction.
9. Molecular Properties of Kinases
Kinases transfer phospho groups from polyphos-
phates such as ATP to oxygen, nitrogen, or sulfur
atoms of a second substrate.
848
Examples include
hexokinase, the enzyme responsible for synthesis of
glucose 6-phosphate from free glucose and ATP (Fig.
11-2, step a); phosphofructokinase, which forms
fructose 1,6 bisphosphate in the glycolysis pathway
(Fig. 11-2, step b); and phosphoglycerate kinase, and
pyruvate kinase, both of which form ATP from ADP
in the glycolysis pathway (Fig. 10-3, steps b, c, and f).
There are many others. Kinases vary greatly in size
and in three-dimensional structure. For example, a
small adenylate kinase, which phosphorylates AMP
to ADP (Eq. 6-65), is a 22-kDa monomer of 194 residues.
Pyruvate kinase is a tetramer of 60-kDa subunits and
muscle phosphofructokinases are tetramers of 75- to
85-kDa subunits. The three-dimensional structures also
vary. While all kinases consist of two domains built
around central sheets (Fig. 12-30), there are several
different folding patterns.
849
The two-domain struc-
tures all have deep clefts which contain the active sites.
Both adenylate kinase (Fig. 12-30) and hexokinase
crystallize in two or more forms with differing confor-
mations.
850
This and other evidence suggests that as a
kinase binds and recognizes its two correct substrates,
the active site cleft closes by a hinging action that
brings together the reacting molecules in the correct
orientation.
851 853
In the crystal structure shown in
Fig. 12-30 both ADP and AMP are bound in a nonpro-
ductive complex. If the ADP were replaced with ATP
(or the AMP with a second ADP) to form a productive
complex the two reacting phospho groups would be
~ 0.8 nm apart. A reaction could not occur without
further closing of the active site cleft.
854
Evidence for
domain closure has been obtained for many other
kinases. For example, substrate complexes of phos-
phoglycerate kinase have been crystallized in both
open forms and closed forms in which a 30
hinge-bending movement has brought the ligands
together for an in-line phospho group transfer.
855
For many enzymes an ATP binding site has been
revealed by study of nonhydrolyzable analogs of ATP
such as AMP-PNP whose structure is shown in
Fig. 12-31.
856
AMP-PNP has been used in thousands
of investigations of ATP-dependent processes.
857
For example, the structure of a phosphoglycerate
Mg
2+
AMP-PNP complex in the active site of phos-
phoglycerate kinase has been determined.
858
Modeling
of a transition state complex indicates that all three
negatively charged oxygens of the ATP portion of struc-
ture are stabilized by hydrogen bonding.
852
Related
analogs such as AMP-PCH
2
P (Fig. 12-3) have been used
in similar ways.
859
Another analog Mg
2+
Ap
5
A (Fig.
12-31) is a bisubstrate inhibitor which binds to adenylate
kinases, fixing the enzymes in a closed conformation
that is thought to resemble the transition state.
859,860
The Mg
2+
complex of ATP is regarded as the true
substrate for kinases. The metal usually also binds
both to the phospho groups of ATP and to groups on
(12-40)
O
OH
HO
HO
OH
O PO
3
2
O
OH
HO
HO
O PO
3
2
O
OH
HO
HO
OH
Glucose-1,6-bisphosphate

Glucose-1-phosphate

Glucose-6-phosphate
O PO
3
2

O PO
3
2

a
b
ESerOPO
3
2
ESerOH

c
ATP
ADP
Kinase
(12-39)
655
Figure 12-30 Stereoscopic -carbon
plots of a 194-residue subunit of adeny-
late kinase from the archaebacterium
Sulfolobus acidocaldarius with ADP (left
side) and AMP (right side) bound into
the active site. From Vonrheim et al.
854
Courtesy of G. E. Schulz.
the enzyme. Crystallographic investigations as well as
studies with exchange-inert ATP complexes (Section 2)
have suggested that the metal ion is bound initially to
the terminal () and adjacent () phospho groups of
ATP. However, this metal bridge could prevent rather
than assist the reaction. For some phosphotransferase
reactions with exchange-inert complexes the product
appears to be the ,-bidentate complex of ADP (see
Section 2) suggesting that movement of the metal ion
from the ,- to the ,-bidentate ATP complex may
occur either prior to or during transfer of the phospho
group. Theoretical calculations also suggest a movement
in the metal complexation as the reaction progresses.
861
EPR studies that involved observation of hyperfine
coupling between
17
O in substrates and Mn
2+
in the
active site led Reed and Leyh to conclude that the
activating metal ion is bound to all three phospho
groups in the transition state.
849
O
HO OH
N
N
N
NH
2
O
P
O
O O
P
N
O
O
P O
HO
O
O P
O
O
O
P
O
O
O
P O
O
HO
Adenosine O
P
O
O O
P
H

O
O

O in ATP
S in ATPS
CF
2
in AMPCF
2
P
AMP-PNP
Ap
5
A, a bisubstrate inhibitor of adenylate kinase
H
N
Figure 12-31 Some useful analogs of ATP.
Adenylate kinase performs the essential function
of recovering AMP formed by many enzymatic pro-
cesses and converting it to ADP (Eq. 6-65) which can
be reconverted to ATP by oxidative or substrate level
phosphorylation. The enzyme is present in all organ-
isms. In vertebrates different isoenzymes function
in the cytosol, mitochondrial intermembrane space,
and mitochondrial matrix.
862,863
A group of other
nucleotide and deoxynucleotide kinases convert
nucleoside monophosphates into diphosphates.
864,865
Some of them, e.g., uridylate kinase are similar in
structure and properties to adenylate kinase.
866,867
Another member of the adenylate kinase family is
phosphoribulokinase, an important photosynthetic
enzyme (see Fig. 17-14, step a).
868
Most kinases transfer chiral phospho groups with
inversion and fail to catalyze partial exchange reactions
that would indicate phosphoenzyme intermediates.
However, nucleoside diphosphate kinase contains
an active site histidine which is phosphorylated to
form a phosphoenzyme.
869
The enzyme catalyzes
phosphorylation of nucleoside diphosphates other
than ADP by a nucleotide triphosphate, usually ATP.
Here, X is usually adenosine and Y is any ribonucleo-
side or deoxyribonucleoside. This enzyme supplies all
of the nucleotide triphosphates except ATP for use in
the many cellular processes that require them.
870 872
The enzyme aligns the substrate, holding the phospho
group with a pair of arginines and a magnesium ion.
The phospho group is aligned for an in-line displace-
ment by N

of His 122, part of a hydrogen-bonded


HisGlu dyad. Formation of the phosphoenzyme occurs
in less than 1 ms without significant conformational
change other than a 30 rotation of the histidine ring.
870,873
XTP
(usually ATP)
XDP
YTP
YDP E His P
E His
(12-41)
D. Displacement on a Phosphorus Atom
656 Chapter 12. Transferring Groups by Displacement Reactions
Hexokinase, the enzyme that phosphorylates
glucose to glucose 6-phosphate, exists as four isoen-
zymes in mammals. Hexokinases I, II, and III are large
~ 100-kDa monomers with similar amino acid sequences,
a single active site, and complex allosteric regulatory
properties. The three-dimensional structure is known
for hexokinase I, which is the pacemaker of glycolysis
in the brain.
874,875
Hexokinase IV (glucokinase) is a
50-kDa protein that is found in liver and in the cells
of the pancreas. It has a low K
m
and is not inhibited by
glucose 6-phosphate, properties that allow for rapid
uptake of glucose after a meal.
876
Its properties are
similar to those of a major isoenzyme of yeast,
877
a
dimer of identical 50-kDa subunits whose structure
is also known.
878
The glucose-binding residues are
conserved in yeast hexokinase, in glucokinase, and in
brain hexokinase. The sequence of the latter suggests
that it may have arisen by a doubling of a shorter
hexokinase gene.
879
Fructose 6-phosphate kinase (phosphofructo-
kinase) has attracted much attention because of its
regulatory properties (Chapter 11).
880 882
Prokaryotic
forms are somewhat simpler.
883
The related fructose
6-phosphate 2-kinase is a component of a bifunctional
kinase-phosphatase (Fig. 11-2, steps d and e) and has a
structure similar to that of adenylate kinase.
884
Phosphoglycerate kinase is encoded by the mam-
malian X chromosome. Several mutant forms are asso-
ciated with hemolytic anemia and mental disorders.
885,886
Mutation of Glu 190, which is in the hinge region far
from the active site, to Gln or Asn markedly reduces
enzymatic activity.
887
Creatine kinase transfers a phospho group to a
nitrogen atom of the quanidinium group of creatine (Eq.
12-31 and Fig. 12-19). Several isoenzymes participate
in its function of buffering the ATP level in tissues
such as muscle fibers, neurons, photoreceptors, and
spermatozoa which experience high and fluctuating
energy needs.
888
A form from the mitochondrial inter-
membrane space of chicken heart is an octomer of 380-
residue subunits.
889
The structurally and mechanisti-
cally similar arginine kinase has an analogous function
in many invertebrates, e.g., in the horseshoe crab, which
provided enzyme for a structure determination.
890
The ~ 500-residue subunits of pyruvate kinase
consist of four domains,
891
the largest of which contains
an 8-stranded barrel similar to that present in triose
phosphate isomerase (Fig. 2-28). Although these two
enzymes catalyze different types of reactions, a com-
mon feature is an enolic intermediate. One could
imagine that pyruvate kinase protonates its substrate
phosphoenolpyruvate (PEP) synchronously with the
phospho group transfer (Eq. 12-42). However, the
enzyme catalyzes the rapid conversion of the enolic
form of pyruvate to the oxo form (Eq. 12-43) adding
the proton sterospecifically to the si face. This and
other evidence favors the enol as a true intermediate
(12-42)
and a product of the phosphotransfer step.
891
Pyruvate
kinase requires not only two equivalents of a divalent
cation such as Mg
2+
or Mn
2+
but also a monovalent
cation, usually K
+
. However, Li
+
, Na
+
, NH
4
+
, Rb
+
, Tl
+
,
and others can substitute. The monovalent cation
induces an essential conformational change. Using
205
Tl NMR it was found that the thallium ion binds
about 0.6 nm from Mn
2+
that is also present in the
active site.
892
All three metals interact directly with the
phospho group of ATP.
891
Pyruvate kinase is a regu-
lated allosteric enzyme present in four isoenzymic
forms in mammals.
893894a
Protein kinases, which were discussed in Chap-
ter 11, phosphorylate selected OH groups of serine,
threonine, and tyrosine side chains in proteins. Exami-
nation of the sequences in the complete genome of
yeast (Saccharomyces cerevisiae) indicates the presence
of at least 113 protein kinase genes, which account for
~ 2% of the total DNA.
895
Higher eukaryotes have
more. While structures of these enzymes vary widely,
they share a common two-domain catalytic core struc-
ture.
896,897
The best known, and one of the simplest of
them, is the catalytic subunit of cyclic AMP-dependent
protein kinase.
897,898
The substrate MgATP binds into
the active site cleft with the -phospho group protrud-
ing to meet the appropriate site of a bound protein
substrate (Fig. 12-32). Most protein kinases are regu-
lated by an activation loop that must be correctly
placed before the ES complex can be formed. As dis-
cussed in Chapter 11, regulation of the cAMP-dependent
kinase depends upon inhibition by a regulatory sub-
unit. In the cAMP-dependent kinases and many other
protein kinases the activation loop (not shown in Fig.
12-32), which helps to form the substrate site, contains
a phosphothreonine residue which is essential for activity.
It is a stable feature of the cAMP-activated kinase,
incorporated at position 197, but for some tyrosine
kinases it is generated by autophosphorylation.
899
HO C
COO
CH
2
O C
COO
CH
3

Enolpyruvate
(12-43)
Adenosine O
P
O P O
O
OH
P
O
O
C
COO
C
O
O
H
H
H

PEP
+
O C
CH
3
COO

Pyruvate
O O

ADP
ATP
657
Figure 12-32 The active site of cyclic AMP-depen-
dent protein kinase with bound ATP and a segment
of an inhibitory peptide (green) blocking the sub-
strate site. The small ball is an alanine side chain; it
would be Ser or Thr in a substrate. The ATP is
clamped by the glycine-rich loop at the top of the
figure. Hydrogen bonds from peptide NH groups
bind the and phospho groups of ATP. A magne-
sium ion (not shown) also binds to the and
phosphate oxygens and to the invariant Asp 184.
The Asp 166 carboxylate is probably the catalytic
base for deprotonation of the substrate OH. From
Grant et al.
896
Courtesy of Susan S. Taylor.
For the enzyme to be activated the phosphothreonine
must form a hydrogen-bonded ion pair with Lys 189
and be hydrogen bonded to His 87, tying together
critical regions of the catalytic domain. The regulatory
subunits, unless occupied by cAMP (Chapter 11) are
competitive inhibitors of the substrates.
900
Cyclic
GMP-activated kinases also have distinct functions.
901
In tryosine kinases
902 904
C-terminal src-homology
domains (Fig. 7-30 and Chapter 11) fold over and
interact in an inhibitory fashion until an appropriate
activating signal is received (see Fig. 11-13).
A relative of the kinases is adenylate cyclase,
whose role in forming the allosteric effector 3',5'-cyclic
AMP (cAMP) was considered in Chapter 11. This
enzyme catalyzes a displacement on P

of ATP by the
3'-hydroxyl group of its ribose ring (see Eq. 11-8 , step
a). The structure of the active site is known.
905
Studies
with ATPS suggest an in-line mechanism resembling
that of ribonuclease (step a, Eq. 12-25). However, it is
Mg
2+
dependent, does not utilize the two-histidine
mechanism of ribonuclease A, and involves an aspar-
tate carboxylate as catalytic base.
906
All isoforms of
adenylate cyclase are activated by the subunits of
some G proteins (Chapter 11). The structures
907
of G
s
and of its complex with adenylate kinase
905
have been
determined. The G
s
activator appears to serve as an
allosteric effector.
Guanylate cyclases, which form cyclic GMP,
occur in particulate and soluble forms.
908
The latter
have been of great interest because they are activated
by nitric oxide (NO). The soluble guanylate cyclases
are heterodimers. The C-terminal regions of both
and subunits are homologous to the catalytic domain
of adenylate cyclase. The N-terminal domain of the
subunits contains heme whose Fe atom is coordinated
by a histidine imidazole.
908,908a
This iron atom is appar-
ently the receptor for NO, a major gaseous hormone,
which is discussed in Chapter 18.
10. Nucleotidyl Transferases
An important group of enzymes transfer substit-
uted phospho groups, most often nucleotidyl groups.
The nucleases, ATPases, and GTPases, which have
already been discussed, belong to this group as do
the nucleic acid synthesizing enzymes, the DNA and
RNA polymerases,
909 911
reverse transcriptase,
912
and topoisomerases. As with other phosphotransfer-
ases, the nucleotidyl transfers occur with inversion
913,913a
and crystallographic investigations also support in-line
mechanisms as illustrated in the following scheme.
Two metal ions assist.
The inorganic pyrophosphate formed is hydrolyzed
to inorganic phosphate by pyrophosphatase. Specific
information about the polymerases and topoisomerases
is given in Chapters 27 and 28.
O
Base
O
HO H (or OH)
P O
P
O P
O
O
O
O
O
O
O
3'
O
H
5'-Polynucleotide

D. Displacement on a Phosphorus Atom


658 Chapter 12. Transferring Groups by Displacement Reactions
E. The Adenylate Kinase Fold, the P Loop, and
ATPases and GTPases
A magnesiumATP-binding fragment consisting
of ~ 40 residues at the N terminus of adenylate kinase
contains sequences homologous to those in the GTP-
binding G proteins such as the protooncogenes ras
(Fig. 11-7A) and also to sequences in myosin and in
mitochondrial ATP synthase.
914
This includes the
glycine-rich P loop which extends from Gly 15 to
Gly 22 in the porcine cytosolic enzyme and contains a
highly conserved lysine [Lys 21 of porcine adenylate
BOX 12-F THE TOXICITY OF ALUMINUM
Aluminum, in the form of oxides and silicate
minerals, is the most abundant element in the earths
crust. Yet it appears to be actively excluded from
most living organisms. It seems surprising that a
naturally occuring Al
3+
-dependent enzyme hasnt
been found, but there is no evidence that Al is an
essential element. Until recently, it was usually
regarded as harmless. The aluminum salts known
as alums are used in baking powders and have been
added to pickles and other foods. Aluminum sulfate
is often used as a coagulant to clarify turbid drink-
ing water. Because of the insolubility of Al(OH)
3
and of aluminum phosphate the concentration of
Al
3+
is very low at neutral pH. However, small
amounts of AlOH
2+
, Al(OH)
2
+
, and Al(OH)
4

are
present in water. Fluoride complexes such as AlF
2+
may also be present. Soluble complexes, such as that
with citrate, may permit some Al
3+
to be absorbed
by the body.
a,b
The toxicity of aluminum has been recognized
most clearly by the development of bone disease
caused by deposition of Al in bones of patients
on hemodialysis
a c
and in infants on intravenous
therapy.
d,e
Excessive Al in the water used for dialysis
may also cause brain damage. Dietary aluminum
may be one cause of Alzheimers disease,
f h
but this
is controversial as is a possible role of aluminum
in vaccines in causing inflammation in muscle.
i,j
Solubilization of soil aluminum by acid rain has
been blamed for the decline of forests in Europe and
North America,
j
for the death of fish in acid waters,
k
and for very large reductions in yield for many
crops.
l,m
An aluminum-resistant strain of buckwheat
makes and secretes from its roots large amounts of
oxalate which binds and detoxifies the Al
3+
ions.
m
Al
3+
has a radius somewhat less than that of Fe
3+
(Table 6-10) and it may sometimes occupy empty
Fe
3+
binding sites. Thus, the transferrin
n
in blood
carries some Al
3+
, although citrate is probably a more
important carrier.
o
Al
3+
binds preferentially to oxy-
gen ligands and can compete with Mg
2+
. However,
the slower rate of ligand exchange reactions with
Al
3+
may interfere with the proper functioning of
the metal. Brain hexokinase is strongly inhibited
p
by Al
3+
and the binding of Al
3+
to tubulin decreases
a
Martin, R. B. (1986) Clinical Chemistry 32, 17971806
b
Macdonald, T. L., and Martin, R. B. (1988) Trends Biochem. Sci.
13, 1519
c
Andress, D. L., Kopp, J. B., Maloney, N. A., Coburn, J. W., and
Sherrard, D. J. (1987) N. Engl. J. Med. 316, 292296
d
Sedman, A. B., Klein, G. L., Merritt, R. J., Miller, N. L., Weber, K.
O., Gill, W. L., Anand, H., and Alfrey, A. C. (1985) N. Engl. J.
Med. 312, 13371342
e
Bishop, N. J., Morley, R., Chir, B., Day, J. P., and Lucas, A. (1997)
N. Engl. J. Med. 336, 15571561
f
Good, P. F., and Perl, D. P. (1993) Nature (London) 362, 418
g
Shen, Z. M., Perczel, A., Hollsi, M., Nagypl, I., and Fasman,
G. D. (1994) Biochemistry 33, 96279636
h
Walker, P. R., LeBlanc, J., and Sikorska, M. (1989) Biochemistry
28, 39113915
i
Landsberg, J. P., McDonald, B., and Watt, F. (1992) Nature
(London) 360, 6568
ij
Malakoff, D. (2000) Science 288, 13231324
j
Godbold, D. L., Fritz, E., and Httermann, A. (1988) Proc. Natl.
Acad. Sci. U.S.A. 85, 38883892
k
Birchall, J. D., Exley, C., Chappell, J. S., and Phillips, M. J. (1989)
Nature (London) 338, 146148
l
Barinaga, M. (1997) Science 276, 1497
m
Ma, J. F., Zheng, S. J., Matsumoto, H., and Hiradate, S. (1997)
Nature (London) 390, 569570
n
Roskams, A. J., and Connor, J. R. (1990) Proc. Natl. Acad. Sci.
U.S.A. 87, 90249027
o
Martin, R. B., Savory, J., Brown, S., Bertholf, R. L., and Wills, M.
R. (1987) Clinical Chemistry 33, 405407
p
Viola, R. E., Morrison, J. F., and Cleland, W. W. (1987) Biochemis-
try 19, 31313137
q
Macdonald, T. L., Humphreys, W. G., and Martin, R. B. (1987)
Science 236, 183186
r
Troullier, A., Girardet, J.-L., and Dupont, Y. (1992) J. Biol. Chem.
267, 2282122829
s
Chabre, M. (1990) Trends Biochem. Sci. 15, 610
t
Merkx, M., and Averill, B. A. (1999) J. Am. Chem. Soc. 121, 6683
6689
the rates of GTP hydrolysis and of Ca
2+
-induced
depolymerization of microtubules.
q
Aluminofluoride
ions, such as AlF
4

, react with phosphates to form


ions such as,
which may be potent competitive inhibitors of
enzymes acting on ATP, GTP, or other phosphate-
containing substrate.
r,s
However, Fe
3+
can be replaced
by Al
3+
in a purple acid phosphatase (Chapter 16)
with retention of good catalytic activity.
t
R O P O
O
AlF
3

O
659
kinase and Lys 17 of the archaeal enzyme (Fig. 12-30)]
and of the ras oncogene product).
915
The lysine side
chain appears to interact with the and / or phospho
group of ATP or GTP. The peptide chain of the P loop
wraps around the phospho group as the chain turns
from the first strand into the first helix. This can be
seen in Fig. 12-30, in which the P loop surrounds the
phospho group of ADP. Two peptide NH groups also
bind to the phospho group on a side opposite to that
occupied by Mg
2+
. A similar loop in the dinucleotide-
binding domain of a dehydrogenase can be seen hydro-
gen bonded to the P

and P

phospho groups of NAD


+
in Fig. 2-13. Consensus sequences for three groups of
glycine-rich loops are:
896
Dinucleotide-binding G X G X X G
P loop G X X X X G K (S/T)
Protein kinase G X G X X G X V
The protein kinase loop is seen at the top of Fig.
12-32 and extends from Gly 50 to Val 57. These con-
served loops help to hold the ATP in place and to orient
it correctly. Do they have any other significance? The
answer is not clear. These glycine-rich loops fold across
the diphosphate linkage that is broken when ATP
or GTP is hydrolyzed. Cleavage of both of these mole-
cules is associated with movement. Kinases close
around ATP, and parts of G proteins (Fig. 11-7) move
when these regulatory devices function.
915,916
Cleavage
of ATP causes movement in the ATPase heads of muscle
myosin, providing the force for muscular contraction
(Chapter 19). Somewhat the opposite occurs in mito-
chondrial ATP synthase (Chapter 18) when movement
in the synthase heads snaps ADP and inorganic phos-
phate together to form ATP. Common to all of these
processes is a movement of charge within the ATP (or
its cleavage products) from the attack nucleophile into
the neighboring phospho group, as is indicated by the
arrows in the following diagram.
Not shown in this diagram is an accompanying flow
of positive charge which may include movement of
a metal ion, addition and loss of protons, and which
may induce conformational changes.
914
The latter
are essential not only to muscle contraction and ATP
synthesis but also to many other processes that depend
upon the Gibbs energy of cleavage of ATP and related
compounds. This includes the pumping of ions
against concentration gradients (Chapter 8), the action
of topoisomerases (Chapter 27) which function to
alter the supercoiling in DNA, and the functioning of
P O
O
O
HO
P
R
O
O
M
2+

Nu:
the phosphotransferase system by which sugars and
other compounds are brought into bacterial cells
(Chapter 8).
F. Displacements on Sulfur Atoms
Nucleophilic displacement reactions occur on
sulfur atoms in various oxidation states. A common
reaction is thioldisulfide exchange (Eqs. 10-9, 11-7),
a reaction in which a nucleophilic thiolate anion
attacks one of the sulfur atoms of a disulfide. Proteins
such as thioredoxin of E. coli and thioltransferases
(Box 15-C), which contain internal disulfide bridges, can
be reduced by disulfide exchange with thiols such as
glutathione (Box 11-B). The reduced proteins may
then undergo similar exchange reactions that cleave
disulfide linkages in other molecules. An example is
glutathione reductase (Fig. 15-12). Thioltransferases
may also serve as protein disulfide isomerases
(Chapter 10, Section D,3). Nucleophilic displacements
on sulfur or on selenium atoms are steps in a variety
of enzymatic reactions. Among these are glutathione
peroxidase (Eq. 15-59) and thiosulfate: cyanide sulfo-
transferase (Eq. 24-45).
While esters of sulfuric acid do not play as central
a role in metabolism as do phosphate esters, they
occur widely. Both oxygen esters (R O SO
3

, often
referred to as O-sulfates) and derivatives of sulfamic
acid (R NH SO
3

, N-sulfates) are found, the latter


occurring in mucopolysaccharides such as heparin.
Sulfate esters of mucopolysaccharides and of steroids
are ubiquitous and sulfation is the most abundant
known modification of tyrosine side chains. Choline
sulfate and ascorbic acid 2-sulfate are also found in
cells. Sulfate esters of phenols and many other organic
sulfates are present in urine.
Sulfotransferases
917 920a
transfer sulfo groups to
O and N atoms of suitable acceptors (reaction type 1D,
Table 10-1). Usually, transfer is from the active sul-
fate, 3'-phosphoadenosine 5'-phosphosulfate
(PAPS),
921
whose formation is depicted in Eq. 17-38.
Sulfatases catalyze hydrolysis of sulfate esters. The
importance of such enzymes is demonstrated by the
genetic mucopolysaccharidoses. In four of these
disease-specific sulfatases that act on iduronate sulfate,
heparan N-sulfate, galactose-6-sulfate, or N-acetylglu-
cosamine-4-sulfate are absent. Some of these, such as
heparan N-sulfatase deficiency, lead to severe mental
retardation, some cause serious skeletal abnormalities,
while others are mild in their effects.
922
Sulfatases are unusual in having a residue of
formylglycine at the active site. This is generated
oxidatively from cysteine in human enzymes
923,923a
and
from serine in some bacterial sulfatases.
924,924a
Absence
of this modification results in a multiple sulfatase
deficiency disease. A probable mechanism of sulfatase
F. Displacements on Sulfur Atoms
660 Chapter 12. Transferring Groups by Displacement Reactions
based on a crystal structure determination is given by
Eq. 12-44.
923
G. Multiple Displacement Reactions and the
Coupling of ATP Cleavage to Endergonic
Processes
A combination of successive displacement reactions
of two types is required in many enzymatic reactions,
including most of those by which the cleavage of ATP
is coupled to biosynthesis. To harness the group trans-
fer potential of ATP to drive an endergonic metabolic
process there must be a mechanism of coupling.
Otherwise, hydrolysis of ATP within a cell would
simply generate heat. An essential part of the coupling
mechanism usually consists of a nucleophilic displacement
on phosphorus followed by displacement on carbon. Like-
wise, the synthesis of ATP and related compounds
often begins with a displacement on carbon followed
by one on phosphorus.
1. Transfer of Phospho, Pyrophospho, and
Adenylyl Groups from ATP
The first step in coupling ATP cleavage to any
process is transfer of part of the ATP molecule to a
nucleophile Y, usually by displacement on one of the
three phosphorus atoms. The nucleophilic attack may
be (a) on the terminal phosphorus (P

) with displace-
ment of ADP or (b) on the internal phosphorus (P

)
with displacement of inorganic pyrophosphate. In the
first case, Y PO
3
H

is formed; in the latter, Y-adenylyl


(sometimes shortened to Y-adenyl) is formed. More
rarely, displacement occurs (c) on the central phospho-
rus (P

) with transfer of a pyrophospho group to the


(12-44)
nucleophile. Still less frequent (d) is a displacement on
C-5' as shown in Eq. 17-37. If the nucleophile Y in any
of these displacement reactions is H
2
O, the resulting
hydrolysis tends to go to completion, i.e., the phospho,
adenylyl, and pyrophospho groups of ATP all have
high group transfer potentials (Table 6-6). If Y is an
OH group in an ordinary alcohol the transfer reaction
also tends to go to completion because the group
transfer potential of a simple phosphate ester is rela-
tively low. Consequently, phosphorylation by ATP is
often used as a means of introducing an essentially
irreversible step in a metabolic pathway.
2. Acyl Phosphates
Transfer of a phospho or adenylyl group from ATP
to the oxygen atom of a carboxylate group yields an
acyl phosphate, a type of metabolic intermediate of
special significance. Acyl phosphates are mixed anhy-
drides of carboxylic and phosphoric acids in which
both the acyl group and the phospho group have high group
transfer potentials. As a consequence, acyl phosphates
can serve as metabolic intermediates through which
the group transfer potential of ATP is transferred into
other molecules and is harnessed to do chemical work.
A typical example is the synthesis of acetyl coen-
zyme A (Eq. 12-45). See Fig. 14-1 for the complete
structure of SH group-containing coenzyme A.
Adenosine
C O
H
H
P
O
O O
P
O
O
O
P
O
O
OH
ATP

5'

d
b
c
a
R
C
O
P
O
O
H (R)
O
O

Acyl phosphate
(12-45)
H
3
C COO HS CoA
H
3
C C
O
S

Acetate
CoA
Coenzyme A
H
2
O
Acetyl coenzyme A
G (pH 7) = +35.1 kJ mol
1
(+8.4 kcal mol
1
)
E CH
2
SH
Enzyme
Oxidation
E C
H
O
R
O
SO
3

(Substrate)
Formylglycine
form
C
H
O
O
H

R O
C
H
O
O
H
SO
3

SO
4
2

H
661
Because the acetyl group in the product also has a high
group transfer potential, G for this reaction is highly
positive and the formation of acetyl-CoA will not
occur spontaneously. However, the sum of G for the
reaction in Eq.12-45 plus that for the hydrolysis of ATP
(Eq. 12-46) is nearly zero (+ 0.6kJ mol
1
).
ATP
4
+ H
2
O ADP
3
+ HPO
4
2
+ H
+
G = 34.5 kJ mol
1
( 8.3 kcal mol
1
)
Coupling of the two reactions is accomplished by first
letting an oxygen atom of the nucleophilic carboxylate
group attack P

of ATP to form acetyl phosphate (Eq.


12-47, step a). In the second step (step b) the sulfur atom
of the SH group of coenzyme A (often abbreviated
CoA SH) attacks the carbon atom of the acetyl phos-
phate with displacement of the good leaving group P
i
.
While G for Eq. 12-47, step a, is moderately positive
(meaning that a relatively low concentration of acetyl
phosphate will accumulate unless the [ATP] / [ADP]
ratio is high), the equilibrium in Eq. 12-47, step b,
favors the products.
The two reactions of Eq. 12-47 are catalyzed by
acetate kinase
925
and an S-acetyltransferase, respec-
tively. The sequence represents an essential first stage
in bacterial utilization of acetate for growth. It is also
used in a few bacteria in reverse as a way of generating
ATP in fermentation reactions. On the other hand,
most eukaryotic cells make acetyl-CoA from acetate by
coupling the synthesis to cleavage of ATP to AMP and
P
i
. A single enzyme acetyl-CoA synthetase (acetate
thiokinase) catalyzes both steps in the reaction (Eq.
10-1). The sequence parallels that of Eq. 12-47, but the
initial displacement is on P

of ATP to form acetyl


adenylate. This intermediate remains tightly bound
(12-46)
a
b
H
3
C C
O
O
H
3
C C
O
O P OH
O
O
CoA S H

ATP
4
Acetyl
phosphate
ATP
3
S C
O
CH
3
G' (pH 7) = +13.2 kJ mol
-1
Acetyl-CoA
CoA
P
i
G' (pH 7) = 12.6 kJ mol
-1
(12-47)
to the enzyme until the second step in the sequence
takes place. When
18
O is present in the acetate (desig-
nated by the asterisks in Eq. 12-48) it appears in the
phospho group of AMP as expected for the indicated
mechanism.
3. General Mechanism of Formation of
Thioesters, Esters, and Amides
The sequences of Eqs. 12-47 and 12-48 are general
ones used by cells for linking carboxylic acids to OH,
SH, and NH
2
groups to form oxygen esters, thioesters,
or amides, respectively. ATP can be cleaved at either
P

or P

. If cleavage is at P

(Eq. 12-48) the hydrolysis


of inorganic pyrophosphate (PP
i
) to P
i
provides an
additional coupling of ATP cleavage to biosynthesis
as is discussed in Chapter 17, Section H.
An enzyme catalyzing a reaction similar to that of
acetyl-CoA synthetase is succinyl-CoA synthetase
(succinate thiokinase).
926,927
The enzyme from E. coli
has been studied most. The first step is formation of
a phosphoenzyme by transfer of the phospho group
from ATP to N

of histidine 246 in the subunit of the


140-kDa
2

2
tetramer.
926
The phospho group is then
transferred to succinate to form succinyl phosphate,
which reacts with coenzyme A, as in step b, Eq. 12-48,
R C
O
O
P O
O
O
O
P P
Adenosine
R C
O
O
P O Adenosine
O
O
R' SH
R' OH
R' NH
2
P
O Adenosine
O O
O
R C
O
S R'
*
R C
O
O R'

ATP
Acyl adenylate
R C
O
N R'

Thioester
Oxygen ester
*
Amide
H
AMP
a
b
PP
i
2 P
i
H
2
O
Pyrophosphatase
(12-48)
G. Multiple Displacement Reactions and the Coupling of ATP Cleavage to Endergonic Processes
662 Chapter 12. Transferring Groups by Displacement Reactions
to form succinyl-CoA. Crystallographic studies sug-
gest that for this step the succinyl phosphate and the
coenzyme A may be bound to opposite subunits in
the tetramer.
926
However, in some bacteria and in
eukaryotes the enzyme appears to operate as an
heterodimer.
927
Glutamine synthetase,
928,929
a large enzyme
containing 12 identical 468-residue subunits with 622
symmetry (as in Fig. 7-12), has a major regulatory func-
tion in nitrogen metabolism, which is discussed in
Chapter 24. Apparently, the intermediate acyl phos-
phate (-glutamyl phosphate) has a transient existence
and all three reactantsglutamate, NH
4
+
, and ATP
must be bound to the enzyme concurrently before the
active site becomes functional. Early evidence for the
acyl phosphate
930,931
included reduction by sodium
borohydride to an alcohol (Eq. 12-49a) and isolation
of the internal amide of glutamic acid 5-oxoproline
(Eq. 12-49b).
Ammonium ions appear to be bound at a specific site
and to be deprotonated by the Asp 50 carboxylate.
929
The aminoacyl-tRNA synthetases join amino
acids to their appropriate tRNA molecules for protein
synthesis. They have the very important task of select-
ing both a specific amino acid and a specific tRNA and
joining them. The enzymes differ in size and other
properties. However, they all appear to function by a
common basic chemistry that makes use of cleavage of
ATP at P

(Eq. 12-48) via an intermediate aminoacyl


adenylate and that is outlined also in Eq. 17-36. These
enzymes are discussed in Chapter 29 .
4. Coenzyme A Transferases
The following problem in energy transfer arises
occasionally: A thioester, such as succinyl-CoA, is
available to a cell and the energy available in its unstable
linkage is needed for synthesis of a different thioester.
It would be possible for a cell to first form ATP or GTP,
OOC C
O
O
H NH
3
PO
3
2
+

_
N
O
H
OOC
H

5-Oxoproline
P
i
OOC CH
2
OH
H NH
3
+

Homoserine
-Glutamyl phosphate
NaBH
4
P
i
a
b
(12-49)
using a synthetase reaction in reverse; then the ATP or
GTP formed could be used to make the new linkage by
the action of another acyl-CoA synthetase. However,
special enzymes, the CoA transferases, function more
directly (Eq. 12-50). The mechanism is not obvious.
How can the CoA be transferred from one acyl group
to another while still retaining the high group transfer
potential of the acyl group?
The following experiments shed some light.
Kinetic studies of succinyl-CoAacetoacetate CoA
transferase indicate a ping-pong mechanism. The
enzyme alternates between two distinct forms, one of
which has been shown to contain bound CoA.
932934
The E-CoA intermediate formed from enzyme plus
acetoacetyl-CoA was reduced with
3
H-containing
sodium borohydride and the protein was completely
hydrolyzed with HCl. Tritium-containing -amino--
hydroxyvaleric acid was isolated. Since thioesters (as
well as oxygen esters) are cleaved in a two-step process
to alcohols by reduction with borohydride, it was con-
cluded that the intermediate E-CoA is a thioester of
the Glu 344 side chain. In exchange reactions
18
O from
labeled succinate entered both the E-CoA intermediate
and the carboxyl group of acetoacetate.
A mechanism involving formation of a transient
anhydride is similar to reactions discussed in preceding
sections.
717,720
The student should be able to write
out the step-by-step detail. Does this mechanism
explain the
18
O exchange data? A second possibility
is a 4-center reaction (Eq. 12-51). However, mecha-
nisms of this type have not been demonstrated for
enzymatic reactions.
R COO
R C
O
S

CoA
Succinyl-CoA
Succinate
Acyl-CoA
G ~ 0
HO
COO
H
3
N H

+
-Amino--hydroxyvaleric acid
E C
O
O
S
C
CoA
O R

E C
O
S
CoA
+
O
C
O R
(12-50)
(12-51)
663
50. Braun, C., Lindhorst, T., Madsen, N. B., and
Withers, S. G. (1996) Biochemistry 35, 54585463
50a. Namchuk, M. N., McCarter, J. D., Becalski, A.,
Andrews, T., and Withers, S. G. (2000) J. Am.
Chem. Soc. 122, 12701277
51. Gorenstein, D. G., Findlay, J. B., Luxon, B. A.,
and Kar, D. (1977) J. Am. Chem. Soc. 99, 3473
3479
52. Kirby, A. J. (1983) The Anomeric Effect and
Related Stereoelectronic Effects at Oxygen,
Springer, Berlin
53. Bennet, A. J., and Sinnott, M. L. (1986) J. Am.
Chem. Soc. 108, 72877294
54. Sinnott, M. L. (1986) in Mechanisms of
Enzymatic Reactions: Stereochemistry A.
Steenbock Symposium (Frey, P. A., ed), pp. 293
305, Elsevier, New York
55. Phillips, D. C. (1966) Sci. Am. 215 (Nov), 7890
56. Sanz, J. M., Garca, P., and Garca, J. L. (1992)
Biochemistry 31, 84958499
57. Artymicek, P. J., and Blake, C. C. F. (1981) J.
Mol. Biol. 152, 737762
58. Harata, K., Muraki, M., and Jigami, Y. (1993) J.
Mol. Biol. 233, 524535
59. Harata, K., Muraki, M., Hayashi, Y., and
Jigami, Y. (1992) Protein Sci. 1, 14471453
60. Grtter, M. G., Weaver, L. H., and Matthews,
B. W. (1983) Nature (London) 303, 828831
61. Zhang, X.-J., Baase, W. A., and Matthews, B.
W. (1991) Biochemistry 30, 20122017
62. Dao-pin, S., Sauer, U., Nicholson, H., and
Matthews, B. W. (1991) Biochemistry 30, 7142
7153
63. Matsumura, M., Becktel, W. J., Levitt, M., and
Matthews, B. W. (1989) Proc. Natl. Acad. Sci.
U.S.A. 86, 65626566
64. Parsons, S. M., and Raftery, M. A. (1972)
Biochemistry 11, 16231628
65. Maenaka, K., Matsushima, M., Song, H.,
Sunada, F., Watanabe, K., and Kumagai, I.
(1995) J. Mol. Biol. 247, 281293
66. Matsumura, I., and Kirsch, J. F. (1996)
Biochemistry 35, 18811889
67. Kuroki, R., Yamada, H., Moriyama, T., and
Imoto, T. (1986) J. Biol. Chem. 261, 1357113574
68. Malcolm, B. A., Rosenberg, S., Corey, M. J.,
Allen, J. S., de Baetselier, A., and Kirsch, J. F.
(1989) Proc. Natl. Acad. Sci. U.S.A. 86, 133137
69. Hadfield, A. T., Harvey, D. J., Archer, D. B.,
MacKenzie, da, Jeenes, D. J., Radford, S. E.,
Lowe, G., Dobson, C. M., and Jonhson, L. N.
(1994) J. Mol. Biol. 243, 856872
70. Weaver, L. H., Grtter, M. G., and Matthews,
B. W. (1995) J. Mol. Biol. 245, 5468
71. Kuroki, R., Ito, Y., Kato, Y., and Imoto, T.
(1997) J. Biol. Chem. 272, 1997619981
72. Matsumura, I., and Kirsch, J. F. (1996)
Biochemistry 35, 18901896
73. Dickerson, R. E., and Geis, I. (1969) The
Structure and Action of Proteins, Harper and
Row, New York (p. 71)
74. Levitt, M. (1974) in Peptides, Polypeptides and
Proteins (Blout, E. R., Bouey, F. A., Goodman,
M., and Lotan, N., eds), pp. 99113, Wiley,
New York
75. Huang, X., Surry, C., Hiebert, T., and Bennet,
A. J. (1995) J. Am. Chem. Soc. 117, 1061410621
76. Matsui, H., Blanchard, J. S., Brewer, C. F., and
Hehre, E. J. (1989) J. Biol. Chem. 264, 87148716
77. Banait, N. S., and Jencks, W. P. (1991) J. Am.
Chem. Soc. 113, 79517958
78. Zhang, Y., Bommuswamy, J., and Sinnott, M.
L. (1994) J. Am. Chem. Soc. 116, 75577563
79. Tanaka, Y., Tao, W., Blanchard, J. S., and Hehre,
E. J. (1994) J. Biol. Chem. 269, 3230632312
80. Knier, B. L., and Jencks, W. P. (1980) J. Am.
Chem. Soc. 102, 67896798
81. Huang, X., Tanaka, K. S. E., and Bennet, A. J.
(1997) J. Am. Chem. Soc. 119, 1114711154
References
1. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York (pp.
78110)
2. Lowe, J. N., and Ingraham, L. L. (1974) An
Introduction to Biochemical Reaction Mechanisms,
Prentice-Hall, Englewood Cliffs, New Jersey
3. Bruice, T. C., and Benkovic, S. J. (1966)
Bioorganic Mechanisms, Benjamin, New York
(2 vols.)
3a. Kyte, J. (1995) Mechanism in Protein Chemistry,
Garland Publ., New York
3b. Fersht, A. (1999) Structure and Mechanism in
Protein Science: A Guide to Enzyme Catalysis and
Protein Folding, Freeman, New York
3c. Silverman, R. B. (1999) The Organic Chemistry
of Enzyme-Catalyzed Reactions, Academic
Press, San Diego, California
3d. Sinnott, M., ed. (1998) Comprehensive Biological
Catalysis. A Mechanistic Reference, Vol. I,
Academic Press, San Diego, California
4. Dixon, J. E., and Bruice, T. C. (1971) J. Am.
Chem. Soc. 93, 65926597
5. Kosower, E. M. (1968) An Introduction to
Physical Organic Chemistry, Wiley, New York
(pp. 28 and 81)
6. Goldman, P., Milne, G. W. A., and Keister, D.
B. (1968) J. Biol. Chem. 243, 428434
7. Liu, J.-Q., Kurihara, T., Miyagi, M., Esaki, N.,
and Soda, K. (1995) J. Biol. Chem. 270, 18309
18312
8. Hisano, T., Hata, Y., Fujii, T., Liu, J.-Q.,
Kurihara, T., Esaki, N., and Soda, K. (1996) J.
Biol. Chem. 271, 2032220330
9. Li, Y.-F., Hata, Y., Fujii, T., Hisano, T.,
Nishihara, M., Kurihara, T., and Esaki, N.
(1998) J. Biol. Chem. 273, 1503515044
10. Grimmelikhuijzen, C. J. P., and Schaller, H. C.
(1979) Trends Biochem. Sci. 4, 265267
11. Koonin, E. V., and Tatusov, R. L. (1994) J. Mol.
Biol. 244, 125132
11a. Nardini, M., Ridder, I. S., Rozeboom, H. J.,
Kalk, K. H., Rink, R., Janssen, D. B., and
Dijkstra, B. W. (1999) J. Biol. Chem. 274, 14579
14586
12. Verschueren, K. H. G., Selje, F., Rozeboom,
H. J., Kalk, K. H., and Dijkstra, B. W. (1993)
Nature (London) 363, 693698
12a. Ridder, I. S., Rozeboom, H. J., Kalk, K. H., and
Dijkstra, B. W. (1999) J. Biol. Chem. 274, 30672
30678
13. Pries, F., Kingma, J., Krooshof, G. H.,
Jeronimus-Stratingh, C. M., Bruins, A. P., and
Janssen, D. B. (1995) J. Biol. Chem. 270, 10405
10411
13a. Schindler, J. F., Naranjo, P. A., Honaberger, D.
A., Chang, C.-H., Brainard, J. R., Vanderberg,
L. A., and Unkefer, C. J. (1999) Biochemistry 38,
57725778
14. Lightstone, F. C., Zheng, Y.-J., Maulitz, A. H.,
and Bruice, T. C. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 84178420
15. Lightstone, F. C., Zheng, Y.-J., and Bruice, T.
C. (1998) J. Am. Chem. Soc. 120, 56115621
16. Pries, F., van den Wijngaard, A. J., Bos, R.,
Pentenga, M., and Janssen, D. B. (1994) J. Biol.
Chem. 269, 1749017494
17. Chiang, P. K., Gordon, R. K., Tal, J., Zeng, G.
C., Doctor, B. P., Pardhasaradhi, K., and
McCann, P. P. (1996) FASEB J. 10, 471480
18. Cornforth, J. W., Reichard, S. A., Talalay, P.,
Carrell, H. L., and Glusker, J. P. (1977) J. Am.
Chem. Soc. 99, 72927301
19. Woodard, R. W., Tsai, M.-D., Floss, H. G.,
Crooks, P. A., and Coward, J. K. (1980) J. Biol.
Chem. 255, 91249127
20. Vidgren, J., Svensson, L. A., and Liljas, A.
(1994) Nature (London) 368, 354357
21. Fu, Z., Hu, Y., Konishi, K., Takata, Y., Ogawa,
H., Gomi, T., Fujioka, M., and Takusagawa, F.
(1996) Biochemistry 35, 1198511993
22. Takata, Y., Konishi, K., Gomi, T., and Fujioka,
M. (1994) J. Biol. Chem. 269, 55375542
23. Schluckebier, G., OGara, M., Saenger, W., and
Cheng, X. (1995) J. Mol. Biol. 247, 1620
24. Szczelkun, M. D., Jones, H., and Connolly, B.
A. (1995) Biochemistry 34, 1073410743
25. OGara, M., Roberts, R. J., and Cheng, X.
(1996) J. Mol. Biol. 263, 597606
25a. Reddy, Y. V. R., and Rao, D. N. (2000) J. Mol.
Biol. 298, 597610
26. Patel, D. J. (1994) Nature (London) 367, 688690
27. Jeltsch, A., Christ, F., Fatemi, M., and Roth, M.
(1999) J. Biol. Chem. 274, 1953819544
28. Lotta, T., Vidgren, J., Tilgmann, C., Ulmanen,
I., Meln, K., Julkunen, I., and Taskinen, J.
(1995) Biochemistry 34, 42024210
29. Zheng, Y.-J., and Bruice, T. C. (1997) J. Am.
Chem. Soc. 119, 81378145
29a. Kahn, K., and Bruice, T. C. (2000) J. Am. Chem.
Soc. 122, 4651
30. Kealey, J. T., and Santi, D. V. (1995) Biochemis-
try 34, 24412446
31. Hrycyna, C. A., Yang, M. C., and Clarke, S.
(1994) Biochemistry 33, 98069812
31a. Cleland, W. W., OLeary, M. H., and
Northrop, D. B., eds. (1977) Isotope Effects on
Enzyme-Catalyzed Reactions, Univ. Park Press,
Baltimore, Maryland
31b. Klinman, J. P. (1978) in Transition States of
Biochemical Processes (Gandour, R. D., and
Schowen, R. L., eds), pp. 165200, Plenum,
New York
31c. Gandour, R. D., and Schowen, R. L., eds.
(1978) Transition States of Biochemical Processes,
Plenum, New York
31d. Schramm, V. L., Horenstein, B. A., and Kline,
P. C. (1994) J. Biol. Chem. 269, 1825918262
32. Hegazi, M. F., Borchardt, R. T., and Schowen,
R. L. (1979) J. Am. Chem. Soc. 101, 43594365
33. Mihel, I., Knipe, J. O., Coward, J. K., and
Schowen, R. L. (1979) J. Am. Chem. Soc. 101,
43494351
34. Rodgers, J., Femec, D. A., and Schowen, R. L.
(1982) J. Am. Chem. Soc. 104, 32633268
35. Boyd, R. J., Kim, C.-K., Shi, Z., Weinberg, N.,
and Wolfe, S. (1993) J. Am. Chem. Soc. 115,
1014710152
36. Glad, S. S., and Jensen, F. (1997) J. Am. Chem.
Soc. 119, 227232
37. Wolfenden, R., Lu, X., and Young, G. (1998) J.
Am. Chem. Soc. 120, 68146815
38. Henrissat, B., and Bairoch, A. (1993) Biochem.
J. 293, 781788
39. Henrissat, B., and Bairoch, A. (1996) Biochem.
J. 316, 695696
40. Henrissat, B., Callebaut, I., Fabrega, S., Lehn,
P., Mornon, J.-P., and Davies, G. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 70907094
41. Sinnot, M. L. (1990) Chem. Rev. 90, 1171 1202
42. Withers, S. G., and Aebersold, R. (1995)
Protein Sci. 4, 361372
43. Silverstein, R., Voet, J., Reed, D., and Abeles,
R. H. (1967) J. Biol. Chem. 242, 13381346
44. Doudoroff, M., Barker, H. A., and Hassid, W.
Z. (1947) J. Biol. Chem. 168, 725732 and 733
746
45. LeBrun, E., and van Rapenbusch, R. (1980) J.
Biol. Chem. 255, 1203412036
46. McCarter, J. D., and Withers, S. G. (1996) J.
Biol. Chem. 271, 68896894
47. Withers, S. G., Rupitz, K., and Street, I. P.
(1988) J. Biol. Chem. 17, 79297932
48. Withers, S. G., and Street, I. P. (1988) J. Am.
Chem. Soc. 110, 85518553
49. Miao, S., Ziser, L., Aebersold, R., and Withers,
S. G. (1994) Biochemistry 33, 70277032
664 Chapter 12. Transferring Groups by Displacement Reactions
References
82. Hardy, L. W., and Poteete, A. R. (1991)
Biochemistry 30, 94579463
82a. Davies, G. J., Mackenzie, L., Varrot, A.,
Dauter, M., Brzozowski, A. M., Schlein, M.,
and Withers, S. G. (1998) Biochemistry 37,
1170711713
82b. Vocadlo, D. J., Mayer, C., He, S., and Withers,
S. G. (2000) Biochemistry 39, 117126
83. Banerjee, S. K., and Rupley, J. A. (1975) J. Biol.
Chem. 250, 82678274
84. Kelly, J. A., Sielecki, A. R., Sykes, B. D., James,
M. N. G., and Phillips, D. C. (1979) Nature
(London) 282, 875878
85. Metzler, D. E. (1979) Adv. Enzymol. 50, 140
86. Dao-pin, S., Liao, D.-I., and Remington, S. J.
(1989) Proc. Natl. Acad. Sci. U.S.A. 86, 5361
5365
87. Post, C. B., and Karplus, M. (1986) J. Am.
Chem. Soc. 108, 13171319
88. Fife, T. H., Jaffe, S. H., and Natarajan, R.
(1991) J. Am. Chem. Soc. 113, 76467653
89. Piszkiewicz, D., and Bruice, T. C. (1968) J. Am.
Chem. Soc. 90, 21562163
90. Lowe, G., and Sheppard, G. (1968) J. Chem.
Soc. Chem. Commun., 529530
91. Capon, B. (1969) Chem. Rev. 69, 407498
92. Hart, P. J., Pfluger, H. D., Monzingo, A. F.,
Hollis, T., and Robertus, J. D. (1995) J. Mol.
Biol. 248, 402413
93. Terwisscha van Scheltinga, A. C., Armand, S.,
Kalk, K. H., Isogai, A., Henrissat, B., and Dijkstra,
B. W. (1995) Biochemistry 34, 1561915623
94. Tews, I., Terwisscha van Scheltinga, A. C.,
Perrakis, A., Wilson, K. S., and Dijkstra, B. W.
(1997) J. Am. Chem. Soc. 119, 79547959
95. Brameld, K. A., and Goddard, W. A., III (1998)
J. Am. Chem. Soc. 120, 35713580
96. Klyosov, A. A. (1990) Biochemistry 29, 10577
10585
97. Watanabe, H., Noda, H., Tokuda, G., and Lo,
N. (1998) Nature (London) 394, 330331
98. Davies, G. J., Tolley, S. P., Henrissat, B., Hjort,
C., and Schlein, M. (1995) Biochemistry 34,
1621016220
99. Damude, H. G., Withers, S. G., Kilburn, D. G.,
Miller, R. C., Jr., and Warren, R. A. J. (1995)
Biochemistry 34, 22202224
100. Barr, B. K., Wolfgang, D. E., Piens, K.,
Claeyssens, M., and Wilson, D. B. (1998)
Biochemistry 37, 92209229
101. Divne, C., Sthlberg, J., Teeri, T. T., and Jones,
T. A. (1998) J. Mol. Biol. 275, 309325
102. Barr, B. K., Hsieh, Y.-L., Ganem, B., and
Wilson, D. B. (1996) Biochemistry 35, 586592
103. Choi, S. K., and Ljungdahl, L. G. (1996)
Biochemistry 35, 49064910
104. Tormo, J., Lamed, R., Chirino, A. J., Morag, E.,
Bayer, E. A., Shoham, Y., and Steitz, T. A.
(1996) EMBO J. 15, 57395751
105. Sulzenbacher, G., Schlein, M., and Davies, G.
J. (1997) Biochemistry 36, 59025911
106. Sulzenbacher, G., Driguez, H., Henrissat, B.,
Schlein, M., and Davies, G. J. (1996)
Biochemistry 35, 1528015287
107. Johnson, P. E., Joshi, M. D., Tomme, P.,
Kilburn, D. G., and McIntosh, L. P. (1996)
Biochemistry 35, 1438114394
108. Fukuda, M., Spooncer, E., Oates, J. E., Dell, A.,
and Klock, J. C. (1984) J. Biol. Chem. 259,
1092510935
108a. Ponyi, T., Szab, L., Nagy, T., Orosz, L.,
Simpson, P. J., Williamson, M. P., and Gilbert,
H. J. (2000) Biochemistry 39, 985991
109. Domnguez, R., Souchon, H., Lascombe, M.-
B., and Alzari, P. M. (1996) J. Mol. Biol. 257,
10421051
110. Roberge, M., Shareck, F., Morosoli, R.,
Kluepfel, D., and Dupont, C. (1997)
Biochemistry 36, 77697775
111. Juy, M., Amit, A. G., Alzari, P. M., Poljak, R. J.,
Claeyssens, M., Bguin, P., and Aubert, J.-P.
(1992) Nature (London) 357, 8991
112. Chen, L., Fincher, G. B., and Hoj, P. B. (1993) J.
Biol. Chem. 268, 1331813326
113. Chen, L., Garrett, T. P. J., Fincher, G. B., and
Hoj, P. B. (1995) J. Biol. Chem. 270, 80938101
114. Mller, J. J., Thomsen, K. K., and Heinemann,
U. (1998) J. Biol. Chem. 273, 34383446
115. Lindley, M. G., Shallenberger, R. S., and
Herbert, S. M. (1976) Food Chemistry 1, 149159
116. Krengel, U., and Dijkstra, B. W. (1996) J. Mol.
Biol. 263, 7078
117. Kasumi, T., Tsumuraya, Y., Brewer, C. F.,
Kersters-Hilderson, H., Claeyssens, M., and
Hehre, E. J. (1987) Biochemistry 26, 3010 3016
118. Lawson, S. L., Wakarchuk, W. W., and Withers,
S. G. (1996) Biochemistry 35, 1011010118
119. Derewenda, U., Swenson, L., Green, R., Wei,
Y., Morosoli, R., Shareck, F., Kluepfel, D., and
Derewenda, Z. S. (1994) J. Biol. Chem. 269,
2081120814
120. McIntosh, L. P., Hand, G., Johnson, P. E.,
Joshi, M. D., Krner, M., Plesniak, L. A., Ziser,
L., Wakarchuk, W. W., and Withers, S. G.
(1996) Biochemistry 35, 99589966
121. He, X. M., and Carter, D. C. (1992) Nature
(London) 358, 209215
122. Braithwaite, K. L., Barna, T., Spurway, T. D.,
Charnock, S. J., Black, G. W., Hughes, N.,
Lakey, J. H., Virden, R., Hazlewood, G. P.,
Henrissat, B., and Gilbert, H. J. (1997)
Biochemistry 36, 1548915500
123. Sanz-Aparicio, J., Hermoso, J. A., Martnez-
Ripoll, M., Lequerica, J. L., and Polaina, J.
(1998) J. Mol. Biol. 275, 491502
124. Wang, Q., Trimbur, D., Graham, R., Warren, R.
A. J., and Withers, S. G. (1995) Biochemistry 34,
1455414562
125. Namchuk, M. N., and Withers, S. G. (1995)
Biochemistry 34, 1619416202
126. Hrmova, M., MacGregor, E. A., Biely, P.,
Stewart, R. J., and Fincher, G. B. (1998) J. Biol.
Chem. 273, 1113411143
127. Febbraio, F., Barone, R., DAuria, S., Rossi, M.,
Nucci, R., Piccialli, G., De Napoli, L., Orr, S.,
and Pucci, P. (1997) Biochemistry 36, 30683075
128. Jacobson, R. H., Zhang, X.-J., DuBose, R. F.,
and Matthews, B. W. (1994) Nature (London)
369, 761766
129. Gebler, J. C., Aebersold, R., and Withers, S. G.
(1992) J. Biol. Chem. 267, 1112611130
130. Richard, J. P., Westerfeld, J. G., Lin, S., and
Beard, J. (1995) Biochemistry 34, 1171311724
131. Richard, J. P., Huber, R. E., Lin, S., Heo, C.,
and Amyes, T. L. (1996) Biochemistry 35,
1237712386
132. Roth, N. J., Rob, B., and Huber, R. E. (1998)
Biochemistry 37, 1009910107
133. Mitchell, E. P., Withers, S. G., Ermert, P.,
Vasella, A. T., Garman, E. F., Oikonomakos, N.
G., and Johnson, L. N. (1996) Biochemistry 35,
73417355
134. Tu, J.-I., Jacobson, G. R., and Graves, D. J.
(1971) Biochemistry 10, 12291236
135. Gold, A. M., Legrand, E., and Sanchez, G. R.
(1971) J. Biol. Chem. 246, 57005706
136. Levvy, G. A., and Snaith, S. M. (1972) Adv.
Enzymol. 36, 151181
137. Barford, D., and Johnson, L. N. (1989) Nature
(London) 340, 609616
138. Oikonomakos, N. G., Johnson, L. N., Acharya,
K. R., Stuart, D. I., Barford, D., Hajdu, J.,
Varvill, K. M., Melpidou, A. E., Papageor-
giou, T., Graves, D. J., and Palm, D. (1987)
Biochemistry 26, 83818389
139. Oikonomakos, N. G., Zographos, S. E.,
Tsitsanou, K. E., Johnson, L. N., and Acharya,
K. R. (1996) Protein Sci. 5, 24162428
140. Helmreich, E. J. M., and Klein, H. W. (1980)
Angew. Chem. Int. Ed. Engl. 19, 441455
141. Klein, H. W., Im, M. J., Palm, D., and
Helmreich, E. J. M. (1984) Biochemistry 23,
58535861
142. Street, I. P., Rupitz, K., and Withers, S. G.
(1989) Biochemistry 28, 15811587
143. Parrish, R. F., Uhing, R. J., and Graves, D. J.
(1977) Biochemistry 16, 48244831
143a. Watson, K. A., McCleverty, C., Geremia, S.,
Cottaz, S., Driguez, H., and Johnson, L. N.
(1999) EMBO J. 18, 46194632
144. Duke, E. M. H., Wakatsuki, S., Hadfield, A., and
Johnson, L. N. (1994) Protein Sci. 3, 11781196
145. Buchbinder, J. L., Luong, C. B. H., Browner,
M. F., and Fletterick, R. J. (1997) Biochemistry
36, 80398044
146. Buchbinder, J. L., and Fletterick, R. J. (1996) J.
Biol. Chem. 271, 2230522309
147. Gregoriou, M., Noble, M. E. M., Watson, K.
A., Garman, E. F., Krulle, T. M., De La Fuente,
C., Fleet, G. W. J., Oikonomakos, N. G., and
Johnson, L. N. (1998) Protein Sci. 7, 915927
148. Nakano, K., and Fukui, T. (1986) J. Biol. Chem.
261, 82308236
149. Watson, K. A., Schinzel, R., Palm, D., and
Johnson, L. N. (1997) EMBO J. 16, 114
150. Becker, S., Palm, D., and Schinzel, R. (1994) J.
Biol. Chem. 269, 24852490
151. Lin, K., Hwang, P. K., and Fletterick, R. J.
(1995) J. Biol. Chem. 270, 2683326839
152. Goldsmith, E. J., Fletterick, R. J., and Withers,
S. G. (1987) J. Biol. Chem. 262, 14491455
153. Robyt, J. F., and French, D. (1970) J. Biol. Chem.
245, 39173927
154. Brayer, G. D., Luo, Y., and Withers, S. G.
(1995) Protein Sci. 4, 17301742
155. MacDonald, R. J., Crerar, M. M., Swain, W. F.,
Pictet, R. L., Thomas, G., and Rutter, W. J.
(1980) Nature (London) 287, 117122
156. Qian, M., Haser, R., Buisson, G., Due, E., and
Payan, F. (1994) Biochemistry 33, 62846294
157. Qian, M., Haser, R., and Payan, F. (1995)
Protein Sci. 4, 747755
158. Larson, S. B., Greenwood, A., Cascio, D., Day,
J., and McPherson, A. (1994) J. Mol. Biol. 235,
15601584
159. Machius, M., Vrtesy, L., Huber, R., and
Wiegand, G. (1996) J. Mol. Biol. 260, 409421
160. Kadziola, A., Sogaard, M., Svensson, B., and
Haser, R. (1998) J. Mol. Biol. 278, 205217
161. Strobl, S., Maskos, K., Betz, M., Wiegand, G.,
Huber, R., Gomis-Rth, F. X., and Glockshuber,
R. (1998) J. Mol. Biol. 278, 617628
162. Fujimoto, Z., Takase, K., Doui, N., Momma,
M., Matsumoto, T., and Mizuno, H. (1998) J.
Mol. Biol. 277, 393407
163. Feller, G., le Bussy, O., Houssier, C., and Gerday,
C. (1996) J. Biol. Chem. 271, 2383623841
164. Aghajari, N., Feller, G., Gerday, C., and Haser,
R. (1998) Protein Sci. 7, 564572
165. Matsuura, Y., Kusunoki, M., Harada, W., and
Kakudo, M. (1984) J. Biochem. 95, 697702
166. Brzozowski, A. M., and Davies, G. J. (1997)
Biochemistry 36, 1083710845
167. Robyt, J. F., and French, D. (1970) Arch.
Biochem. Biophys. 138, 662670
168. Robyt, J. F., and French, D. (1967) Arch.
Biochem. Biophys. 122, 816
169. delCardayr, S. B., and Raines, R. T. (1994)
Biochemistry 33, 60316037
169a. Dauter, Z., Dauter, M., Brzozowski, A. M.,
Christensen, S., Borchert, T. V., Beier, L.,
Wilson, K. S., and Davies, G. J. (1999)
Biochemistry 38, 83858392
169b. Park, K. H., Kim, M. J., Lee, H. S., Han, N. S.,
Kim, D., and Robyt, J. F. (1998) Carbohydr. Res.
313, 235246
665
215. Cromlish, J. A., Seidah, N. G., Marcinkiewicz,
M., Hamelin, J., Johnson, D. A., and Chrtien,
M. (1987) J. Biol. Chem. 262, 13631373
216. Schaner, P., Todd, R. B., Seidah, N. G., and
Nillni, E. A. (1997) J. Biol. Chem. 272, 19958
19968
217. Rose, C., Vargas, F., Facchinetti, P., Bourgeat,
P., Bambal, R. B., Bishop, P. B., Chan, S. M. T.,
Moore, A. N. J., Ganellin, C. R., and Schwartz,
J.-C. (1996) Nature (London) 380, 403409
218. Shilton, B. H., Thomas, D. Y., and Cygler, M.
(1997) Biochemistry 36, 90029012
219. Bullock, T. L., Branchaud, B., and Remington,
S. J. (1994) Biochemistry 33, 1112711134
220. Mortensen, U. H., Remington, S. J., and
Breddam, K. (1994) Biochemistry 33, 508517
221. Brayer, G. D., Delbaere, L. T. J., and James, M.
N. G. (1979) J. Mol. Biol. 131, 743775
222. Plou, F. J., Kowlessur, D., Malthouse, J. P. G.,
Mellor, G. W., Hartshorn, M. J., Pinitglang, S.,
Patel, H., Topham, C. M., Thomas, E. W.,
Verma, C., and Brocklehurst, K. (1996) J. Mol.
Biol. 257, 10881111
223. Wells, J. A., and Estell, D. A. (1988) Trends
Biochem. Sci. 13, 291297
224. Kossiakoff, A. A., Ultsch, M., White, S., and
Eigenbrot, C. (1991) Biochemistry 30, 12111221
225. Sorensen, S. B., Bech, L. M., Meldal, M., and
Breddam, K. (1993) Biochemistry 32, 89948999
225a. Smith, C. A., Toogood, H. S., Baker, H. M.,
Daniel, R. M., and Baker, E. N. (1999) J. Mol.
Biol. 294, 10271040
226. Koszelak, S., Ng, J. D., Day, J., Ko, T. P.,
Greenwood, A., and McPherson, A. (1997)
Biochemistry 36, 65976604
227. Delbaere, L. T. J., and Brayer, G. D. (1980) J.
Mol. Biol. 139, 4551
228. Blanchard, H., and James, M. N. G. (1994) J.
Mol. Biol. 241, 574587
228a. Tomkinson, B. (1999) Trends Biochem. Sci. 24,
355359
228b. Rose, C., Vargas, F., Facchinetti, P., Bourgeat,
P., Bambal, R. B., Bishop, P. B., Chan, S. M. T.,
Moore, A. N. J., Ganellin, C. R., and Schwartz,
J.-C. (1996) Nature (London) 380, 403409
228c. Renn, S. C. P., Tomkinson, B., and Taghert, P.
H. (1998) J. Biol. Chem. 273, 1917319182
229. Wharton, C. W. (1998) in Comprehensive
Biological Catalysis. A Mechanistic Reference,
Vol. I (Sinnott, M., ed), pp. 345379,
Academic Press, San Diego, California
230. Blow, D. M. (1997) Trends Biochem. Sci. 22,
405408
231. Tsukada, H., and Blow, D. M. (1985) J. Mol.
Biol. 184, 703711
232. Bode, W., and Schwager, P. (1975) J. Mol. Biol.
98, 693717
233. Gaboriaud, C., Serre, L., Guy-Crotte, O.,
Forest, E., and Fontecilla-Camps, J.-C. (1996)
J. Mol. Biol. 259, 9951010
234. Sawyer, L., Shotten, D. M., Campbell, J. W.,
Wendell, P. L., Muirhead, H., and Watson, H.
C. (1978) J. Mol. Biol. 118, 137208
235. Bode, W., Meyer, E., Jr., and Powers, J. C.
(1989) Biochemistry 28, 19511963
236. Stubbs, M. T., and Bode, W. (1995) Trends
Biochem. Sci. 20, 2328
237. Katz, B. A., Liu, B., Barnes, M., and Springman,
E. B. (1998) Protein Sci. 7, 875885
238. Timm, D. E. (1997) Protein Sci. 6, 14181425
239. Blow, D. M. (1971) in The Enzymes, 3rd ed.,
Vol. 3 (Boyer, P. D., ed), pp. 185212,
Academic Press, New York
240. Huber, R., and Bode, W. (1977) in NMR in
Biology (Dwek, R. A., Campbell, I. D.,
Richards, R. E., and Williams, R. J. P., eds),
pp. 131 , Academic Press, New York
241. Bachovchin, W. W., and Roberts, J. D. (1978)
J. Am. Chem. Soc. 100, 80418047
192a. Lu, D., Ftterer, K., Korolev, S., Zheng, X.,
Tan, K., Waksman, G., and Sadler, J. E. (1999)
J. Mol. Biol. 292, 361373
193. Light, A., and Janska, H. (1989) Trends
Biochem. Sci. 14, 110112
194. Matsushima, M., Ichinose, M., Yahagi, N.,
Kakei, N., Tsukada, S., Miki, K., Kurokawa,
K., Tashiro, K., Shiokawa, K., Shinomiya, K.,
Umeyama, H., Inoue, H., Takahashi, T., and
Takahashi, K. (1994) J. Biol. Chem. 269, 19976
19982
195. Kitamoto, Y., Veile, R. A., DonisKeller, H.,
and Sadler, J. E. (1995) Biochemistry 34, 4562
4568
196. Khan, A. R., and James, M. N. G. (1998)
Protein Sci. 7, 815836
197. Hedstrom, L., Lin, T.-Y., and Fast, W. (1996)
Biochemistry 35, 45154523
197a. Khan, A. R., Khazanovich-Bernstein, N.,
Bergman, E. M., and James, M. N. G. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 1096810975
198. Kramer, K. J., Felsted, R. L., and Law, J. H.
(1973) J. Biol. Chem. 248, 30213028
199. Lesk, A. M., and Fordham, W. D. (1996) J. Mol.
Biol. 258, 501537
200. Cygler, M., Schrag, J. D., Sussman, J. L., Harel,
M., Silman, I., Gentry, M. K., and Doctor, B. P.
(1993) Protein Sci. 2, 366382
201. Pereira, P. J. B., Bergner, A., Macedo, R., S,
Huber, R., Matschiner, G., Fritz, H.,
Sommerhoff, C. P., and Bode, W. (1998) Nature
(London) 392, 306311
201a. Sommerhoff, C. P., Bode, W., Pereira, P. J. B.,
Stubbs, M. T., Strzebecher, J., Piechottka, G.
P., Matschiner, G., and Bergner, A. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1098410991
202. Schechter, N. M., Eng, G. Y., Selwood, T., and
McCaslin, D. R. (1995) Biochemistry 34, 10628
10638
203. Johnson, D. A., and Barton, G. J. (1992) Protein
Sci. 1, 370377
204. Kido, H., Yokogoshi, Y., Sakai, K., Tashiro, M.,
Kishino, Y., Fukutomi, A., and Katunuma, N.
(1992) J. Biol. Chem. 267, 1357313579
204a. Wong, G. W., Tang, Y., Feyfant, E., Sali, A., Li,
L., Li, Y., Huang, C., Friend, D. S., Krilis, S. A.,
and Stevens, R. L. (1999) J. Biol. Chem. 274,
3078430793
205. Remington, S. J., Woodbury, R. G., Reynolds,
R. A., Matthews, B. W., and Neurath, H.
(1988) Biochemistry 27, 80978105
206. McGrath, M. E., Mirzadegan, T., and Schmidt,
B. F. (1997) Biochemistry 36, 1431814324
207. Sali, A., Matsumoto, R., McNeil, H. P.,
Karplus, M., and Stevens, R. L. (1993) J. Biol.
Chem. 268, 90239034
208. Springman, E. B., Dikov, M. M., and Serafin,
W. E. (1995) J. Biol. Chem. 270, 13001307
209. Hof, P., Mayr, I., Huber, R., Korzus, E.,
Potempa, J., Travis, J., Powers, J. C., and Bode,
W. (1996) EMBO J. 15, 54815491
210. Rao, N. V., Wehner, N. G., Marshall, B. C.,
Gray, W. R., Gray, B. H., and Hoidal, J. R.
(1991) J. Biol. Chem. 266, 95409548
211. Hershberger, R. J., Gershenfeld, H. K.,
Weissman, I. L., and Su, L. (1992) J. Biol. Chem.
267, 2548825493
212. Beresford, P. J., Kam, C.-M., Powers, J. C., and
Lieberman, J. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 92859290
213. Caputo, A., Garner, R. S., Winkler, U., Hudig,
D., and Bleackley, R. C. (1993) J. Biol. Chem.
268, 1767217675
213a. Harris, J. L., Peterson, E. P., Hudig, D.,
Thornberry, N. A., and Craik, C. S. (1998) J.
Biol. Chem. 273, 2736427373
214. Fuller, R. S., Brake, A. J., and Thorner, J. (1989)
Science 246, 482486
170. Hermans, M. M. P., Kroos, M. A., van
Beeuman, J., Oostra, B. A., and Reuser, A. J. J.
(1991) J. Biol. Chem. 266, 1350713512
171. Raben, N., Nagaraju, K., Lee, E., Kessler, P.,
Byrne, B., Lee, L., LaMarca, M., King, C.,
Ward, J., Sauer, B., and Plotz, P. (1998)
J. Biol. Chem. 273, 1908619092
172. Howard, S., and Withers, S. G. (1998)
Biochemistry 37, 38583864
173. Jespersen, H. M., MacGregor, E. A., Henrissat,
B., Sierks, M. R., and Svensson, B. (1993)
Journal of Protein Chemistry 12, 791805
174. Liu, W., Madsen, N. B., Fan, B., Zucker, K. A.,
Glew, R. H., and Fry, D. E. (1995) Biochemistry
34, 70567061
175. Watanabe, K., Hata, Y., Kizaki, H., Katsube, Y.,
and Suzuki, Y. (1997) J. Mol. Biol. 269, 142153
176. Knegtel, R. M. A., Strokopytov, B., Penninga,
D., Faber, O. G., Rozeboom, H. J., Kalk, K. H.,
Dijkhuizen, L., and Dijkstra, B. W. (1995)
J. Biol. Chem. 270, 2925629264
177. Schmidt, A. K., Cottaz, S., Driguez, H., and
Schulz, G. E. (1998) Biochemistry 37, 59095915
177a. Uitdehaag, J. C. M., Mosi, R., Kalk, K. H., van
der Veen, B. A., Dijkhuizen, L., Withers, S. G.,
and Dijkstra, B. W. (1999) Nature Struct. Biol.
6, 432436
177b. van der Veen, B. A., Uitdehaag, J. C. M.,
Penninga, D., van Alebeek, G.-J. W. M., Smith,
L. M., Dijkstra, B. W., and Dijkhuizen, L.
(2000) J. Mol. Biol. 296, 10271038
178. Terada, Y., Yanase, M., Takata, H., Takaha, T.,
and Okada, S. (1997) J. Biol. Chem. 272, 15729
15733
179. Fierobe, H.-P., Clarke, A. J., Tull, D., and
Svensson, B. (1998) Biochemistry 37, 37533759
180. Aleshin, A. E., Stoffer, B., Firsov, L. M.,
Svensson, B., and Honzatko, R. B. (1996)
Biochemistry 35, 83198328
181. Firsov, L. M., Neustroev, K. N., Aleshin, A. E.,
Metzler, C. M., Metzler, D. E., Scott, R. D.,
Stoffer, B., Christensen, T., and Svensson, B.
(1994) Eur. J. Biochem. 223, 293302
182. Natarajan, S. K., and Sierks, M. R. (1997)
Biochemistry 36, 1494614955
183. Williamson, M. P., Le Gal-Coffet, M.-F.,
Sorimachi, K., Furniss, C. S. M., Archer, D. B.,
and Williamson, G. (1997) Biochemistry 36,
75357539
184. Penninga, D., van der Veen, B. A., Knegtel, R.
M. A., van Hijum, S. A. F. T., Rozeboom, H. J.,
Kalk, K. H., Dijkstra, B. W., and Dijkhuizen, L.
(1996) J. Biol. Chem. 271, 3277732784
184a. Mikami, B., Adachi, M., Kage, T., Sarikaya, E.,
Nanmori, T., Shinke, R., and Utsumi, S. (1999)
Biochemistry 38, 70507061
185. Mikami, B., Degano, M., Hehre, E. J., and
Sacchettini, J. C. (1994) Biochemistry 33, 7779
7787
186. Adachi, M., Mikami, B., Katsube, T., and Utsumi,
S. (1998) J. Biol. Chem. 273, 1985919865
187. Amado, M., Almeida, R., Carneiro, F., Levery,
S. B., Holmes, E. H., Nomoto, M., Hollings-
worth, M. A., Hassan, H., Schwientek, T.,
Nielsen, P. A., Bennett, E. P., and Clausen, H.
(1998) J. Biol. Chem. 273, 1277012778
188. Baenziger, J. U. (1994) FASEB J. 8, 10191025
189. Radzicka, A., and Wolfenden, R. (1996) J. Am.
Chem. Soc. 118, 61056109
190. Barrett, A. J., Rawlings, N. D., and Woessner,
J. F., eds. (1998) Handbook of Proteolytic
Enzymes, Academic Press, San Diego,
California
191. Fushiki, T., and Iwai, K. (1989) FASEB J. 3,
121126
192. Pignol, D., Granon, S., Chapus, C., and
Carlos, J. (1995) J. Mol. Biol. 252, 2024
References
666 Chapter 12. Transferring Groups by Displacement Reactions
242. Smith, S. O., Farr-Jones, S., Griffin, R. G., and
Bachovchin, W. W. (1989) Science 244, 961964
243. Kossiakoff, A. A., and Spencer, S. A. (1981)
Biochemistry 20, 64626474
244. Polgr, L. (1989) Mechanisms of Protease Action,
CRC Press, Boca Raton, Florida
245. Warshel, A., Naray-Szabo, G., Sussman, F.,
and Hwang, J.-K. (1989) Biochemistry 28,
36303637
246. Robillard, G., and Schulman, R. G. (1972) J.
Mol. Biol. 71, 507511
247. Markley, J. L., and Westler, W. M. (1996)
Biochemistry 35, 1109211097
248. Ash, E. L., Sudmeier, J. L., De Fabo, E. C.,
and Bachovchin, W. W. (1997) Science 278,
11281132
249. Halkides, C. J., Wu, Y. Q., and Murray, C. J.
(1996) Biochemistry 35, 1594115948
250. Zhong, S., Haghjoo, K., Kettner, C., and
Jordan, F. (1995) J. Am. Chem. Soc. 117, 7048
7055
251. Bachovchin, W. W., Wong, W. Y. L., Farr-Jones,
S., Shenvi, A. B., and Kettner, C. A. (1988)
Biochemistry 27, 76897697
252. Cassidy, C. S., Lin, J., and Frey, P. A. (1997)
Biochemistry 36, 45764584
253. Tsilikounas, E., Rao, T., Gutheil, W. G., and
Bachovchin, W. W. (1996) Biochemistry 35,
24372444
254. Tobin, J. B., Whitt, S. A., Cassidy, C. S., and
Frey, P. A. (1995) Biochemistry 34, 69196924
255. Singer, P. T., Smals, A., Carty, R. P., Mangel,
W. F., and Sweet, R. M. (1993) Science 259,
669673
256. Shieh, H.-S., Kurumbail, R. G., Stevens, A. M.,
Stegeman, R. A., Sturman, E. J., Pak, J. Y.,
Wittwer, A. J., Palmier, M. O., Wiegand, R. C.,
Holwerda, B. C., and Stallings, W. C. (1996)
Nature (London) 383, 279282
257. Tong, L., Quin, C., Massariol, M.-J., Bonneau,
P. R., Cordingley, M. G., and Lagac, L. (1996)
Nature (London) 383, 272275
258. Carter, P., and Wells, J. A. (1988) Nature
(London) 332, 564568
259. Corey, D. R., and Craik, C. S. (1992) J. Am.
Chem. Soc. 114, 17841790
260. Sprang, S., Standing, T., Fletterick, R. J.,
Stroud, R. M., Finer-Moore, J., Xuong, N.-H.,
Hamlin, R., Rutter, W. J., and Craik, C. S.
(1987) Science 237, 905909
261. McGrath, M. E., Vsquez, J. R., Craik, C. S.,
Yang, A. S., Honig, B., and Fletterick, R. J.
(1992) Biochemistry 31, 30593064
262. Robertus, J. D., Kraut, J., Alden, R. A., and
Birktoft, J. J. (1972) Biochemistry 11, 42934303
263. Whiting, A. K., and Peticolas, W. L. (1994)
Biochemistry 33, 552561
264. Bryan, P., Pantoliano, M. W., Quill, S. G.,
Hsiao, H.-Y., and Poulos, T. (1986) Proc. Natl.
Acad. Sci. U.S.A. 83, 37433745
265. Asbth, B., and Polgr, L. (1983) Biochemistry
22, 117122
266. Ortiz, C., Tellier, C., Williams, H., Stolowich,
N. J., and Scott, A. I. (1991) Biochemistry 30,
1002610034
267. Bullock, T. L., Breddam, K., and Remington,
S. J. (1996) J. Mol. Biol. 255, 714725
268. Tonge, P. J., and Carey, P. R. (1989) Biochemis-
try 28, 67016709
269. Tonge, P. J., and Carey, P. R. (1992) Biochemistry
31, 91229125
270. Birktoft, J. J., Kraut, J., and Freer, S. T. (1976)
Biochemistry 15, 44814485
271. Brnger, A. T., Huber, R., and Karplus, M.
(1987) Biochemistry 26, 5153 5162
272. Polgr, L., and Halsz, R. (1982) Biochem. J.
207, 110
273. Bizzozero, S. A., and Dutler, H. (1981) Bioorg.
Chem. 10, 4662
References
274. Cruickshank, W. H., and Kaplan, H. (1974) J.
Mol. Biol. 83, 267274
274a. Ash, E. L., Sudmeier, J. L., Day, R. M., Vincent,
M., Torchilin, E. V., Haddad, K. C., Bradshaw,
E. M., Sanford, D. G., and Bachovchin, W. W.
(2000) Proc. Natl. Acad. Sci. U.S.A. 97, 10371
10376
274b. Kidd, R. D., Sears, P., Huang, D.-H., Witte, K.,
Wong, C.-H., and Farber, G. K. (1999) Protein
Sci. 8, 410417
275. Meyer, E. F., Jr., Clore, G. M., Gronenborn, A.
M., and Hansen, H. A. S. (1988) Biochemistry
27, 725730
276. Wells, G. B., Mustafi, D., and Makinen, M. W.
(1994) J. Biol. Chem. 269, 45774586
277. Ding, X., Rasmussen, B. F., Petsko, G. A., and
Ringe, D. (1994) Biochemistry 33, 92859293
278. Oppenheimer, H. L., Labouesse, B., and Hess,
G. P. (1966) J. Biol. Chem. 241, 27202730
279. Fersht, A. (1985) Enzyme Structure and
Mechanism, 2nd ed., Freeman, San Francisco,
California
280. Perona, J. J., Hedstrom, L., Rutter, W. J., and
Fletterick, R. J. (1995) Biochemistry 34, 14891499
281. Graf, L., Craik, C. S., Patthy, A., Roczniak, S.,
Fletterick, R. J., and Rutter, W. J. (1987)
Biochemistry 26, 26162623
282. Schellenberger, V., Turck, C. W., and Rutter,
W. J. (1994) Biochemistry 33, 42514257
283. Kurth, T., Ullmann, D., Jakubke, H.-D., and
Hedstrom, L. (1997) Biochemistry 36, 10098
10104
284. Le Bonniec, B. F., Myles, T., Johnson, T.,
Knight, C. G., Tapparelli, C., and Stone, S. R.
(1996) Biochemistry 35, 71147122
285. Wangikar, P. P., Rich, J. O., Clark, D. S., and
Dordick, J. S. (1995) Biochemistry 34, 12302
12310
286. Perona, J. J., and Craik, C. S. (1995) Protein Sci.
4, 337360
287. Davis, J. H., and Agard, D. A. (1998)
Biochemistry 37, 76967707
288. Baumann, W. K., Bizzozero, S. A., and Dutler,
H. (1970) FEBS Letters 8, 257260
289. Baumann, W. K., Bizzozero, S. A., and Dutler,
H. (1973) Eur. J. Biochem. 39, 381387
290. Stein, R. L., Elrod, J. P., and Schowen, R. L.
(1983) J. Am. Chem. Soc. 105, 24462452
291. Chang, T. K., Chiang, Y., Guo, H.-X., Kresge,
A. J., Mathew, L., Powell, M. F., and Wells, J.
A. (1996) J. Am. Chem. Soc. 118, 88028807
292. Stein, R. L., and Stimpler, A. M. (1987) J. Am.
Chem. Soc. 109, 43874390
293. Baker, E. N. (1980) J. Mol. Biol. 141, 441484
294. Maes, D., Bouckaert, J., Poortmans, F., Wyns,
L., and Looze, Y. (1996) Biochemistry 35,
1629216298
295. OHara, B. P., Hemmings, A. M., Buttle, D. J.,
and Pearl, L. H. (1995) Biochemistry 34,
1319013195
296. Berti, P. J., and Storer, A. C. (1995) J. Mol. Biol.
246, 273283
297. Drenth, J., Jansonius, J. N., Koekoek, R., and
Wolthers, B. G. (1971) Adv. Prot. Chem. 25,
79115
298. Lewis, S. D., Johnson, F. A., and Shafer, J. A.
(1981) Biochemistry 20, 4851
299. Roberts, D. D., Lewis, S. D., Ballou, D. P.,
Olson, S. T., and Shafer, J. A. (1986)
Biochemistry 25, 55955601
300. Johnson, F. A., Lewis, S. D., and Shafer, J. A.
(1981) Biochemistry 20, 4448
301. Keillor, J. W., and Brown, R. S. (1992) J. Am.
Chem. Soc. 114, 79837989
302. Pinitglang, S., Watts, A. B., Patel, M., Reid, J.
D., Noble, M. A., Gul, S., Bokth, A., Naeem,
A., Patel, H., Thomas, E. W., Sreedharan, S.
K., Verma, C., and Brocklehurst, K. (1997)
Biochemistry 36, 99689982
303. Mnard, R., Khouri, H. E., Plouffe, C.,
Laflamme, P., Dupras, R., Vernet, T., Tessier,
D. C., Thomas, D. Y., and Storer, A. C. (1991)
Biochemistry 30, 55315538
304. Katerelos, N. A., and Goodenough, P. W.
(1996) Biochemistry 35, 1476314772
305. Vernet, T., Tessier, D. C., Chatellier, J., Plouffe,
C., Lee, T. S., Thomas, D. Y., Storer, A. C., and
Mnard, R. (1995) J. Biol. Chem. 270, 16645
16652
306. Mnard, R., Carrire, J., Laflamme, P., Plouffe,
C., Khouri, H. E., Vernet, T., Tessier, D. C.,
Thomas, D. Y., and Storer, A. C. (1991)
Biochemistry 30, 89248928
307. Mnard, R., Laflamme, P., Plouffe, C., Vernet,
T., Tessier, D. C., Thomas, D. Y., and Storer, A.
C. (1995) Biochemistry 34, 464471
308. Doran, J. D., Tonge, P. J., Mort, J. S., and Carey,
P. R. (1996) Biochemistry 35, 1248712494
309. Gamcsik, M. P., Malthouse, J. P. G., Primrose,
W. U., Mackenzie, N. E., Boyd, A. S. F.,
Russell, R. A., and Scott, A. I. (1983) J. Am.
Chem. Soc. 105, 63246325
310. Zheng, Y.-J., and Bruice, T. C. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 42854288
311. Arad, D., Langridge, R., and Kollman, P. A.
(1990) J. Am. Chem. Soc. 112, 491502
312. Brocklehurst, K., Watts, A. B., Patel, M.,
Verma, C., and Thomas, E. W. (1998) in
Comprehensive Biological Catalysis. A
Mechanistic Reference, Vol. I (Sinnott, M., ed),
Academic Press, San Diego, California
313. Bond, J. S., and Butler, P. E. (1987) Ann. Rev.
Biochem. 56, 333364
314. Takahashi, T., Dehdarani, A. H., and Tang, J.
(1988) J. Biol. Chem. 263, 1095210957
315. Coulombe, R., Grochulski, P., Sivaraman, J.,
Mnard, R., Mort, J. S., and Cygler, M. (1996)
EMBO J. 15, 54925503
316. Jia, Z., Hasnain, S., Hirama, T., Lee, X., Mort,
J. S., To, R., and Huber, C. P. (1995) J. Biol.
Chem. 270, 55275533
317. Taralp, A., Kaplan, H., Sytwu, I.-I., Vlattas, I.,
Bohacek, R., Knap, A. K., Hirama, T., Huber,
C. P., and Hasnain, S. (1995)
J. Biol. Chem. 270, 1803618043
318. McGrath, M. E., Palmer, J. T., Brmme, D., and
Somoza, J. R. (1998) Protein Sci. 7, 12941302
319. Shi, G.-P., Webb, A. C., Foster, K. E., Knoll, J.
H. M., Lemere, C. A., Munger, J. S., and
Chapman, H. A. (1994) J. Biol. Chem. 269,
1153011536
320. Gelb, B. D., Shi, G.-P., Chapman, H. A., and
Desnick, R. J. (1996) Science 273, 12361238
321. Ishidoh, K., Muno, D., Sato, N., and Kominami,
E. (1991) J. Biol. Chem. 266, 1631216317
322. Wolters, P. J., Raymond, W. W., Blount, J. L.,
and Caughey, G. H. (1998) J. Biol. Chem. 273,
1551415520
323. Schiller, M. R., Mende-Mueller, L., Moran, K.,
Meng, M., Miller, K. W., and Hook, V. Y. H.
(1995) Biochemistry 34, 79887995
324. Friedman, T. C., Kline, T. B., and Wilk, S.
(1985) Biochemistry 24, 39073913
325. Matsui, S.-I., Sandberg, A. A., Negoro, S.,
Seon, B. K., and Goldstein, G. (1982) Proc.
Natl. Acad. Sci. U.S.A. 79, 15351539
326. Joshua-Tor, L., Xu, H. E., Johnston, S. A., and
Rees, D. C. (1995) Science 269, 945950
327. Richards, F. M. (1991) Sci. Am. 264 (Jan), 5463
328. Ohno, S., Emori, Y., Imajoh, S., Kawasaki, H.,
Kisaragi, M., and Suzuki, K. (1984) Nature
(London) 312, 566570
329. Saido, T. C., Sorimachi, H., and Suzuki, K.
(1994) FASEB J. 8, 814822
330. Chian, H.-L., and Dice, J. F. (1988) J. Biol.
Chem. 263, 67976805
331. Tompa, P., Baki, A., Schd, ., and Friedrich,
P. (1996) J. Biol. Chem. 271, 3316133164
667
385. Blundell, T. L., Lapatto, R., Wilderspin, A. F.,
Hemmings, A. M., Hobart, P. M., Danley, D.
E., and Whittle, P. J. (1990) Trends Biochem. Sci.
15, 425430
386. Katz, R. A., and Skalka, A. M. (1994) Ann. Rev.
Biochem. 63, 133173
387. Wlodawer, A., Miller, M., Jasklski, M.,
Sathyanarayana, B. K., Baldwin, E., Weber, I.
T., Selk, L. M., Clawson, L., Schneider, J., and
Kent, S. B. H. (1989) Science 245, 616620
388. Rose, R. B., Craik, C. S., Douglas, N. L., and
Stroud, R. M. (1996) Biochemistry 35, 1293312944
389. Chatfield, D. C., and Brooks, B. R. (1995) J.
Am. Chem. Soc. 117, 55615572
390. Rodriguez, E. J., Angeles, T. S., and Meek, T.
D. (1993) Biochemistry 32, 1238012385
391. Liu, H., Mller-Plathe, F., and van Gunsteren,
W. F. (1996) J. Mol. Biol. 261, 454469
392. Silva, A. M., Cachau, R. E., Sham, H. L., and
Erickson, J. W. (1996) J. Mol. Biol. 255, 321346
393. Reverter, D., Ventura, S., Villegas, V., Vendrell,
J., and Avils, F. X. (1998) J. Biol. Chem. 273,
35353541
394. Varlamov, O., Leiter, E. H., and Fricker, L.
(1996) J. Biol. Chem. 271, 1398113986
395. Rodrguez, C., Brayton, K. A., Brownstein, M.,
and Dixon, J. E. (1989) J. Biol. Chem. 264,
59885995
396. Tan, F., Chan, S. J., Steiner, D. F., Schilling, J.
W., and Skidgel, R. A. (1989) J. Biol. Chem. 264,
1316513170
397. Tan, F., Weerasinghe, D. K., Skidgel, R. A.,
Tamei, H., Kaul, R. K., Roninson, I. B.,
Schilling, J. W., and Erds, E. G. (1990) J. Biol.
Chem. 265, 1319
398. Ehlers, M. R. W., and Riordan, J. F. (1989)
Biochemistry 28, 53115318
399. Dideberg, O., Charlier, P., Dive, G., Joris, B.,
Frere, J. M., and Ghuysen, J. M. (1982) Nature
(London) 299, 469470
400. Weaver, L. H., Kester, W. R., and Matthews, B.
W. (1977) J. Mol. Biol. 114, 119132
401. Hausrath, A. C., and Matthews, B. W. (1994) J.
Biol. Chem. 269, 1883918842
402. Holland, D. R., Hausrath, A. C., Juers, D., and
Matthews, B. W. (1995) Protein Sci. 4, 19551965
403. Holden, H. M., Tronrud, D. E., Monzingo, A.
F., Weaver, L. H., and Matthews, B. W. (1987)
Biochemistry 26, 85428553
404. Mock, W. L., and Stanford, D. J. (1996)
Biochemistry 35, 73697377
405. Marie-Claire, C., Ruffet, E., Antonczak, S.,
Beaumont, A., ODonohue, M., Roques, B. P.,
and Fourni-Zaluski, M.-C. (1997) Biochemis-
try 36, 1393813945
406. Lipman, M. L., Panda, D., Bennett, H. P. J.,
Henderson, J. E., Shane, E., Shen, Y.,
Goltzman, D., and Karaplis, A. C. (1998) J.
Biol. Chem. 273, 1372913737
407. Ogishima, T., Niidome, T., Shimokata, K.,
Kitada, S., and Ito, A. (1995) J. Biol. Chem. 270,
3032230326
408. Luciano, P., Tokatlidis, K., Chambre, I.,
Germanique, J.-C., and Gli, V. (1998) J. Mol.
Biol. 280, 193199
409. Christianson, D. W., and Lipscomb, W. N. (1986)
Proc. Natl. Acad. Sci. U.S.A. 83, 75687572
410. Mock, W. L. (1998) in Comprehensive Biological
Catalysis. A Mechanistic Reference, Vol. I
(Sinnott, M., ed), pp. 425453, Academic
Press, San Diego, California
411. Campbell, P., and Nashed, N. T. (1982) J. Am.
Chem. Soc. 104, 52215226
412. Makinen, M. W., Fukuyama, J. M., and Kuo,
L. C. (1982) J. Am. Chem. Soc. 104, 26672669
413. Sander, M. E., and Witzel, H. (1985) Biochem.
Biophys. Res. Commun. 132, 681687
414. Kuo, L. C., Fukuyama, J. M., and Makinen, M.
W. (1983) J. Mol. Biol. 163, 63105
332. Stabach, P. R., Cianci, C. D., Glantz, S. B.,
Zhang, Z., and Morrow, J. S. (1997) Biochemistry
36, 5765
332a. Hosfield, C. M., Elce, J. S., Davies, P. L., and
Jia, Z. (1999) EMBO J. 18, 68806889
333. Nicholson, D. W., and Thornberry, N. A.
(1997) Trends Biochem. Sci. 22, 299306
334. Villa, P., Kaufmann, S. H., and Earnshaw, W.
C. (1997) Trends Biochem. Sci. 22, 388393
335. Munday, N. A., Vaillancourt, J. P., Ali, A.,
Casano, F. J., Miller, D. K., Molineaux, S. M.,
Yamin, T.-T., Yu, V. L., and Nicholson, D. W.
(1995) J. Biol. Chem. 270, 1587015876
336. McGrath, M. E., Eakin, A. E., Engel, J. C.,
McKerrow, J. H., Craik, C. S., and Fletterick,
R. J. (1995) J. Mol. Biol. 247, 251259
337. Gillmor, S. A., Craik, C. S., and Fletterick, R. J.
(1997) Protein Sci. 6, 16031611
338. Porter, W. H., Cunningham, L. W., and Mitchell,
W. M. (1971) J. Biol. Chem. 246, 76757682
339. Pavloff, N., Pemberton, P. A., Potempa, J.,
Chen, W.-C. A., Pike, R. N., Prochazka, V.,
Kiefer, M. C., Travis, J., and Barr, P. J. (1997)
J. Biol. Chem. 272, 15951600
339a. Nelson, D., Potempa, J., Kordula, T., and Travis,
J. (1999) J. Biol. Chem. 274, 1224512251
340. Malcolm, B. A. (1995) Protein Sci. 4, 14391445
341. Mosimann, S. C., Cherney, M. M., Sia, S.,
Plotch, S., and James, M. N. G. (1997) J. Mol.
Biol. 273, 10321047
342. Ding, J., McGrath, W. J., Sweet, R. M., and
Mangel, W. F. (1996) EMBO J. 15, 17781783
343. Fox, T., de Miguel, E., Mort, J. S., and Storer,
A. C. (1992) Biochemistry 31, 1257112576
344. Podobnik, M., Kuhelj, R., Turk, V., and Turk,
D. (1997) J. Mol. Biol. 271, 774788
345. Seemller, E., Lupas, A., Stock, D., Lwe, J.,
Huber, R., and Baumeister, W. (1995) Science
268, 579582
346. Paetzel, M., and Dalbey, R. E. (1997) Trends
Biochem. Sci. 22, 2831
346a. Kisselev, A. F., Songyang, Z., and Goldberg,
A. L. (2000) J. Biol. Chem. 275, 1483114837
347. Eleuteri, A. M., Kohanski, R. A., Cardozo, C.,
and Orlowski, M. (1997) J. Biol. Chem. 272,
1182411831
348. Kopp, F., Kristensen, P., Hendil, K. B.,
Johnsen, A., Sobek, A., and Dahlmann, B.
(1995) J. Mol. Biol. 248, 264272
349. Cardozo, C., Michaud, C., and Orlowski, M.
(1999) Biochemistry 38, 97689777
349a. Groll, M., Heinemeyer, W., Jger, S., Ullrich,
T., Bochtler, M., Wolf, D. H., and Huber, R.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 10976
10983
349b. Witt, E., Zantopf, D., Schmidt, M., Kraft, R.,
Kloetzel, P.-M., and Krger, E. (2000) J. Mol.
Biol. 301, 19
349c. Orlowski, M., and Wilk, S. (2000) Arch.
Biochem. Biophys. 383, 116
350. Tikkanen, R., Riikonen, A., Oinonen, C.,
Rouvinen, J., and Peltonen, L. (1996) EMBO J.
15, 29542960
351. Xuan, J., Tarentino, A. L., Grimwood, B. G.,
Plummer, T. H., Jr., Cui, T., Guan, C., and Van
Roey, P. (1998) Protein Sci. 7, 774781
352. Liu, Y., Guan, C., and Aronson, N. N., Jr.
(1998) J. Biol. Chem. 273, 96889694
353. Guan, C., Liu, Y., Shao, Y., Cui, T., Liao, W.,
Ewel, A., and Whitaker, R. (1998) J. Biol. Chem.
273, 96959702
354. Brannigan, J. A., Dodson, G., Duggleby, H. J.,
Moody, P. C. E., Smith, J. L., Tomchick, D. R.,
and Murzin, A. G. (1995) Nature (London) 378,
416419
355. Dalbey, R. E., and Wickner, W. (1988) J. Biol.
Chem. 263, 404408
356. Tschantz, W. R., Sung, M., Delgado-Partin, V.
M., and Dalbey, R. E. (1993) J. Biol. Chem. 268,
2734927354
357. Dalbey, R. E., Lively, M. O., Bron, S., and Van
Dijl, J. M. (1997) Protein Sci. 6, 11291138
358. Fenteany, G., and Schreiber, S. L. (1998) J. Biol.
Chem. 273, 85458548
359. Bogyo, M., McMaster, J. S., Gaczynska, M.,
Tortorella, D., Goldberg, A. L., and Ploegh, H.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 66296634
360. Schmidtke, G., Kraft, R., Kostka, S., Henklein,
P., Frmmel, C., Lwe, J., Huber, R., Kloetzel,
P. M., and Schmidt, M. (1996) EMBO J. 15,
68876898
361. Heinemeyer, W., Fischer, M., Krimmer, T.,
Stachon, U., and Wolf, D. H. (1997) J. Biol.
Chem. 272, 2520025209
362. Ditzel, L., Huber, R., Mann, K., Heinemeyer,
W., Wolf, D. H., and Groll, M. (1998) J. Mol.
Biol. 279, 11871191
363. Andreeva, N. S., Zdanov, A. S., Gustchina, A.
E., and Fedorov, A. A. (1984) J. Biol. Chem. 259,
1135311365
364. Fusek, M., and Vetvicka, V., eds. (1995)
Aspartic Proteinases: Physiology and Pathology,
CRC Press, Boca Raton, Florida
365. Moore, S. A., Sielecki, A. R., Chernaia, M. M.,
Tarasova, N. I., and James, M. N. G. (1995) J.
Mol. Biol. 247, 466485
366. Strop, P., Sedlacek, J., Stys, J., Kaderabkova,
Z., Blaha, I., Pavlickova, L., Pohl, J., Fabry, M.,
Kostka, V., Newman, M., Frazao, C., Shearer,
A., Tickle, I. J., and Blundell, T. L. (1990)
Biochemistry 29, 98639871
367. Vance, J. E., LeBlanc, D. A., and London, R. E.
(1997) Biochemistry 36, 1323213240
368. Majer, P., Collins, J. R., Gulnik, S. V., and
Erickson, J. W. (1997) Protein Sci. 6, 14581466
369. Krieger, T. J., and Hook, V. Y. H. (1992)
Biochemistry 31, 42234231
370. Fineschi, B., and Miller, J. (1997) Trends
Biochem. Sci. 22, 377382
371. Wilk, S., Wilk, E., and Magnusson, R. P. (1998)
J. Biol. Chem. 273, 1596115970
372. Tong, L., Pav, S., Lamarre, D., Pilote, L.,
LaPlante, S., Anderson, P. C., and Jung, G.
(1995) J. Mol. Biol. 250, 211222
373. Yang, J., Teplyakov, A., and Quail, J. W. (1997)
J. Mol. Biol. 268, 449459
374. Gmez, J., and Freire, E. (1995) J. Mol. Biol.
252, 337350
375. Symersky, J., Monod, M., and Foundling, S. I.
(1997) Biochemistry 36, 1270012710
376. Green, D. W., Aykent, S., Gierse, J. K., and
Zupec, M. E. (1990) Biochemistry 29, 31263133
377. Baldwin, E. T., Bhat, T. N., Gulnik, S., Hosur,
M. V., Sowder, R. C., Cachau, R. E., Collins, J.,
Silva, A. M., and Erickson, J. W. (1993) Proc.
Natl. Acad. Sci. U.S.A. 90, 67966800
378. Goldblum, A. (1988) Biochemistry 27, 16531658
379. Subramanian, E. (1978) Trends Biochem. Sci. 3,
13
380. Andreeva, N. S., Zdanov, A. S., Gustchina, A.
E., and Fedorev, A. A. (1984) J. Biol. Chem. 259,
1135311365
381. James, M. N. G., Sielecki, A. R., Hayakawa,
K., and Gelb, M. H. (1992) Biochemistry 31,
38723886
382. Meek, T. D. (1998) in Comprehensive Biological
Catalysis. A Mechanistic Reference, Vol. I
(Sinnott, M., ed), pp. 327344, Academic
Press, San Diego, California
383. Blum, M., Cunningham, A., Pang, H., and
Hofmann, T. (1991) J. Biol. Chem. 266, 9501
9507
384. James, M. N. G., and Sielecki, A. R. (1986)
Nature (London) 319, 3338
References
668 Chapter 12. Transferring Groups by Displacement Reactions
415. Suh, J., Hong, S.-B., and Chung, S. (1986) J.
Biol. Chem. 261, 71127114
416. Mustafi, D., and Makinen, M. W. (1994) J. Biol.
Chem. 269, 45874595
417. Kuo, L. C., and Makinen, M. W. (1985) J. Am.
Chem. Soc. 107, 52555261
418. Cleland, W. W. (1977) Adv. Enzymol. 45, 273286
419. Kim, H., and Lipscomb, W. N. (1990)
Biochemistry 29, 55465555
420. Osumi, A., Rahmo, A., King, S. W., Przystas,
T. J., and Fife, T. H. (1994) Biochemistry 33,
1475014757
421. Mock, W. L., and Zhang, J. Z. (1991) J. Biol.
Chem. 266, 63936400
422. Zhang, K., and Auld, D. S. (1995) Biochemistry
34, 1630616312
423. Hilver, D., Gardell, S. J., Rutter, W. J., and
Kaiser, E. T. (1986) J. Am. Chem. Soc. 108,
52985304
424. Kam, C.-M., Nishino, N., and Powers, J. C.
(1979) Biochemistry 18, 30323038
425. Bartlett, P. A., and Marlowe, C. K. (1983)
Biochemistry 22, 46184624
426. Holden, H. M., and Matthews, B. W. (1988) J.
Biol. Chem. 263, 32563260
427. Massova, I., Kotra, L. P., Fridman, R., and
Mobashery, S. (1998) FASEB J. 12, 10751095
428. Bond, J. S., and Beynon, R. J. (1995) Protein Sci.
4, 12471261
429. Stcker, W., Grams, F., Baumann, U.,
Reinemer, P., Gomis-Rth, F.-X., McKay, D. B.,
and Bode, W. (1995) Protein Sci. 4, 823840
429a. Morgunova, E., Tuuttila, A., Bergmann, U.,
Isupov, M., Lindqvist, Y., Schneider, G., and
Tryggvason, K. (1999) Science 284, 16671670
429b. Tortorella, M. D., and 27 other authors. (1999)
Science 284, 16641666
429c. Nagase, H., and Woessner, J. F., Jr. (1999) J.
Biol. Chem. 274, 2149121494
430. Becker, J. W., Marcy, A. I., Rokosz, L. L., Axel,
M. G., Burbaum, J. J., Fitzgerald, P. M. D.,
Cameron, P. M., Esser, C. K., Hagmann, W. K.,
Hermes, J. D., and Springer, J. P. (1995) Protein
Sci. 4, 19661976
431. Wetmore, D. R., and Hardman, K. D. (1996)
Biochemistry 35, 65496558
432. Arumugam, S., Hemme, C. L., Yoshida, N.,
Suzuki, K., Nagase, H., Berjanskii, M., Wu, B.,
and Van Doren, S. R. (1998) Biochemistry 37,
96509657
433. Gomis-Rth, F. X., Stcker, W., Huber, R.,
Zwilling, R., and Bode, W. (1993) J. Mol. Biol.
229, 945968
434. Gomis-Rth, F. X., Kress, L. F., Kellermann, J.,
Mayr, I., Lee, X., Huber, R., and Bode, W.
(1994) J. Mol. Biol. 239, 513544
435. Llano, E., Pends, A. M., Knuper, V., Sorsa,
T., Salo, T., Salido, E., Murphy, G., Simmer, J.
P., Bartlett, J. D., and Lpez-Otn, C. (1997)
Biochemistry 36, 1510115108
436. Gomis-Rth, F. X., Gohlke, U., Betz, M.,
Knuper, V., Murphy, G., Lpez-Otn, C., and
Bode, W. (1996) J. Mol. Biol. 264, 556566
437. Taylor, A. (1993) FASEB J. 7, 290298
438. Strter, N., and Lipscomb, W. N. (1995)
Biochemistry 34, 1479214800
438a. Strter, N., Sun, L., Kantrowitz, E. R., and
Lipscomb, W. N. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 1115111155
439. Chen, G., Edwards, T., Dsouza, V. M., and
Holz, R. C. (1997) Biochemistry 36, 42784286
440. Lowther, W. T., Orville, A. M., Madden, D. T.,
Lim, S., Rich, D. H., and Matthews, B. W.
(1999) Biochemistry 38, 76787688
441. Bradshaw, R. A., Brickey, W. W., and Walker,
K. W. (1998) Trends Biochem. Sci. 23, 263267
References
442. Wilce, M. C. J., Bond, C. S., Dixon, N. E.,
Freeman, H. C., Guss, J. M., Lilley, P. E., and
Wilce, J. A. (1998) Proc. Natl. Acad. Sci. U.S.A.
95, 34723477
443. Chan, M. K., Gong, W., Rajagopalan, P. T. R.,
Hao, B., Tsai, C. M., and Pei, D. (1997)
Biochemistry 36, 1390413909
444. Dardel, F., Ragusa, S., Lazennec, C., Blanquet,
S., and Meinnel, T. (1998) J. Mol. Biol. 280,
501513
445. Becker, A., Schlichting, I., Kabsch, W., Schultz,
S., and Wagner, A. F. V. (1998) J. Biol. Chem.
273, 1141311416
446. Andrews, D. W., and Johnson, A. E. (1996)
Trends Biochem. Sci. 21, 365369
446a. Fabunmi, R. P., Wigley, W. C., Thomas, P. J.,
and DeMartino, G. N. (2000) J. Biol. Chem. 275,
409413
447. Sommer, T., and Wolf, D. H. (1997) FASEB J.
11, 12271233
448. Ciechanover, A., and Schwartz, A. L. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 27272730
449. Rubin, D. M., Glickman, M. H., Larsen, C. N.,
Dhruvakumar, S., and Finley, D. (1998) EMBO
J. 17, 49094919
450. Lee, Y. S., Park, S. C., Goldberg, A. L., and
Chung, C. H. (1988) J. Biol. Chem. 263, 6643
6646
451. Shin, D. H., Lee, C. S., Chung, C. H., and Suh,
S. W. (1996) J. Mol. Biol. 262, 7176
452. Chin, D. T., Goff, S. A., Webster, T., Smith, T.,
and Goldberg, A. L. (1988) J. Biol. Chem. 263,
1171811728
453. Flanagan, J. M., Wall, J. S., Capel, M. S.,
Schneider, D. K., and Shanklin, J. (1995)
Biochemistry 34, 1091010917
454. Kessel, M., Maurizi, M. R., Kim, B., Kocsis, E.,
Trus, B. L., Singh, S. K., and Steven, A. C.
(1995) J. Mol. Biol. 250, 587594
455. Suzuki, C. K., Rep, M., Maarten van Dijl, J.,
Suda, K., Grivell, L. A., and Schatz, G. (1997)
Trends Biochem. Sci. 22, 118123
456. Roudiak, S. G., and Shrader, T. E. (1998)
Biochemistry 37, 1125511263
457. Wang, N., Maurizi, M. R., Emmert-Buck, L.,
and Gottesman, M. M. (1994) J. Biol. Chem.
269, 2930829313
458. Savelev, A. S., Novikova, L. A., Kovaleva, I.
E., Luzikov, V. N., Neupert, W., and Langer, T.
(1998) J. Biol. Chem. 273, 2059620602
459. Singh, S. K., Guo, F., and Maurizi, M. R.
(1999) Biochemistry 38, 1490614915
460. Kihara, A., Akiyama, Y., and Ito, K. (1998) J.
Mol. Biol. 279, 175188
461. Lupas, A., Flanagan, J. M., Tamura, T., and
Baumeister, W. (1997) Trends Biochem. Sci. 22,
399404
461a. de Sagarra, M. R., Mayo, I., Marco, S.,
Rodrguez-Vilarino, S., Oliva, J., Carrascosa, J.
L., and Castano, J. G. (1999) J. Mol. Biol. 292,
819825
461b. Kim, K. I., Cheong, G.-W., Park, S.-C., Ha, J.-
S., Woo, K. M., Choi, S. J., and Chung, C. H.
(2000) J. Mol. Biol. 303, 655-666
462. Rohrwild, M., Coux, O., Huang, H.-C.,
Moerschell, R. P., Yoo, S. J., Seol, J. H., Chung,
C. H., and Goldberg, A. L. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 58085813
463. Missiakas, D., Schwager, F., Betton, J.-M.,
Georgopoulos, C., and Raina, S. (1996) EMBO
J. 15, 68996909
463a. Bochtler, M., Hartmann, C., Song, H. K.,
Bourenkov, G. P., Bartunik, H. D., and Huber,
R. (2000) Nature (London) 403, 800805
463b. Shotland, Y., Teff, D., Koby, S., Kobiler, O.,
and Oppenheim, A. B. (2000) J. Mol. Biol. 299,
953964
464. Krusslich, H.-G., and Wimmer, E. (1988)
Ann. Rev. Biochem. 57, 701754
465. Ypma-Wong, M. F., Filman, D. J., Hogle, J. M.,
and Semler, B. L. (1988) J. Biol. Chem. 263,
1784617856
466. Lpez-Otn, C., Simn-Mateo, C., Martnez,
L., and Vinuela, E. (1989) J. Biol. Chem. 264,
91079110
466a. Wu, Z., Yao, N., Le, H. V., and Weber, P. C.
(1998) Trends Biochem. Sci. 23, 9293
466b. Barbato, G., Cicero, D. O., Cordier, F., Narjes,
F., Gerlach, B., Sambucini, S., Grzesiek, S.,
Matassa, V. G., De Francesco, R., and Bazzo,
R. (2000) EMBO J. 19, 11951206
467. Roseman, J. E., and Levine, R. L. (1987) J. Biol.
Chem. 262, 21012110
468. Grune, T., Reinheckel, T., and Davies, K. J. A.
(1997) FASEB J. 11, 526534
469. Erds, E. G., and Skidgel, R. A. (1989) FASEB
J. 3, 145151
470. Hui, K.-S. (1988) J. Biol. Chem. 263, 66136618
471. Zisfein, J. B., Graham, R. M., Dreskin, S. V.,
Wildey, G. M., Fischman, A. J., and Honey, C.
J. (1987) Biochemistry 26, 86908697
471a. Loew, D., Perrault, C., Morales, M., Moog, S.,
Ravanat, C., Schuhler, S., Arcone, R., Pietropa-
olo, C., Cazenave, J.-P., van Dorsselaer, A.,
and Lanza, F. (2000) Biochemistry 39, 10812
10822
472. Lobe, C. G., Finlay, B. B., Paranchych, W.,
Paetkau, V. H., and Bleackley, R. C. (1986)
Science 232, 858861
473. Melloni, E., Pontremoki, S., Salamino, F.,
Sparatore, B., Michetti, M., Sacco, O., and
Horecker, B. L. (1986) J. Biol. Chem. 261,
1143711439
474. Pohlner, J., Halter, R., Beyreuther, K., and
Meyer, T. F. (1987) Nature (London) 325, 458462
475. Newport, G. R., McKerrow, J. H., Hedstrom,
R., Petitt, M., McGarrigle, L., Barr, D. J., and
Agabian, N. (1988) J. Biol. Chem. 263, 13179
13184
476. Folk, J. E. (1980) Ann. Rev. Biochem. 49, 517531
477. McGrath, M. E., Gillmor, S. A., and Fletterick,
R. J. (1995) Protein Sci. 4, 141 148
478. Salier, J.-P. (1990) Trends Biochem. Sci. 15, 435
439
479. Laskowski, M., Jr., and Kato, I. (1980) Ann.
Rev. Biochem. 49, 593626
480. Barrett, A. J. (1987) Trends Biochem. Sci. 12,
193196
481. Freije, J. P., Balbn, M., Abrahamson, M.,
Velasco, G., Dalboge, H., Grubb, A., and
Lpez-Otn, C. (1993) J. Biol. Chem. 268,
1573715744
482. Martin, J. R., Craven, C. J., Jerala, R., Kroon-
Zitko, L., Zerovnik, E., Turk, V., and Waltho, J.
P. (1995) J. Mol. Biol. 246, 331343
483. Brown, W. M., and Dziegielewska, K. M.
(1997) Protein Sci. 6, 512
484. Williamson, R. A., Carr, M. D., Frenkiel, T. A.,
Feeney, J., and Freedman, R. B. (1997)
Biochemistry 36, 1388213889
485. Apte, S. S., Olsen, B. R., and Murphy, G.
(1995) J. Biol. Chem. 270, 1431314318
486. Baumann, U., Bauer, M., Ltoff, S.,
Delepelaire, P., and Wandersman, C. (1995)
J. Mol. Biol. 248, 653661
487. Martzen, M. R., McMullen, B. A., Smith, N. E.,
Fujikawa, K., and Peanasky, R. J. (1990)
Biochemistry 29, 73667372
488. Huang, K., Lu, W., Anderson, S., Laskowski,
M., Jr., and James, M. N. G. (1995) Protein Sci.
4, 19851997
489. Lu, W., Qasim, M. A., and Kent, S. B. H. (1996)
J. Am. Chem. Soc. 118, 85188523
490. Huber, R., Kukla, D., Bode, W., Schwager, P.,
Bartels, K., Diesenhofer, J., and Steigemann,
W. (1974) J. Mol. Biol. 89, 73101
669
543. Everse, S. J., Spraggon, G., Veerapandian, L.,
Riley, M., and Doolittle, R. F. (1998)
Biochemistry 37, 86378642
544. Spraggon, G., Everse, S. J., and Doolittle, R. F.
(1997) Nature (London) 389, 455462
545. Murthy, S. N. P., Wilson, J. H., Lukas, T. J.,
Veklich, Y., Weisel, J. W., and Lorand, L. (2000)
Proc. Natl. Acad. Sci. U.S.A. 97, 4448
546. Pedersen, L. C., Yee, V. C., Bishop, P. D., Le
Trong, I., Teller, D. C., and Stenkamp, R. E.
(1994) Protein Sci. 3, 11311135
547. Bernardo, M. M., Day, D. E., Olson, S. T., and
Shore, J. D. (1993) J. Biol. Chem. 268, 12468
12476
548. Beaubien, G., Rosinski-Chupin, I., Mattei, M.
G., Mbikay, M., Chrtien, M., and Seidah, N.
G. (1991) Biochemistry 30, 16281635
549. Herwald, H., Renn, T., Meijers, J. C. M.,
Chung, D. W., Page, J. D., Colman, R. W., and
Mller-Esterl, W. (1996) J. Biol. Chem. 271,
1306113067
550. Baglia, F. A., Jameson, B. A., and Walsh, P. N.
(1993) J. Biol. Chem. 268, 38383844
551. Hamaguchi, N., Charifson, P. S., Pedersen, L.
G., Brayer, G. D., Smith, K. J., and Stafford, D.
W. (1991) J. Biol. Chem. 266, 1521315220
552. Kurachi, S., Hitomi, Y., Furukawa, M., and
Kurachi, K. (1995) J. Biol. Chem. 270, 52765281
552a. Kurachi, S., Deyashiki, Y., Takeshita, J., and
Kurachi, K. (1999) Science 285, 739743
553. Freedman, S. J., Furie, B. C., Furie, B., and
Baleja, J. D. (1995) Biochemistry 34, 1212612137
554. Freedman, S. J., Blostein, M. D., Baleja, J. D.,
Jacobs, M., Furie, B. C., and Furie, B. (1996) J.
Biol. Chem. 271, 1622716236
555. Lenting, P. J., Christophe, O. D., ter Maat, H.,
Rees, D. J. G., and Mertens, K. (1996) J. Biol.
Chem. 271, 2533225337
556. Tagliavacca, L., Moon, N., Dunham, W. R.,
and Kaufman, R. J. (1997) J. Biol. Chem. 272,
2742827434
557. Pittman, D. D., Wang, J. H., and Kaufman, R.
J. (1992) Biochemistry 31, 33153325
558. Gilbert, G. E., and Baleja, J. D. (1995)
Biochemistry 34, 30223031
559. Gilbert, G. E., and Drinkwater, D. (1993)
Biochemistry 32, 95779585
560. Leyte, A., van Schijndel, H. B., Niehrs, C.,
Huttner, W. B., Verbeet, M. P., Mertens, K.,
and van Mourik, J. A. (1991) J. Biol. Chem. 266,
740746
561. Gitschier, J., Wood, W. I., Shuman, M. A., and
Lawn, R. M. (1986) Science 232, 1415 1416
562. Titani, K., and Walsh, K. A. (1988) Trends
Biochem. Sci. 13, 9497
563. Sadler, J. E. (1998) Ann. Rev. Biochem. 67, 395
424
564. Ruggeri, Z. M., and Ware, J. (1993) FASEB J. 7,
308316
565. Huizinga, E. G., van der Plas, R. M., Kroon, J.,
Sixma, J. J., and Gros, P. (1997) Structure 5,
11471156
566. Emsley, J., Cruz, M., Handin, R., and
Liddington, R. (1998) J. Biol. Chem. 273,
1039610401
567. Sugimoto, M., Dent, J., McClintock, R., Ware,
J., and Ruggeri, Z. M. (1993) J. Biol. Chem. 268,
1218512192
568. George, J. N., Nurden, A. T., and Phillips, D.
R. (1984) N. Engl. J. Med. 311, 10841098
569. Cruz, M. A., Yuan, H., Lee, J. R., Wise, R. J.,
and Handin, R. I. (1995) J. Biol. Chem. 270,
1082210827
570. Beacham, D. A., Wise, R. J., Turci, S. M., and
Handin, R. I. (1992) J. Biol. Chem. 267, 3409
3415
571. Zhang, Z. P., Blombck, M., Nyman, D., and
Anvret, M. (1993) Proc. Natl. Acad. Sci. U.S.A.
90, 79377940
491. Mer, G., Hietter, H., Kellenberger, C., Renatus,
M., Luu, B., and Lefvre, J.-F. (1996) J. Mol.
Biol. 258, 158171
492. Conconi, A., and Ryan, C. A. (1993) J. Biol.
Chem. 268, 430435
493. Werner, M. H., and Wemmer, D. E. (1991)
Biochemistry 30, 33563364
494. McBride, J. D., Brauer, A. B. E., Nievo, M., and
Leatherbarrow, R. J. (1998) J. Mol. Biol. 282,
447457
495. Song, H. K., and Suh, S. W. (1998) J. Mol. Biol.
275, 347363
496. Brandt, P., and Woodward, C. (1987)
Biochemistry 26, 31563162
497. Huber, R., and Bode, W. (1978) Acc. Chem. Res.
11, 114122
498. Potempa, J., Korzus, E., and Travis, J. (1994) J.
Biol. Chem. 269, 1595715960
499. Whisstock, J., Skinner, R., and Lesk, A. M.
(1998) Trends Biochem. Sci. 23, 6367
500. Lukacs, C. M., Rubin, H., and Christianson,
D. W. (1998) Biochemistry 37, 32973304
500a. Whisstock, J. C., Skinner, R., Carrell, R. W., and
Lesk, A. M. (2000) J. Mol. Biol. 296, 685699
501. Lawrence, D. A., Ginsburg, D., Day, D. E.,
Berkenpas, M. B., Verhamme, I. M., Kvassman,
J.-O., and Shore, J. D. (1995) J. Biol. Chem. 270,
2530925312
502. Kaslik, G., Kardos, J., Szab, E., Szilgyi, L.,
Zvodszky, P., Westler, W. M., Markley, J. L.,
and Grf, L. (1997) Biochemistry 36, 54555464
503. Bode, W., Wei, A.-Z., Huber, R., Meyer, E.,
Travis, J., and Neumann, S. (1986) EMBO J. 5,
24532458
504. Boswell, D. R., and Carrell, R. (1986) Trends
Biochem. Sci. 11, 102103
505. Curiel, D. T., Holmes, M. D., Okayama, H.,
Brantly, M. L., Vogelmeier, C., Travis, W. D.,
Stier, L. E., Perks, W. H., and Crystal, R. G.
(1989) J. Biol. Chem. 264, 1393813945
506. Lomas, D. A., Finch, J. T., Seyama, K.,
Nukiwa, T., and Carrell, R. W. (1993) J. Biol.
Chem. 268, 1533315335
507. Carrell, R. W., Jeppsson, J.-O., Laurell, C.-B.,
Brennan, S. O., Owen, M. C., Vaughan, L., and
Boswell, D. R. (1982) Nature (London) 298,
329334
508. Carp, H., Miller, F., Hoidal, J. R., and Janoff,
A. (1982) Proc. Natl. Acad. Sci. U.S.A. 79, 2041
2045
509. Radinsky, L. (1983) Science 221, 11871191
510. Culliton, B. J. (1989) Science 246, 750751
511. Matheson, N. R., Gibson, H. E., Hallewell, R.
A., Barr, P. J., and Travis, J. (1986) J. Biol. Chem.
261, 1040410409
512. Rosenfeld, M. A., Siegfried, W., Yoshimura,
K., Yoneyama, K., Fukayama, M., Stier, L. E.,
Pkk, P. K., Gilardi, P., Stratford-
Perricaudet, L. D., Perricaudet, M., Jallat, S.,
Pavirani, A., Lecocq, J.-P., and Crystal, R. G.
(1991) Science 252, 431434
513. Owen, M. C., Brennan, S. O., Lewis, J. H., and
Carrell, R. W. (1983) N. Engl. J. Med. 309, 694
698
514. Davie, E. W., Fujikawa, K., and Kisiel, W.
(1991) Biochemistry 30, 1036410370
515. Smith, E. L., Hill, R. L., Lehman, I. R.,
Lefkowitz, R. J., Handler, P., and White, A.
(1983) in Principles of Biochemistry, Mammalian
Biochemistry, 7th ed., pp. 1737, McGraw-Hill,
New York
516. Mann, K. G., Jenny, R. J., and Krishnaswamy,
S. (1988) Ann. Rev. Biochem. 57, 915956
517. Zucker, M. B. (1980) Sci. Am. 242 (Jun), 86103
518. Gibbs, C. S., McCurdy, S. N., Leung, L. L. K.,
and Paborsky, L. R. (1994) Biochemistry 33,
1400314010
519. Muller, Y. A., Ultsch, M. H., Kelley, R. F., and de
Vos, A. M. (1994) Biochemistry 33, 1086410870
520. Harlos, K., Martin, D. M. A., OBrien, D. P.,
Jones, E. Y., Stuart, D. I., Polikarpov, I., Miller,
A., Tuddenham, E. G. D., and Boys, C. W. G.
(1994) Nature (London) 370, 662666
521. Martin, D. M. A., Boys, C. W. G., and Ruf, W.
(1995) FASEB J. 9, 852859
522. Ruf, W., Kelly, C. R., Schullek, J. R., Martin, D.
M. A., Polikarpov, I., Boys, C. W. G.,
Tuddenham, E. G. D., and Edgington, T. S.
(1995) Biochemistry 34, 63106315
523. Muller, Y. A., Ultsch, M. H., and de Vos, A. M.
(1996) J. Mol. Biol. 256, 144159
524. Doolittle, R. F. (1981) Sci. Am. 245 (Dec), 126
135
525. Fu, Y., Weissbach, L., Plant, P. W., Oddoux, C.,
Cao, Y., Liang, T. J., Roy, S. N., Redman, C. M.,
and Grieninger, G. (1992) Biochemistry 31,
1196811972
526. Hunziker, E. B., Straub, P. W., and Haeberli,
A. (1990) J. Biol. Chem. 265, 74557463
527. Banner, D. W., DArcy, A., Chne, C., Winkler,
F. K., Guha, A., Konigsberg, W. H., Nemerson,
Y., and Kirchhofer, D. (1996) Nature (London)
380, 4146
527a. Banner, D. W. (2000) Nature (London) 404,
449450
527b. Chang, Y.-J., Hamaguchi, N., Chang, S.-C.,
Ruf, W., Shen, M.-C., and Lin, S.-W. (1999)
Biochemistry 38, 1094010948
528. Muranyi, A., Finn, B. E., Gippert, G. P.,
Forsn, S., Stenflo, J., and Drakenberg, T.
(1998) Biochemistry 37, 1060510615
529. Andrews, B. S. (1991) Trends Biochem. Sci. 16,
3136
530. Neuenschwander, P. F., and Morrissey, J. H.
(1992) J. Biol. Chem. 267, 1447714482
531. Altieri, D. C. (1995) FASEB J. 9, 860865
532. Brandstetter, H., Khne, A., Bode, W., Huber,
R., von der Saal, W., Wirthensohn, K., and Engh,
R. A. (1996) J. Biol. Chem. 271, 2998829992
533. Sabharwal, A. K., Padmanabhan, K., Tulinsky,
A., Mathur, A., Gorka, J., and Bajaj, S. P. (1997)
J. Biol. Chem. 272, 2203722045
534. Dharmawardana, K. R., and Bock, P. E. (1998)
Biochemistry 37, 1314313152
534a. Macedo-Ribeiro, S., Bode, W., Huber, R.,
Quinn-Allen, M. A., Kim, S. W., Ortel, T. L.,
Bourenkov, G. P., Bartunik, H. D., Stubbs, M.
T., Kane, W. H., and Fuentes-Prior, P. (1999)
Nature (London) 402, 434439
535. Xue, J., Kalafatis, M., Silveira, J. R., Kung, C.,
and Mann, K. G. (1994) Biochemistry 33,
1310913116
536. Comfurius, P., Smeets, E. F., Willems, G. M.,
Bevers, E. M., and Zwaal, R. F. A. (1994)
Biochemistry 33, 1031910324
537. Betz, A., and Krishnaswamy, S. (1998) J. Biol.
Chem. 273, 1070910718
538. Bottenus, R. E., Ichinose, A., and Davie, E. W.
(1990) Biochemistry 29, 1119511209
539. Slon-Usakiewicz, J. J., Purisima, E., Tsuda, Y.,
Sulea, T., Pedyczak, A., Fthire, J., Cygler,
M., and Konishi, Y. (1997) Biochemistry 36,
1349413502
540. Hughes, P. E., Morgan, G., Rooney, E. K.,
Brownlee, G. G., and Handford, P. (1993) J.
Biol. Chem. 268, 1772717733
540a. Mathur, A., and Bajaj, S. P. (1999) J. Biol. Chem.
274, 18477 18486
540b. Tolkatchev, D., Ng, A., Zhu, B., and Ni, F.
(2000) Biochemistry 39, 1036510372
541. Xu, W.-f, Chung, D. W., and Davie, E. W.
(1996) J. Biol. Chem. 271, 2794827953
541a. Brown, J. H., Volkmann, N., Jun, G., Hen-
schen-Edman, A. H., and Cohen, C. (2000)
Proc. Natl. Acad. Sci. U.S.A. 97, 8590
542. Doolittle, R. F., Everse, S. J., and Spraggon, G.
(1996) FASEB J. 10, 14641470
References
670 Chapter 12. Transferring Groups by Displacement Reactions
572. Kaufman, R. J. (1989) Nature (London) 342,
207208
573. Dwarki, V. J., Belloni, P., Nijjar, T., Smith, J.,
Couto, L., Rabier, M., Clift, S., Berns, A., and
Cohen, L. K. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 10231027
574. Kay, M. A., Rothenberg, S., Landen, C. N.,
Bellinger, D. A., Leland, F., Toman, C.,
Finegold, M., Thompson, A. R., Read, M. S.,
Brinkhous, K. M., and Woo, S. L. C. (1993)
Science 262, 117119
575. Jesty, J., Wun, T.-C., and Lorenz, A. (1994)
Biochemistry 33, 1268612694
576. Hajjar, K. A. (1994) N. Engl. J. Med. 331, 1585
1587
577. Bjrk, I., Nordling, K., and Olson, S. T. (1993)
Biochemistry 32, 65016505
578. Streusand, V. J., Bjrk, I., Gettins, P. G. W.,
Petitou, M., and Olson, S. T. (1995) J. Biol.
Chem. 270, 90439051
578a. Whisstock, J. C., Pike, R. N., Jin, L., Skinner,
R., Pei, X. Y., Carrell, R. W., and Lesk, A. M.
(2000) J. Mol. Biol. 301, 12871305
579. Di Cera, E., Guinto, E. R., Vindigni, A., Dang,
Q. D., Ayala, Y. M., Wuyi, M., and Tulinsky, A.
(1995) J. Biol. Chem. 270, 2208922092
580. Weisel, J. W., Nagaswami, C., Young, T. A.,
and Light, D. R. (1996) J. Biol. Chem. 271,
3148531490
580a. Fuentes-Prior, P., Iwanaga, Y., Huber, R.,
Pagila, R., Rumennik, G., Seto, M., Morser, J.,
Light, D. R., and Bode, W. (2000) Nature
(London) 404, 518525
580b. Baerga-Ortiz, A., Rezaie, A. R., and Komives,
E. A. (2000) J. Mol. Biol. 296, 651658
581. Esmon, C. T. (1995) FASEB J. 9, 946 955
582. Colpitts, T. L., Prorok, M., and Castellino, F. J.
(1995) Biochemistry 34, 24242430
582a. Shen, L., Dahlbck, B., and Villoutreix, B. O.
(2000) Biochemistry 39, 28532860
583. hlin, A., Landes, G., Bourden, P.,
Oppenheimer, C., Wydro, R., and Stenflo, J.
(1988) J. Biol. Chem. 263, 1924019248
584. Plow, E. F., Herren, T., Redlitz, A., Miles, L. A.,
and Hoover-Plow, J. L. (1995) FASEB J. 9, 939
945
585. Menhart, N., Hoover, G. J., McCance, S. G.,
and Castellino, F. J. (1995) Biochemistry 34,
14821488
586. Bendixen, E., Harpel, P. C., and Sottrup-Jensen,
L. (1995) J. Biol. Chem. 270, 1792917933
587. Pirie-Shepherd, S. R., Jett, E. A., Andon, N. L.,
and Pizzo, S. V. (1995) J. Biol. Chem. 270, 5877
5881
588. Marti, D. N., Hu, C.-K., An, S. S. A., von
Haller, P., Schaller, J., and Llins, M. (1997)
Biochemistry 36, 1159111604
589. Bogusky, M. J., Dobson, C. M., and Smith, R.
A. G. (1989) Biochemistry 28, 67286735
590. Byeon, I.-J. L., Kelley, R. F., Mulkerrin, M. G.,
An, S. S. A., and Llins, M. (1995) Biochemistry
34, 27392750
591. Parry, M. A. A., Zhang, X. C., and Bode, W.
(2000) Trends Biochem. Sci. 25, 5359
592. Oates, J. A., Wood, A. J. J., Loscalzo, J., and
Braunwald, E. (1988) N. Engl. J. Med. 319,
925931
593. Mottonen, J., Strand, A., Symersky, J., Sweet,
R. M., Danley, D. E., Geoghegan, K. F.,
Gerard, R. D., and Goldsmith, E. J. (1992)
Nature (London) 355, 270273
594. Szyperski, T., Antuch, W., Schick, M., Betz, A.,
Stone, S. R., and Wthrich, K. (1994)
Biochemistry 33, 93039310
595. Betz, A., Hofsteenge, J., and Stone, S. R. (1992)
Biochemistry 31, 45574562
References
596. Vitali, J., Martin, P. D., Malkowski, M. G.,
Robertson, W. D., Lazar, J. B., Winant, R. C.,
Johnson, P. H., and Edwards, B. F. P. (1992) J.
Biol. Chem. 267, 1767017678
597. van de Locht, A., Lamba, D., Bauer, M., Huber,
R., Friedrich, T., Krger, B., Hffken, W., and
Bode, W. (1995) EMBO J. 14, 51495157
598. Stark, K. R., and James, A. A. (1998) J. Biol.
Chem. 273, 2080220809
599. Lim-Wilby, M. S. L., Hallenga, K., De Maeyer,
M., Lasters, I., Vlasuk, G. P., and Brunck, T. K.
(1995) Protein Sci. 4, 178186
600. van de Locht, A., Stubbs, M. T., Bode, W.,
Friedrich, T., Bollschweiler, C., Hffken, W.,
and Huber, R. (1996) EMBO J. 15, 60116017
601. Tabernero, L., Chang, C. Y. Y., Ohringer, S. L.,
Lau, W. F., Iwanowicz, E. J., Han, W.-C.,
Wang, T. C., Seiler, S. M., Roberts, D. G. M.,
and Sack, J. S. (1995) J. Mol. Biol. 246, 1420
602. Cheng, Y., Slon-Usakiewicz, J. J., Wang, J.,
Purisima, E. O., and Konishi, Y. (1996)
Biochemistry 35, 1302113029
603. Krishnan, R., Tulinsky, A., Vlasuk, G. P.,
Pearson, D., Vallar, P., Bergum, P., Brunck, T. K.,
and Ripka, W. C. (1996) Protein Sci. 5, 422433
604. Kamata, K., Kawamoto, H., Honma, T.,
Iwama, T., and Kim, S.-H. (1998) Proc. Natl.
Acad. Sci. U.S.A. 95, 66306635
605. Lu, D., Bovill, E. G., and Long, G. L. (1994) J.
Biol. Chem. 269, 2903229038
606. Ichinose, A. (1992) Biochemistry 31, 31133118
607. Olds, R. J., Lane, D. A., Chowdhury, V., De
Stefano, V., Leone, G., and Thein, S. L. (1993)
Biochemistry 32, 42164224
608. Watton, J., Longstaff, C., Lane, D. A., and
Barrowcliffe, T. W. (1993) Biochemistry 32,
72867293
609. Taylor, P. (1991) J. Biol. Chem. 266, 40254028
610. Sussman, J. L., Harel, M., Frolow, F., Oefner,
C., Goldman, A., Toker, L., and Silman, I.
(1991) Science 253, 872879
611. Maelicke, A. (1991) Trends Biochem. Sci. 16,
355356
611a. Koellner, G., Kryger, G., Millard, C. B.,
Silman, I., Sussman, J. L., and Steiner, T.
(2000) J. Mol. Biol. 296, 713735
612. Vellom, D. C., Radic, Z., Li, Y., Pickering, N.
A., Camp, S., and Taylor, P. (1993) Biochemistry
32, 1217
613. Kovach, I. M., Huber, J. H.-A., and Schowen,
R. L. (1988) J. Am. Chem. Soc. 110, 590593
614. Radic, Z., Kirchhoff, P. D., Quinn, D. M.,
McCammon, J. A., and Taylor, P. (1997) J. Biol.
Chem. 272, 2326523277
615. Wlodek, S. T., Antosiewicz, J., and Briggs, J.
M. (1997) J. Am. Chem. Soc. 119, 81598165
616. Haas, R., Marshall, T. L., and Rosenberry, T. L.
(1988) Biochemistry 27, 64536457
617. Krejci, E., Thomine, S., Boschetti, N., Legay,
C., Sketelj, J., and Massouli, J. (1997) J. Biol.
Chem. 272, 2284022847
618. Pindel, E. V., Kedishvili, N. Y., Abraham, T. L.,
Brzezinski, M. R., Zhang, J., Dean, R. A., and
Bosron, W. F. (1997) J. Biol. Chem. 272, 14769
14775
619. Li, J., Szittner, R., Derewenda, Z. S., and
Meighen, E. A. (1996) Biochemistry 35, 9967
9973
620. Derewenda, Z. S., and Sharp, A. M. (1993)
Trends Biochem. Sci. 18, 2025
621. Quinn, D. M., and Feaster, S. R. (1998) in
Comprehensive Biological Catalysis. A
Mechanistic Reference, Vol. I (Sinnott, M., ed),
pp. 455482, Academic Press, San Diego,
California
622. Bourne, Y., Martinez, C., Kerfelec, B.,
Lombardo, D., Chapus, C., and Cambillau, C.
(1994) J. Mol. Biol. 238, 709732
623. Egloff, M.-P., Marguet, F., Buono, G., Verger,
R., Cambillau, C., and van Tilbeurgh, H.
(1995) Biochemistry 34, 27512762
624. Dahim, M., and Brockman, H. (1998)
Biochemistry 37, 83698377
625. Jentoft, N. (1990) Trends Biochem. Sci. 15, 291294
626. Chen, J. C.-H., Miercke, L. J. W., Krucinski, J.,
Starr, J. R., Saenz, G., Wang, X., Spilburg, C.
A., Lange, L. G., Ellsworth, J. L., and Stroud,
R. M. (1998) Biochemistry 37, 51075117
627. Gargouri, Y., Moreau, H., Pieroni, G., and
Verger, R. (1988) J. Biol. Chem. 263, 21592162
628. Warner, T. G., Dambach, L. M., Shin, J. H., and
OBrien, J. S. (1981) J. Biol. Chem. 256, 29522957
629. Emmerich, J., Beg, O. U., Peterson, J.,
Previato, L., Brunzell, J. D., Brewer, J., HB,
and Santamarina-Fojo, S. (1992) J. Biol. Chem.
267, 41614165
630. Kobayashi, J., Applebaum-Bowden, D., Dugi,
K. A., Brown, D. R., Kashyap, V. S., Parrott, C.,
Duarte, C., Maeda, N., and Santamarina-Fojo,
S. (1996) J. Biol. Chem. 271, 2629626301
631. Shen, W.-J., Patel, S., Natu, V., and Kraemer, F.
B. (1998) Biochemistry 37, 89738979
632. Nicolas, A., Egmond, M., Verrips, C. T., de
Vlieg, J., Longhi, S., Cambillau, C., and
Martinez, C. (1996) Biochemistry 35, 398410
633. Grochulski, P., Bouthillier, F., Kazlauskas, R.
J., Serreqi, A. N., Schrag, J. D., Ziomek, E., and
Cygler, M. (1994) Biochemistry 33, 34943500
634. Lang, D., Hofmann, B., Haalck, L., Hecht, H.-
J., Spener, F., Schmid, R. D., and Schomburg,
D. (1996) J. Mol. Biol. 259, 704717
635. Uppenberg, J., hrner, N., Norin, M., Hult,
K., Kleywegt, G. J., Patkar, S., Waagen, V.,
Anthonsen, T., and Jones, T. A. (1995)
Biochemistry 34, 1683816851
636. Brocca, S., Schmidt-Dannert, C., Lotti, M.,
Alberghina, L., and Schmid, R. D. (1998)
Protein Sci. 7, 14151422
637. Ordentlich, A., Barak, D., Kronman, C., Ariel,
N., Segall, Y., Velan, B., and Shafferman, A.
(1998) J. Biol. Chem. 273, 1950919517
638. Axelsen, P. H., Harel, M., Silman, I., and
Sussman, J. L. (1994) Protein Sci. 3, 188197
639. Brady, L., Brzozowski, A. M., Derewenda, Z.
S., Dodson, E., Dodson, G., Tolley, S.,
Turkenburg, J. P., Christiansen, L., Huge-
Jensen, B., Norskov, L., Thim, L., and Menge,
U. (1990) Nature (London) 343, 767770
640. Dennis, E. A. (1997) Trends Biochem. Sci. 22, 12
641. Balsinde, J., and Dennis, E. A. (1997) J. Biol.
Chem. 272, 1606916072
642. Leslie, C. C. (1997) J. Biol. Chem. 272, 16709
16712
643. Scott, D. L., White, S. P., Otwinowski, Z.,
Yuan, W., Gelb, M. H., and Sigler, P. B. (1990)
Science 250, 15411546
644. Brunie, S., Bolin, J., Gewirth, D., and Sigler, P.
B. (1985) J. Biol. Chem. 260, 97429749
645. Scott, D. L., Achari, A., Vidal, J. C., and Sigler,
P. B. (1992) J. Biol. Chem. 267, 2264522657
646. van den Berg, B., Tessari, M., de Haas, G. H.,
Verheij, H. M., Boelens, R., and Kaptein, R.
(1995) EMBO J. 14, 41234131
647. Gelb, M. H., Jain, M. K., Hanel, A. M., and
Berg, O. G. (1995) Ann. Rev. Biochem. 64, 653
688
648. Thuren, T., Tulkki, A.-P., Virtanen, J. A., and
Kinnunen, P. K. J. (1987) Biochemistry 26,
49074910
649. Huber, M., Yee, V. C., Burri, N., Vikerfors, E.,
Lavrijsen, A. P. M., Paller, A. S., and Hohl, D.
(1997) J. Biol. Chem. 272, 2101821026
650. Yee, V. C., Pedersen, L. C., Le Trong, I.,
Bishop, P. D., Stenkamp, R. E., and Teller, D.
C. (1994) Proc. Natl. Acad. Sci. U.S.A. 91,
72967300
671
718. Buchwald, S. L., Saini, M. S., Knowles, J. R.,
and Van Etten, R. L. (1984) J. Biol. Chem. 259,
22082213
719. Ma, L., and Kantrowitz, E. R. (1996)
Biochemistry 35, 23942402
720. Sugiura, Y., Kawabe, H., Tanaka, H.,
Fujimoto, S., and Ohara, A. (1981) J. Biol.
Chem. 256, 1066410670
721. Hong, S.-B., Kuo, J. M., Mullins, L. S., and
Raushel, F. M. (1995) J. Am. Chem. Soc. 117,
75807581
722. Benning, M. M., Kuo, J. M., Raushel, F. M.,
and Holden, H. M. (1995) Biochemistry 34,
79737978
723. Vanhooke, J. L., Benning, M. M., Raushel, F.
M., and Holden, H. M. (1996) Biochemistry 35,
60206025
723a. Benning, M. M., Hong, S.-B., Raushel, F. M.,
and Holden, H. M. (2000) J. Biol. Chem. 275,
3055630560
724. Kuo, J. M., Chae, M. Y., and Raushel, F. M.
(1997) Biochemistry 36, 19821988
724a. Shim, H., and Raushel, F. M. (2000)
Biochemistry 39, 73577364
725. Williams, N. H. (1998) in Comprehensive
Biological Catalysis. A Mechanistic Reference,
Vol. I (Sinnott, M., ed), pp. 543561,
Academic Press, San Diego, California
726. Villeret, V., Huang, S., Zhang, Y., and
Lipscomb, W. N. (1995) Biochemistry 34,
43074315
727. Kurbanov, F. T., Choe, J.-y, Honzatko, R. B.,
and Fromm, H. J. (1998) J. Biol. Chem. 273,
1751117516
728. Choe, J.-Y., Fromm, H. J., and Honzatko, R. B.
(2000) Biochemistry 39, 85658574
728a. Lee, Y.-H., Ogata, C., Pflugrath, J. W., Levitt,
D. G., Sarma, R., Banaszak, L. J., and Pilkis, S.
J. (1996) Biochemistry 35, 60106019
729. Lu, G., Giroux, E. L., and Kantrowitz, E. R.
(1997) J. Biol. Chem. 272, 50765081
730. Villeret, V., Huang, S., Zhang, Y., Xue, Y., and
Lipscomb, W. N. (1995) Biochemistry 34, 4299
4306
731. Lei, K.-J., Pan, C.-J., Liu, J.-L., Shelly, L. L.,
and Chou, J. Y. (1995) J. Biol. Chem. 270,
1188211886
732. Clottes, E., and Burchell, A. (1998) J. Biol.
Chem. 273, 1939119397
733. Josse, J., and Wong, S. C. K. (1971) in The
Enzymes, 3rd ed., Vol. 4 (Boyer, P. D., ed), pp.
499 541, Academic Press, New York
734. Harutyunyan, E. H., Oganessyan, V. Y.,
Oganessyan, N. N., Avaeva, S. M., Nazarova,
T. I., Vorobyeva, N. N., Kurilova, S. A., Huber,
R., and Mather, T. (1997) Biochemistry 36,
77547760
735. Salminen, A., Efimova, I. S., Parfenyev, A. N.,
Magretova, N. N., Mikalahti, K., Goldman, A.,
Baykov, A. A., and Lahti, R. (1999) J. Biol.
Chem. 274, 3389833904
736. Pohjanjoki, P., Lahti, R., Goldman, A., and
Cooperman, B. S. (1998) Biochemistry 37,
17541761
736a. Lappnen, V.-M., Nummelin, H., Hansen, T.,
Lahti, R., Schfer, G., and Goldman, A. (1999)
Protein Sci. 8, 12181231
736b. Baykov, A. A., Fabrichniy, I. P., Pohjanjoki, P.,
Zyryanov, A. B., and Lahti, R. (2000)
Biochemistry 39, 1193911947
737. Barford, D. (1996) Trends Biochem. Sci. 21,
407412
738. Goldberg, J., Huang, H., Kwon, Y., Greengard,
P., Nairn, A. C., and Kuriyan, J. (1995) Nature
(London) 376, 745753
739. Egloff, M.-P., Cohen, P. T. W., Reinemer, P., and
Barford, D. (1995) J. Mol. Biol. 254, 942959
740. Zhang, J., Zhang, Z., Brew, K., and Lee, E. Y.
C. (1996) Biochemistry 35, 62766282
651. Kulkarni, M. S., and Sherman, F. (1994) J. Biol.
Chem. 269, 1314113147
652. Wu, D., and Hersh, L. B. (1995) J. Biol. Chem.
270, 2911129116
653. Schwartz, B., and Drueckhammer, D. G.
(1996) J. Am. Chem. Soc. 118, 98269830
654. Ellis, J., Bagshaw, C. R., and Shaw, W. V.
(1995) Biochemistry 34, 1685216859
655. Murray, I. A., Cann, P. A., Day, P. J., Derrick, J.
P., Sutcliffe, M. J., Shaw, W. V., and Leslie, A.
G. W. (1995) J. Mol. Biol. 254, 9931005
656. Guillaume, G., Vanhove, M., Lamotte-
Brasseur, J., Ledent, P., Jamin, M., Joris, B.,
and Frre, J.-M. (1997) J. Biol. Chem. 272,
54385444
657. Maveyraud, L., Pratt, R. F., and Samama, J.-P.
(1998) Biochemistry 37, 26222628
658. Crowder, M. W., Wang, Z., Franklin, S. L.,
Zovinka, E. P., and Benkovic, S. J. (1996)
Biochemistry 35, 1212612132
659. Orellano, E. G., Girardini, J. E., Cricco, J. A.,
Ceccarelli, E. A., and Vila, A. J. (1998)
Biochemistry 37, 1017310180
660. Lubkowski, J., Wlodawer, A., Ammon, H. L.,
Copeland, T. D., and Swain, A. L. (1994)
Biochemistry 33, 1025710265
661. Stewart, A. E., Arfin, S. M., and Bradshaw, R.
A. (1995) J. Biol. Chem. 270, 2528
662. Nakamura, N., Inoue, N., Watanabe, R.,
Takahashi, M., Takeda, J., Stevens, V. L., and
Kinoshita, T. (1997) J. Biol. Chem. 272, 15834
15840
663. Knowles, J. R. (1980) Ann. Rev. Biochem. 49,
877919
664. Westheimer, F. H. (1968) Acc. Chem. Res. 1,
7078
665. Westheimer, F. H. (1987) Science 235, 11731178
666. Hengge, A. C. (1998) in Comprehensive
Biological Catalysis. A Mechanistic Reference,
Vol. I (Sinnott, M., ed), pp. 517542,
Academic Press, San Diego, California
667. Vincent, J. B., Crowder, M. W., and Averill, B.
A. (1992) Trends Biochem. Sci. 17, 105110
668. Mislow, K. (1970) Acc. Chem. Res. 3, 321331
669. Bunton, C. A. (1970) Acc. Chem. Res. 3, 257265
670. Gorenstein, D. G., Luxon, B. A., and Findlay, J.
B. (1979) J. Am. Chem. Soc. 101, 58695875
671. Taira, K., Mock, W. L., and Gorenstein, D. G.
(1984) J. Am. Chem. Soc. 106, 78317835
672. Calvo, K. C., and Westheimer, F. H. (1984) J.
Am. Chem. Soc. 106, 42054210
673. Friedman, J. M., Freeman, S., and Knowles, J.
R. (1988) J. Am. Chem. Soc. 110, 12681275
674. Wu, Y.-D., and Houk, K. N. (1993) J. Am.
Chem. Soc. 115, 1199712002
675. Buchwald, S. L., Friedman, J. M., and
Knowles, J. R. (1984) J. Am. Chem. Soc. 106,
49114916
676. Herschlag, D., and Jencks, W. P. (1989) J. Am.
Chem. Soc. 111, 75797586
677. Hengge, A. C., Edens, W. A., and Elsing, H.
(1994) J. Am. Chem. Soc. 116, 50455049
678. Jankowski, S., Quin, L. D., Paneth, P., and
OLeary, M. H. (1994) J. Am. Chem. Soc. 116,
1167511677
679. Herschlag, D., and Jencks, W. P. (1990)
Biochemistry 29, 51725179
680. Admiraal, S. J., and Herschlag, D. (1999) J.
Am. Chem. Soc. 121, 58375845
681. Cohn, M., and Reed, G. H. (1982) Ann. Rev.
Biochem. 51, 365394
682. Villafranca, J. J., and Raushel, F. M. (1980)
Annu Rev Biophys Bioeng. 9, 363392
683. McLaughlin, A. C., Leigh, J. S., Jr., and Cohn,
M. (1976) J. Biol. Chem. 251, 27772787
684. Leyh, T. S., Goodhart, P. J., Nguyen, A. C.,
Kenyon, G. L., and Reed, G. H. (1985)
Biochemistry 24, 308316
685. Nageswara Rao, B. D., Kayne, F. J., and Cohn,
M. (1979) J. Biol. Chem. 254, 26892696
686. Stackhouse, J., Nambiar, K. P., Burbaum, J. J.,
Stauffer, D. M., and Benner, S. A. (1985) J. Am.
Chem. Soc. 107, 27572763
687. Rao, B. D. N., and Cohn, M. (1981) J. Biol.
Chem. 256, 17161721
688. Gadian, D. G., Radda, G. K., Brown, T. R.,
Chance, E. M., Dawson, M. J., and Wilke, D.
R. (1981) Biochem. J. 194, 215228
689. Mendz, G. L., Robinson, G., and Kuchel, P. W.
(1986) J. Am. Chem. Soc. 108, 169173
690. Midelfort, C. F., and Rose, I. A. (1976) J. Biol.
Chem. 251, 58815887
691. Lowe, G., and Tuck, S. P. (1986) J. Am. Chem.
Soc. 108, 13001301
692. Abbott, S. J., Jones, S. R., Weinman, S. A.,
Bockhoff, F. M., McLafferty, F. W., and
Knowles, J. R. (1979) J. Am. Chem. Soc. 101,
43234332
693. Pliura, D. H., Schomburg, D., Richard, J. P.,
Frey, P. A., and Knowles, J. R. (1980)
Biochemistry 19, 325329
694. Sammons, R. D., and Frey, P. A. (1982) J. Biol.
Chem. 257, 11381141
695. Eckstein, F. (1980) Trends Biochem. Sci. 5, 157
159
696. Baraniak, J., and Frey, P. A. (1988) J. Am. Chem.
Soc. 110, 40594060
697. Richard, J. P., and Frey, P. A. (1982) J. Am.
Chem. Soc. 104, 34763481
698. Hassett, A., Blttler, W., and Knowles, J. R.
(1982) Biochemistry 21, 63356340
699. Mehdi, S., and Gerlt, J. A. (1984) Biochemistry
23, 48444852
700. Sammons, R. D., Frey, P. A., Bruzik, K., and
Tsai, M.-D. (1983) J. Am. Chem. Soc. 105, 5455
5461
701. Buchwald, S. L., and Knowles, J. R. (1980) J.
Am. Chem. Soc. 102, 66026604
702. Cornelius, R. D., and Cleland, W. W. (1978)
Biochemistry 17, 32793286
703. Dunaway-Mariano, D., and Cleland, W. W.
(1980) Biochemistry 19, 15061515
704. Speckhard, D. C., Pecoraro, V. L., Knight, W.
B., and Cleland, W. W. (1986) J. Am. Chem. Soc.
108, 41674171
705. Lin, I., and Dunaway-Mariano, D. (1988) J.
Am. Chem. Soc. 110, 950956
706. Burgers, P. M. J., and Eckstein, F. (1980) J. Biol.
Chem. 255, 82298233
707. Lester, L. M., Rusch, L. A., Robinson, G. J.,
and Speckhard, D. C. (1998) Biochemistry 37,
53495355
708. Matlin, A. R., Kendall, D. A., Carano, K. S.,
Banzon, J. A., Klecka, S. B., and Solomon, N.
M. (1992) Biochemistry 31, 81968200
709. Simopoulos, T. T., and Jencks, W. P. (1994)
Biochemistry 33, 1037510380
710. Han, R., and Coleman, J. E. (1995) Biochemistry
34, 42384245
711. Craig, D. B., Arriaga, E. A., Wong, J. C. Y., Lu,
H., and Dovichi, N. J. (1996) J. Am. Chem. Soc.
118, 52455253
712. Kimura, E., Kodama, Y., Koike, T., and Shiro,
M. (1995) J. Am. Chem. Soc. 117, 83048311
713. Stec, B., Hehir, M. J., Brennan, C., Nolte, M.,
and Kantrowitz, E. R. (1998) J. Mol. Biol. 277,
647662
713a. Stec, B., Holtz, K. M., and Kantrowitz, E. R.
(2000) J. Mol. Biol. 299, 13031311
714. Florin, J., and Warshel, A. (1997) J. Am. Chem.
Soc. 119, 54735474
715. Herschman, H. R. (1980) Trends Biochem. Sci. 5,
8284
716. Ostanin, K., and Van Etten, R. L. (1993) J. Biol.
Chem. 268, 2077820784
717. VanEtten, R. L., Waymack, P. P., and Rehkop,
D. M. (1974) J. Am. Chem. Soc. 96, 67836785
References
672 Chapter 12. Transferring Groups by Displacement Reactions
741. Zhang, M., Zhou, M., Van Etten, R. L., and
Stauffacher, C. V. (1997) Biochemistry 36, 1523
742. Hengge, A. C., Sowa, G. A., Wu, L., and Zhang,
Z.-Y. (1995) Biochemistry 34, 1398213987
743. Yuvaniyama, J., Denu, J. M., Dixon, J. E., and
Saper, M. A. (1996) Science 272, 13281331
744. DAlessio, G., and Riordan, J. F., eds. (1997)
Ribonuclease Structure and Functions, Academic
Press, San Diego, California
745. Moore, S., and Stein, W. H. (1973) Science 180,
458464
746. Wlodawer, A., Svensson, L. A., Sjlin, L., and
Gilliland, G. L. (1988) Biochemistry 27, 2705
2717
747. Santoro, J., Gonzlez, C., Bruix, M., Neira, J.
L., Nieto, J. L., Herranz, J., and Rico, M. (1993)
J. Mol. Biol. 229, 722734
748. Tilton, R. F., Jr., Dewan, J. C., and Petsko, G.
A. (1992) Biochemistry 31, 24692481
749. Fedorov, A. A., Joseph-McCarthy, D., Fedorov,
E., Sirakova, D., Graf, I., and Almo, S. C.
(1996) Biochemistry 35, 1596215979
750. Boix, E., Nogus, M. V., Schein, C. H., Benner,
S. A., and Cuchillo, C. M. (1994) J. Biol. Chem.
269, 25292534
751. Schultz, L. W., Quirk, D. J., and Raines, R. T.
(1998) Biochemistry 37, 88868898
752. Jackson, D. Y., Burnier, J., Quan, C., Stanley,
M., Tom, J., and Wells, J. A. (1994) Science 266,
243247
753. Gutte, B. (1977) J. Biol. Chem. 252, 663 670
754. Breslow, R., Dong, S. D., Webb, Y., and Xu, R.
(1996) J. Am. Chem. Soc. 118, 65886600
755. Martin, P. D., Coscha, M. S., and Edwards, B.
F. P. (1987) J. Biol. Chem. 262, 1593015938
756. Wladkowski, B. D., Svensson, L. A., Sjolin, L.,
Ladner, J. E., and Gilliland, G. L. (1998) J. Am.
Chem. Soc. 120, 54885498
757. Eckstein, F. (1979) Acc. Chem. Res. 12, 204210
758. Matta, M. S., and Vo, D. T. (1986) J. Am. Chem.
Soc. 108, 53165318
759. Herschlag, D. (1994) J. Am. Chem. Soc. 116,
1163111635
760. Breslow, R., and Chapman, W. H., Jr. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1001810021
761. Sowa, G. A., Hengge, A. C., and Cleland, W.
W. (1997) J. Am. Chem. Soc. 119, 23192320
762. Kawata, Y., Sakiyama, F., Hayashi, F., and
Kyogoku, Y. (1990) Eur. J. Biochem. 187, 255262
763. Pletinckx, J., Steyaert, J., Zegers, I., Choe, H.-
W., Heinemann, U., and Wyns, L. (1994)
Biochemistry 33, 16541662
764. Kurihara, H., Nonaka, T., Mitsui, Y., Ohgi, K.,
Irie, M., and Nakamura, K. T. (1996) J. Mol.
Biol. 255, 310320
764a. Noguchi, S., Satow, Y., Uchida, T., Sasaki, C.,
and Matsuzaki, T. (1995) Biochemistry 34,
1558315591
765. Cordes, F., Starikov, E. B., and Saenger, W.
(1995) J. Am. Chem. Soc. 117, 1036510372
766. Buckle, A. M., and Fersht, A. R. (1994)
Biochemistry 33, 16441653
767. Deutscher, M. P. (1988) Trends Biochem. Sci. 13,
136139
768. Benner, S. A., and Alleman, R. K. (1989) Trends
Biochem. Sci. 14, 396397
769. Davies, J. F., II, Hostomska, Z., Hostomsky, Z.,
Jordan, S. R., and Matthews, D. A. (1991)
Science 252, 8895
770. Katayanagi, K., Miyagawa, M., Matsushima,
M., Ishikawa, M., Kanaya, S., Ikehara, M.,
Matsuzaki, T., and Morikawa, K. (1990)
Nature (London) 347, 306309
771. Yang, W., Hendrickson, W. A., Crouch, R. J.,
and Satow, Y. (1990) Science 249, 13981405
772. Acharya, K. R., Shapiro, R., Riordan, J. F., and
Vallee, B. L. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 29492953
References
773. Lequin, O., Albaret, C., Bontems, F., Spik, G.,
and Lallemand, J.-Y. (1996) Biochemistry 35,
88708880
774. Shapiro, R. (1998) Biochemistry 37, 68476856
775. Rosenberg, H. F., Tenen, D. G., and
Ackerman, S. J. (1989) Proc. Natl. Acad. Sci.
U.S.A. 86, 44604464
776. Leland, P. A., Schultz, L. W., Kim, B.-M., and
Raines, R. T. (1998) Proc. Natl. Acad. Sci. U.S.A.
95, 1040710412
777. DAlessio, G., Di Donato, A., Parente, A., and
Piccoli, R. (1991) Trends Biochem. Sci. 16, 104
106
778. Hofsteenge, J., Kieffer, B., Matthies, R.,
Hemmings, B. A., and Stone, S. R. (1988)
Biochemistry 27, 85378544
779. Kole, R., and Altman, S. (1981) Biochemistry
20, 19021906
780. Altman, S., Baer, M., Guerrier-Takada, C., and
Vioque, A. (1986) Trends Biochem. Sci. 11, 515
518
781. Altman, S., Kirsebom, L., and Talbot, S. (1993)
FASEB J. 7, 714
782. Yuan, Y., and Altman, S. (1995) EMBO J. 14,
159168
782a. Massire, C., Jaeger, L., and Westhof, E. (1998)
J. Mol. Biol. 279, 773793
783. Waugh, D. S., Green, C. J., and Pace, N. R.
(1989) Science 244, 15691571
784. True, H. L., and Celander, D. W. (1998) J. Biol.
Chem. 273, 71937196
785. Cech, T. R. (1987) Science 236, 15321539
786. Zaug, A. J., and Cech, T. R. (1986) Science 231,
470475
787. Cech, T. R. (1986) Sci. Am. 255 (Nov), 6475
788. Brown, J. W., Haas, E. S., Gilbert, D. G., and
Pace, N. R. (1994) Nucleic Acids Res. 22,
36603662
789. Pan, T. (1995) Biochemistry 34, 902909
790. Golden, B. L., Gooding, A. R., Podell, E. R.,
and Cech, T. R. (1998) Science 282, 259264
791. Cate, J. H., Gooding, A. R., Podell, E., Zhou,
K., Golden, B. L., Kundrot, C. E., Cech, T. R.,
and Doudna, J. A. (1996) Science 273, 16781685
792. Ferr-DAmar, A. R., Zhou, K., and Doudna,
J. A. (1998) Nature (London) 395, 567574
793. Baidya, N., and Uhlenbeck, O. C. (1997)
Biochemistry 36, 11081114
794. Pley, H. W., Flaherty, K. M., and McKay, D. B.
(1994) Nature (London) 372, 6874
795. Scott, W. G., Murray, J. B., Arnold, J. R. P.,
Stoddard, B. L., and Klug, A. (1996) Science
274, 20652069
796. Bevers, S., Xiang, G., and McLaughlin, L. W.
(1996) Biochemistry 35, 64836490
796a. Bevers, S., Ha, S. B., and McLaughlin, L. W.
(1999) Biochemistry 38, 77107718
796b. Kore, A. R., and Eckstein, F. (1999) Biochemis-
try 38, 1091510918
796c. Lyne, P. D., and Karplus, M. (2000) J. Am.
Chem. Soc. 122, 166167
796d. Torres, R. A., and Bruice, T. C. (2000) J. Am.
Chem. Soc. 122, 781791
797. Simorre, J.-P., Legault, P., Baidya, N.,
Uhlenbeck, O. C., Maloney, L., Wincott, F.,
Usman, N., Beigelman, L., and Pardi, A.
(1998) Biochemistry 37, 40344044
798. Sargueil, B., Pecchia, D. B., and Burke, J. M.
(1995) Biochemistry 34, 77397748
798a. Nissen, P., Hansen, J., Ban, N., Moore, P. B.,
and Steitz, T. A. (2000) Science 289, 920930
798b. Muth, G. W., Ortoleva-Donnelly, L., and
Strobel, S. A. (2000) Science 289, 947950
798c. Nakano, S.-i, Chadalavada, D. M., and
Bevilacqua, P. C. (2000) Science 287, 14931497
799. Scott, W. G., and Klug, A. (1996) Trends
Biochem. Sci. 21, 220224
800. Grasby, J. A. (1998) in Comprehensive Biological
Catalysis. A Mechanistic Reference, Vol. I
(Sinnott, M., ed), pp. 563571, Academic
Press, San Diego, California
801. Hampel, K. J., Walter, N. G., and Burke, J. M.
(1998) Biochemistry 37, 1467214682
802. Pan, T., and Uhlenbeck, O. C. (1992) Nature
(London) 358, 560563
803. Chartrand, P., Usman, N., and Cedergren, R.
(1997) Biochemistry 36, 31453150
804. Santoro, S. W., and Joyce, G. F. (1998)
Biochemistry 37, 1333013342
805. Beebe, J. A., Kurz, J. C., and Fierke, C. A.
(1996) Biochemistry 35, 1049310505
806. Torres, R. A., and Bruice, T. C. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 1107711082
807. Pyle, A. M. (1993) Science 261, 709714
808. Peracchi, A., Beigelman, L., Scott, E. C.,
Uhlenbeck, O. C., and Herschlag, D. (1997) J.
Biol. Chem. 272, 2682226826
809. Pontius, B. W., Lott, W. B., and von Hippel, P.
H. (1997) Proc. Natl. Acad. Sci. U.S.A. 94,
22902294
810. Bruice, T. C., Tsubouchi, A., Dempcy, R. O.,
and Olson, L. P. (1996) J. Am. Chem. Soc. 118,
98679875
811. Weinstein, L. B., Jones, B. C. N. M., Cosstick,
R., and Cech, T. R. (1997) Nature (London) 388,
805808
812. Christian, E. L., and Yarus, M. (1993)
Biochemistry 32, 44754480
813. Orita, M., Vinayak, R., Andrus, A., Warashina,
M., Chiba, A., Kaniwa, H., Nishikawa, F.,
Nishikawa, S., and Taira, K. (1996)
J. Biol. Chem. 271, 94479454
814. Hertel, K. J., Peracchi, A., Uhlenbeck, O. C.,
and Herschlag, D. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 84978502
815. Peracchi, A., Karpeisky, A., Maloney, L.,
Beigelman, L., and Herschlag, D. (1998)
Biochemistry 37, 1476514775
816. Joyce, G. F. (1992) Sci. Am. 267 (Dec), 9097
817. Ekland, E. H., Szostak, J. W., and Bartel, D. P.
(1995) Science 269, 364370
818. Wilson, C., and Szostak, J. W. (1995) Nature
(London) 374, 777782
819. Wright, M. C., and Joyce, G. F. (1997) Science
276, 614617
820. Ohmichi, T., and Sugimoto, N. (1997)
Biochemistry 36, 35143521
821. Suga, H., Cowan, J. A., and Szostak, J. W.
(1998) Biochemistry 37, 1011810125
822. Tsang, J., and Joyce, G. F. (1996) J. Mol. Biol.
262, 3142
823. Geyer, C. R., and Sen, D. (1998) J. Mol. Biol.
275, 483489
824. Ceska, T. A., and Sayers, J. R. (1998) Trends
Biochem. Sci. 23, 331336
825. Weber, D. J., Libson, A. M., Gittis, A. G.,
Lebowitz, M. S., and Mildvan, A. S. (1994)
Biochemistry 33, 80178028
826. Libson, A. M., Gittis, A. G., and Lattman, E. E.
(1994) Biochemistry 33, 80078016
827. Hale, S. P., Poole, L. B., and Gerlt, J. A. (1993)
Biochemistry 32, 74797487
828. Grissom, C. B., and Markley, J. L. (1989)
Biochemistry 28, 21162124
829. Serpersu, E. H., Hibler, D. W., Gerlt, J. A., and
Mildvan, A. S. (1989) Biochemistry 28, 1539
1548
830. Suck, D., Lahm, A., and Oefner, C. (1988)
Nature (London) 332, 464468
831. Jones, S. J., Worrall, A. F., and Connolly, B. A.
(1996) J. Mol. Biol. 264, 11541163
832. Frederick, C. A., Grable, J., Melia, M.,
Samudzi, C., Jen-Jacobson, L., Wang, B.-C.,
Greene, P., Boyer, H. W., and Rosenberg, J. M.
(1984) Nature (London) 309, 327331
673
833. McClarin, J. A., Frederick, C. A., Wang, B.-C.,
Greene, P., Boyer, H. W., Grable, J., and
Rosenberg, J. M. (1986) Science 234, 15261541
834. McCarthy, A. D., and Hardie, D. G. (1984)
Trends Biochem. Sci. 9, 6063
835. Kostrewa, D., and Winkler, F. K. (1995)
Biochemistry 34, 683696
836. Newman, M., Strzelecka, T., Dorner, L. F.,
Schildkraut, I., and Aggarwal, A. K. (1995)
Science 269, 656663
837. Bozic, D., Grazulis, S., Siksnys, V., and Huber,
R. (1996) J. Mol. Biol. 255, 176186
838. Connolly, B. A., Echstein, F., and Pingoud, A.
(1984) J. Biol. Chem. 259, 1076010763
839. Corey, D. R., Pei, D., and Schultz, P. G. (1989)
Biochemistry 28, 82778786
840. Ray, W. J., Jr., Hermodsen, M. A., Puvathingal,
J. M., and Mahoney, W. C. (1983) J. Biol. Chem.
258, 91669174
841. Percival, M. D., and Withers, S. G. (1992)
Biochemistry 31, 505512
842. Dai, J.-B., Liu, Y., Ray, W. J., Jr., and Konno, M.
(1992) J. Biol. Chem. 267, 63226337
843. Rose, Z. B. (1986) Trends Biochem. Sci. 11, 253
255
844. Shanske, S., Sakoda, S., Hermodson, M. A.,
DiMauro, S., and Schon, E. A. (1987) J. Biol.
Chem. 262, 1461214617
845. Rigden, D. J., Alexeev, D., Phillips, S. E. V.,
and Fothergill-Gilmore, L. A. (1998) J. Mol.
Biol. 276, 449459
846. Ravel, P., Craescu, C. T., Arous, N., Rosa, J.,
and Garel, M. C. (1997) J. Biol. Chem. 272,
1404514050
847. Collet, J.-F., Stroobant, V., Pirard, M.,
Delpierre, G., and Van Schaftingen, E. (1998) J.
Biol. Chem. 273, 1410714112
848. Kalckar, H. M. (1985) Trends Biochem. Sci. 10,
291293
849. Reed, G. H., and Leyh, T. S. (1980) Biochemis-
try 19, 54725480
850. Schulz, G. E., and Schirmer, R. H. (1979)
Principles of Protein Structure, Springer-Verlag,
New York (pp. 222226)
851. Gerstein, M., Schulz, G., and Chothia, C.
(1993) J. Mol. Biol. 229, 494501
852. Bernstein, B. E., and Hol, W. G. J. (1998)
Biochemistry 37, 44294436
853. Matte, A., Tari, L. W., and Delbaere, L. T. J.
(1998) Structure 6, 413419
854. Vonrhein, C., Bnisch, H., Schfer, G., and
Schulz, G. E. (1998)
J. Mol. Biol. 282, 167179
855. Bernstein, B. E., Michels, P. A. M., and Hol, W.
G. J. (1997) Nature (London) 385, 275278
856. Yount, R. G., Babcock, D., Ballantyne, W., and
Ojala, D. (1971) Biochemistry 10, 24842489
857. Yount, R. G. (1975) Adv. Enzymol. 43, 156
858. McPhillips, T. M., Hsu, B. T., Sherman, M. A.,
Mas, M. T., and Rees, D. C. (1996) Biochemistry
35, 41184127
859. Schlauderer, G. J., Proba, K., and Schulz, G. E.
(1996) J. Mol. Biol. 256, 223227
860. Byeon, I.-J. L., Shi, Z., and Tsai, M.-D. (1995)
Biochemistry 34, 31723182
861. Ray, B. D., Chau, M. H., Fife, W. K., Jarori, G.
K., and Nageswara Rao, B. D. (1996)
Biochemistry 35, 72397246
862. Schlauderer, G. J., and Schulz, G. E. (1996)
Protein Sci. 5, 434441
863. Schricker, R., Magdolen, V., Strobel, G.,
Bogengruber, E., Breitenbach, M., and Bandlow,
W. (1995) J. Biol. Chem. 270, 3110331110
864. Teplyakov, A., Sebastiao, P., Obmolova, G.,
Perrakis, A., Brush, G. S., Bessman, M. J., and
Wilson, K. S. (1996) EMBO J. 15, 34873497
865. Zhang, Y., Li, Y., Wu, Y., and Yan, H. (1997) J.
Biol. Chem. 272, 2934329350
References
866. Mller-Dieckmann, H.-J., and Schulz, G. E.
(1995) J. Mol. Biol. 246, 522530
867. Schlichting, I., and Reinstein, J. (1997)
Biochemistry 36, 92909296
868. Runquist, J. A., Harrison, D. H. T., and Miziorko,
H. M. (1998) Biochemistry 37, 12211226
869. Morra, S., Chiadmi, M., LeBras, G., Lascu, I.,
and Janin, J. (1995) Biochemistry 34, 1106211070
870. Webb, P. A., Perisic, O., Mendola, C. E.,
Backer, J. M., and Williams, R. L. (1995) J. Mol.
Biol. 251, 574587
871. Abdulaev, N. G., Karaschuk, G. N., Ladner, J.
E., Kakuev, D. L., Yakhyaev, A. V., Tordova,
M., Gaidarov, I. O., Popov, V. I., Fujiwara, J.
H., Chinchilla, D., Eisenstrein, E., Gilliland, G.
L., and Ridge, K. D. (1998) Biochemistry 37,
1395813967
872. Mesnildrey, S., Agou, F., Karlsson, A., Bonne,
D. D., and Vron, M. (1998) J. Biol. Chem. 273,
44364442
873. Turano, A., Furey, W., Pletcher, J., Sax, M.,
Pike, D., and Kluger, R. (1982) J. Am. Chem.
Soc. 104, 30893095
874. Aleshin, A. E., Zeng, C., Bourenkov, G. P.,
Bartunik, H. D., Fromm, H. J., and Honzatko,
R. B. (1998) Structure 6, 3950
875. Aleshin, A. E., Zeng, C., Bartunik, H. D.,
Fromm, H. J., and Honzatko, R. B. (1998) J.
Mol. Biol. 282, 345357
876. Lundblad, V. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 84158416
877. Behlke, J., Heidrich, K., Naumann, M., Mller,
E.-C., Otto, A., Reuter, R., and Kriegel, T.
(1998) Biochemistry 37, 1198911995
878. Anderson, C. M., Stenkamp, R. E., and Steitz,
T. A. (1978) J. Mol. Biol. 123, 1523
879. Schwab, D. A., and Wilson, J. E. (1989) Proc.
Natl. Acad. Sci. U.S.A. 86, 25632567
880. Schirmer, T., and Evans, P. R. (1990) Nature
(London) 343, 140145
881. Auzat, I., Le Bras, G., and Garel, J.-R. (1995) J.
Mol. Biol. 246, 248253
882. Blake, C. C. F., and Evans, P. R. (1974) J. Mol.
Biol. 484, 585601
883. Berger, S. A., and Evans, P. R. (1992)
Biochemistry 31, 92379242
884. Mizuguchi, H., Cook, P. F., Hasemann, C. A.,
and Uyeda, K. (1997) Biochemistry 36, 8775
8784
885. Fujii, H., Krietsch, W. K. G., and Yoshida, A.
(1980) J. Biol. Chem. 255, 64216423
886. Tanaka, K. R., and Paglia, D. E. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease (Scriver, C. R., Beaudet, A. L., Sly, W.
S., and Valle, D., eds), pp. 34853511,
McGraw-Hill, New York
887. Mas, M. T., Bailey, J. M., and Resplandor, Z. E.
(1988) Biochemistry 27, 11681172
888. Forstner, M., Mller, A., Stolz, M., and
Wallimann, T. (1997) Protein Sci. 6, 331339
889. Fritz-Wolf, K., Schnyder, T., Wallimann, T.,
and Kabsch, W. (1996) Nature (London) 381,
341345
890. Zhou, G., Somasundaram, T., Blanc, E.,
Parthasarathy, G., Ellington, W. R., and
Chapman, M. S. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 84498454
891. Larsen, T. M., Benning, M. M., Rayment, I.,
and Reed, G. H. (1998) Biochemistry 37, 6247
6255
892. Loria, J. P., and Nowak, T. (1998) Biochemistry
37, 69676974
893. Lovell, S. C., Mullick, A. H., and Muirhead,
H. (1998) J. Mol. Biol. 276, 839851
894. Cheng, X., Friesen, R. H. E., and Lee, J. C.
(1996) J. Biol. Chem. 271, 63136321
894a. Valentini, G., Chiarelli, L., Fortin, R.,
Speranza, M. L., Galizzi, A., and Mattevi, A.
(2000) J. Biol. Chem. 275, 1814518152
895. Hunter, T., and Plowman, G. D. (1997) Trends
Biochem. Sci. 22, 1822
896. Grant, B. D., Hemmer, W., Tsigelny, I., Adams,
J. A., and Taylor, S. S. (1998) Biochemistry 37,
77087715
897. Cheng, X., Shaltiel, S., and Taylor, S. S. (1998)
Biochemistry 37, 1400514013
898. Engh, R. A., Girod, A., Kinzel, V., Huber, R.,
and Bossemeyer, D. (1996) J. Biol. Chem. 271,
2615726164
899. Herberg, F. W., Zimmermann, B., McGlone,
M., and Taylor, S. S. (1997) Protein Sci. 6, 569
579
900. Su, Y., Dostmann, W. R. G., Herberg, F. W.,
Durick, K., Xuong, N.-h, Eyck, L. T., Taylor, S.
S., and Varughese, K. I. (1995) Science 269,
807813
901. Lohmann, S. M., Vaandrager, A. B.,
Smolenski, A., Walter, U., and DeJonge, H. R.
(1997) Trends Biochem. Sci. 22, 307312
902. Xu, W., Harrison, S. C., and Eck, M. J. (1997)
Nature (London) 385, 595602
903. Sicheri, F., Moarefi, I., and Kuriyan, J. (1997)
Nature (London) 385, 602609
904. Adams, J. A. (1996) Biochemistry 35, 10949
10956
905. Tesmer, J. J. G., Sunahara, R. K., Gilman, A.
G., and Sprang, S. R. (1997) Science 278, 1907
1916
906. Tesmer, J. J. G., Sunahara, R. K., Johnson, R.
A., Gosselin, G., Gilman, A. G., and Sprang, S.
R. (1999) Science 285, 756760
907. Sunahara, R. K., Tesmer, J. J. G., Gilman, A. G.,
and Sprang, S. R. (1997) Science 278, 19431947
908. Zhao, Y., Schelvis, J. P. M., Babcock, G. T., and
Marletta, M. A. (1998) Biochemistry 37, 4502
4509
908a. Zhao, Y., Brandish, P. E., DiValentin, M.,
Schelvis, J. P. M., Babcock, G. T., and Marletta,
M. A. (2000) Biochemistry 39, 1084810854
909. Joyce, C. M., and Steitz, T. A. (1994) Ann. Rev.
Biochem. 63, 777822
910. Kim, Y., Eom, S. H., Wang, J., Lee, D.-S., Suh,
S. W., and Steitz, T. A. (1995) Nature (London)
376, 612616
911. Doubli, S., Tabor, S., Long, A. M.,
Richardson, C. C., and Ellenberger, T. (1998)
Nature (London) 391, 251258
912. Patel, P. H., Jacobo-Molina, A., Ding, J.,
Tantillo, C., Clark, A. D., Jr., Raag, R., Nanni,
R. G., Hughes, S. H., and Arnold, E. (1995)
Biochemistry 34, 53515363
913. Bartlett, P. A., and Eckstein, F. (1988) J. Biol.
Chem. 257, 88798884
913a. Stivers, J. T., Nawrot, B., Jagadeesh, G. J., Stec,
W. J., and Shuman, S. (2000) Biochemistry 39,
55615572
914. Smith, C. A., and Rayment, I. (1996) Biophys. J.
70, 15901602
915. Wittinghofer, A., and Pai, E. F. (1991) Trends
Biochem. Sci. 16, 382387
916. Coleman, D. E., and Sprang, S. R. (1998)
Biochemistry 37, 1437614385
917. Falany, C. N. (1997) FASEB J. 11, 206216
918. Varin, L., Marsolais, F., Richard, M., and
Rouleau, M. (1997) FASEB J. 11, 517 525
919. Kakuta, Y., Petrotchenko, E. V., Pedersen, L.
C., and Negishi, M. (1998) J. Biol. Chem. 273,
2732527330
920. Zhang, H., Varmalova, O., Vargas, F. M.,
Falany, C. N., and Leyh, T. S. (1998) J. Biol.
Chem. 273, 1088810892
920a. Hiraoka, N., Nakagawa, H., Ong, E., Akama,
T. O., Fukuda, M. N., and Fukuda, M. (2000) J.
Biol. Chem. 275, 20188 20196
921. Klaassen, C. D., and Boles, J. W. (1997) FASEB
J. 11, 404418
674 Chapter 12. Transferring Groups by Displacement Reactions
References
922. Neufeld, E. F., and Muenzer, J. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2465
2494, McGraw-Hill, New York
923. Lukatela, G., Krauss, N., Theis, K., Selmer, T.,
Gieselmann, V., von Figura, K., and Saenger,
W. (1998) Biochemistry 37, 36543664
923a. Dierks, T., Lecca, M. R., Schlotterhose, P.,
Schmidt, B., and von Figura, K. (1999) EMBO
J. 18, 20842091
924. Waldow, A., Schmidt, B., Dierks, T., von
Blow, R., and von Figura, K. (1999) J. Biol.
Chem. 274, 1228412288
924a. Szameit, C., Miech, C., Balleininger, M.,
Schmidt, B., von Figura, K., and Dierks, T.
(1999) J. Biol. Chem. 274, 1537515381
925. Fox, D. K., and Roseman, S. (1986) J. Biol.
Chem. 261, 1348713497
926. Joyce, M. A., Fraser, M. E., James, M. N. G.,
Bridger, W. A., and Wolodko, W. T. (2000)
Biochemistry 39, 1725
927. Johnson, J. D., Muhonen, W. W., and
Lambeth, D. O. (1998)
J. Biol. Chem. 273, 2757327579
928. Liaw, S.-H., and Eisenberg, D. (1994)
Biochemistry 33, 675681
929. Alibhai, M., and Villafranca, J. J. (1994)
Biochemistry 33, 682686
930. Meister, A. (1968) Adv. Enzymol. 31, 183 218
931. Meister, A. (1974) in The Enzymes, 3rd ed., Vol.
10 (Boyer, P. D., ed), pp. 669754, Academic
Press, New York
932. Whitty, A., Fierke, C. A., and Jencks, W. P.
(1995) Biochemistry 34, 1167811689
933. Rochet, J.-C., and Bridger, W. A. (1994) Protein
Sci. 3, 975981
934. Selmer, T., and Buckel, W. (1999) J. Biol. Chem.
274, 2077220778
675
1. Outline the reactions by which glyceraldehyde 3-
phosphate is converted to 3-phosphoglycerate
with coupled synthesis of ATP in the glycolysis
pathway. Show important mechanistic details.
2. Papain is a protein-hydrolyzing (proteolytic)
enzyme with an SH group and an imidazole
group at the active site. Write a reasonable
structure for a tetrahedral intermediate that
would be expected to arise during formation of an
acyl enzyme intermediate.
3. Adenylate kinase catalyzes the interconversion of
ATP, AMP, and ADP.
a) Draw a reasonable structure for a penta-
covalent intermediate derived from ATP and
AMP.
b) Draw a reasonable structure for the transition
state leading from ATP + AMP to two molecules
of ADP in an S
N
2-like reaction.
4. Penicillin inhibits a D-alanyl-D-alanine
transpeptidase that catalyzes the reaction
R-D-alanyl-D-alanine + H
2
N-R
R-D-alanyl-NH-R + D-alanine
where R and R are different parts of a bacterial
peptidoglycan. Write a step-by-step mechanism
for this reaction and indicate how penicillin may
inhibit the enzyme by combining with it irrevers-
ibly. See Strynodka et al., Nature 359, 700 705,
1992, for related reaction of penicillin with a
penicillinase.
Study Questions
5. Write out step-by-step chemical mechanisms for
the following enzymatic reaction. Use small
arrows to indicate directions of electron flow.
Remember to have all electrons move in the same
direction in any single structure.
Amino acid + tRNA + ATP
Aminoacyl-tRNA + AMP + PP
i
6. Trypsin in which Asp 102 has been replaced by
Asn has 10
4
times less catalytic activity than
natural trypsin at neutral pH. From the crystal
structure of the mutant enzyme it appears that the
imidazole group of His 57 is held by the Asn side
chain in the wrong tautomeric form for catalysis.
Explain. Compare this incorrect tautomeric form
with that in the initial structure shown in Fig. 12-11.
7. A recent discovery in biochemistry is that RNA
can act as an enzyme in chemical reactions,
usually reactions involving RNA hydrolysis.
Discuss the features of RNA structure that might
favor evolution of enzymes composed entirely of
a single polyribonucleotide chain, and describe a
proposed mechanism for RNA-catalyzed hydroly-
sis of RNA molecules.
N C C
H H
C N
O
CH
S
CH
3
CH
3
COO
Penicillin

H
C
O
R
676 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
A. Addition of ROH, RNH
2
, and RSH to
Polarized Double Bonds
1. Carbonic Anhydrase
2. Imines (Schiff Bases)
3. Stereochemistry of Addition to Trigonal Carbon
Atoms
4. Addition of CarbonCarbon Double Bonds, Often
Reversible Reactions
Enoyl-CoA hydratase
Glutathione S-transferases
Chlorobenzoyl-CoA dehalogenase
5. Addition to Double Bonds Adjacent to Carboxylate
Groups
Fumarate hydratase
Some other fumarate-forming reactions
Enolase
Pectate lyase and related enzymes
6. Aconitase and Related IronSulfur Proteins
7. Addition to or Formation of Isolated Double Bonds
8. Conjugative and Decarboxylative Elimination
Reactions
9. Isomerization Assisted by Addition
10. Reversibility of Addition and Elimination
Reactions
B. Enolic Intermediates in Enzymatic Reactions
1. Mandelate Racemase and Related Enzymes
2. Isomerases
Aldose ketose interconversions
Catalysis of ring opening by isomerases
Triose phosphate isomerase
Xylose isomerase and the hydride shift mechanism
3-Oxosteroid isomerases
4-Oxalocrotonate tautomerase
3. Internal OxidationReduction by Dehydration
of Dihydroxyacids
4. Formation and Metabolism of Methylglyoxal
(Pyruvaldehyde)
677
677
679
680
681
681
682
682
683
683
685
685
686
686
688
689
690
690
691
691
692
692
693
693
695
696
697
697
697
680
687
701
Boxes
Box 13-A Zinc
Box 13-B EPSP Synthase and the Herbicide
Glyphosate
Tables
Table 13-1 Products Arising from Reactions of
Acetyl-CoA with a Second Substrate
with Catalysis by a Polycarboxylate
Synthase
C. Beta Cleavage and Condensation
1. Displacement on a Carbonyl Group
2. Addition of an Enolate Anion to a Carbonyl
Group or an Imine; Aldolases
Polycarboxylic acid synthases
Citrate cleaving enzymes
3. Chiral Acetates and Their Use in Stereochemical
Studies
4. Addition of an Enolate Ion to Carbon Dioxide and
Decarboxylation
Decarboxylation of -oxoacids
Linked oxidation and decarboxylation
Phosphoenolpyruvate, a key metabolic intermediate
Ribulose bisphosphate carboxylase
Carbon dioxide or bicarbonate ion?
5. Incorporation of Bicarbonate into Carboxyl Groups
PEP mutase and the synthesis of phosphonates
D. Some Isomerization and Rearrangement
Reactions
References
Study Questions
698
698
699
700
703
704
705
705
705
705
706
710
711
711
712
713
717
Contents
T199
N244
H96
H94
H119
E117
Zn
Q92
N
N
N
N
N
N
C
C O
O
O O
O
H
H
Fe S
S
C
H
CH2 O
O
R580
H101
D100
N
H
H
N
O
H
Fe
S
Fe
Fe S
S
C424
C358
C421
S
R452
+

C H
+

O
Ser
+
R644

O
O
D568

+
O
O

S
R401
H
C
+
N
O
N
N
H
H
H
H
O
C

R329
N
N
N
H
H
H
H
H
C
+
O
C
C
O
O
C
D375 H274
R421
N
N
H
H
N
H
N H
H
O
O
C
C
H
H
O
S
CoA
C
H2N

One of the simplest biochemical addition reactions is the hydration of carbon


dioxide to form carbonic acid, which is released from the zinc-containing carbonic
anhydrase (left, Fig. 13-1) as HCO
3

. Aconitase (center, Fig. 13-4) is shown here


removing a water molecule from isocitrate, an intermediate compound in the citric
acid cycle. The H
2
O that is removed will become bonded to an iron atom of the
Fe
4
S
4
cluster at the active site as indicated by the black H
2
O. An enolate anion
derived from acetyl-CoA adds to the carbonyl group of oxaloacetate to form citrate
in the active site of citrate synthase (right, Fig. 13-9) to initiate the citric acid cycle.
677
Enzymatic Addition, Elimination, Condensation, and Isomerization:
Roles for Enolate and Carbocation Intermediates
13
In Chapter 12 we considered reactions by which
living cells are able to transfer groups from one mole-
cule to another using nucleophilic displacements. We
also showed how transfer reactions can be utilized by
ligases to join two molecules together with the Gibbs
energy of cleavage of ATP or of a related molecule
driving the reaction. In this chapter we will examine
addition reactions, which provide a simple way of
joining two molecules by means of CO, CN, CS, or
CC bonds. Among these are the aldol and Claisen-
type condensations by which CC bonds are formed.
We will also consider elimination reactions and decar-
boxylations, which are the reverse of addition and
condensation reactions, as well as mechanistically
related isomerizations. Many reactions of these types
occur in the major pathways of metabolism.
A. Addition of ROH, RNH
2
, and RSH to
Polarized Double Bonds
Next to nucleophilic displacement, the commonest
mechanistic processes in enzymatic catalysis are addi-
tion to double bonds and elimination to form double
bonds. These often involve addition of a nucleophile
together with a proton to a highly polarized double
bond such as C=O or C=N. In other reactions, which
are discussed in Section C,2, the nucleophile attacks one
end of a C=C bond that is polarized by conjugation
with C=O or C=N.
Alcohols, amines, and thiols add readily to the
electrophilic carbon of the carbonyl group to form
hemiacetals, carbinolamines, hemiketals, and
hemimercaptals. An example is the formation of ring
structures of sugars (Eq. 4-1). Water can also add to
carbonyl groups and most aliphatic carbonyl compounds
exist in water as an equilibrium mixture with a cova-
lent hydrate (Eq. 13-1). For example, acetaldehyde in
aqueous solution consists of a mixture of about 50%
free aldehyde and 50% hydrate in rapidly reversible
equilibrium
1,2
and formaldehyde is over 99.9%
hydrated.
3
Addition reactions often occur as parts of more
complex reactions. For example, a thiol group of
glyceraldehyde-3-phosphate dehydrogenase reacts
with the aldehyde substrate to form a hemimercaptal,
which is subsequently oxidized to a thioester (see Fig.
15-6).
1. Carbonic Anhydrase
Another simple addition reaction is the hydration
of CO
2
to form the bicarbonate ion. Without catalysis
the reaction may require several seconds,
4,5
the appar-
ent first-order rate constant being ~ 0.03 s
1
at 25C.
Cells must often hasten the process. The specific catalyst
carbonic anhydrase is widespread in its distribution
CH
3
C
O
H
CH
3
C
OH
H
OH + H
2
O
(13-1)
(13-2)
O C O HO C
OH
O
HO C
O

O
H
2
O H
+
T199
N244
H96
H94
H119
E117
Zn
Q92
N
N
N
N
N
N
C
C
O
O
O O
O
H
H
Fe S
S
C
H
CH
2 O
O
R580
H101
D100
N
H
H
N
O
H
Fe
S
Fe
Fe S
S
C424
C358
C421
S
R452
+

C H
+

O
Ser
+
R644

O
O
D568

+
O
O

S
R401
H
C
+
N
O
N
N
H
H
H
H
O
C

R329
N
N
N
H
H
H
H
H
C
+
O
C
C
O
O
C
D375 H274
R421
N
N
H
H
N
H
N
H
H
O
O
C
C
H
H
O
S
CoA
C
H
2
N

678 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
and is especially active in tissues (e.g., red blood cells,
and lungs) that are involved in respiration. One liter
of mammalian blood contains 1 2 g of this enzyme,
a monomeric 30-kDa protein containing ~ 260 amino
acids and one tightly bound ion of Zn
2+
. Erythrocytes
contain two isoenzymes (I and II) of carbonic anhydrase
and the human body contains at least eight distinct
isoenzymes (I VIII).
5 8
They are found wherever there
is a high demand for CO
2
or bicarbonate. Isoenzyme I,
II, III, and VII are cytosolic. Carbonic anhydrase I is
specific to erythrocytes, while isoenzyme II is present
in most cells. Hereditary lack of carbonic anhydrase II
is associated with osteopetrosis (marble bone disease),
a condition involving failure of bone resorption and
the calcification of other tissues.
9
The generation of
acidity according to Eq. 13-2 is presumably required
for dissolution of bone by the osteoclasts.
Isoenzymes III and VII have a more specialized
distribution. Carbonic anhydrase III is abundant in
adipocytes which use bicarbonate in fatty acid syn-
thesis.
7
Isoenzyme V is present in the mitochondrial
matrix and is also abundant in both adipocytes and
liver.
7,8
Isoenzyme IV is a larger membrane-associated
form, while VI is secreted into the saliva.
10
Carbonic
anhydrase has also been identified in E. coli.,
11
in a
methanobacterium,
12
and in green plants.
13,13a
A 60-kDa
carbonic anhydrase called nacrein is found in the
organic matrix of the nacreous layer of the pearl oyster,
the layer that forms aragonite (orthorhombic calcium
carbonate) in the shell and in pearls.
14
X-ray studies of carbonic anhydrases I and II, from
human blood, revealed that both have an ellipsoidal
shape of dimensions ~ 4.1 x 4.1 x 4.7 nm.
15,16
The zinc
atom in each molecule lies in a deep pocket ~ 1.2 nm
from the surface and is surrounded by three histidine
side chains and one H
2
O or OH

ion, the four ligands


forming a distorted tetrahedron (Fig. 13-1). The coor-
dinating imidazole group from His 119 is hydrogen
bonded to a carboxylate group of Glu 117, a feature
reminiscent of the charge-relay system of serine pro-
tease. This carboxylate group is also bound into a
more extended hydrogen-bonded network, part of
which is indicated in Fig. 13-1. The other imidazole
groups also form hydrogen bonds to protein groups
and the zinc-bound H
2
O is involved in an extensive
hydrogen-bonded network with several other bound
water molecules and protein side chains.
17a
Most of
these structural features are conserved in the other
mammalian isoenzymes.
5
However, from X-ray absorp-
tion spectroscopy (EXAFS) it appears that spinach
carbonic anhydrase contains one or more sulfur ligands
to the zinc,
13
while the enzymes from the archaeon
Methanosarcina thermophila have a left-handed helix
structure (see Figs. 2-17 and 13-3).
12
Nevertheless, this
enzyme has the same three-histidine Zn OH structure
found in the mammalian enzymes.
Carbonic anhydrase II is among the most rapid
enzymes known, with the turnover number at 25C
for hydration of CO
2
being ~ 10
6
s
1
. The same enzyme
catalyzes hydration of acetaldehyde (Eq. 13-1) but at a
1000-fold slower rate. A pK
a
of ~ 7 controls the activity.
This appears to represent the loss of H
+
from the Zn
2+

OH
2
complex
18
to give Zn
+
OH. The latter is in effect
a stabilized hydroxide ion existing at a pH at which
OH

is normally not present in quantity. It is this


hydroxide ion that adds to the CO
2
or to the aldehyde
substrate (Eq. 13-3, step a).
18 20
In step b a water molecule
replaces the departing bicarbonate.
17
A variety of data
indicate that proton transfers mediated by the enzyme
are essential parts of the carbonic anhydrase mecha-
nism.
18 19b,21 23
One proposal is that the nearby imida-
zole group of His 64 (not shown in Fig. 13-1) deproto-
nates the bound H
2
O via a hydrogen-bonded network
of bound water molecules (Eq. 13-3, step c). The side
chain of Thr 199 may function in a cyclic proton trans-
fer in step c.
19
The proton generated in step c is released
to the solvent in a process that is catalyzed by buffer
anions or by amines such as histamine. The latter
binds at the edge of the active site and forms an addi-
tional hydrogen-bonded pathway to the zinc-bound
H
2
O.
24
A different proton shuttle pathway has been
proposed for the slower carbonic anhydrase III.
25
Related to the reaction catalyzed by carbonic
anhydrase is the addition of an amino group to CO
2
(Eq. 2-21) to form a carbamino group ( NH COO

).
This reaction is essential to the functioning of hemo-
globin, which must carry large amounts of CO
2
, as
carbamino groups, from tissues to the lungs, (Eq. 7-47)
and to some enzymes such as ribulose bisphosphate
carboxylase (Figs. 13-10, 13-11).
Zn O
H
C
O
H
O
Thr
O
Zn O C
O
O
Zn OH
2
2

+
2+
2+
H
a
b
c
H
+
slow via shuttle
H
2
O
HCO
3

H
+

(13-3)
679 A. Addition of R OH, RNH
2
, and R SH to Polarized Double Bonds
Figure 13-1 (A) Stereoscopic
view showing the binding pocket
for HCO
3

in the active site of


human carbonic acid anhydrase I.
Also shown are the hydrogen-
bonded pairs E117 H119 and
E106 T199. ZNC, zinc ion; HCO,
bicarbonate. From Kumar and
Kannan.
16
(B) View of the active
site of carbonic anhydrase II. The
orientation is a little different than
in (A). The location of H
2
O or

OH
bound to the zinc ion is marked X.
Hydrogen bonds from the three
histidines that form coordinate
bonds to the zinc, Q92, E117, the
backbone carbonyl of N244, and
the hydrogen bond from the zinc-
bound hydroxyl to the T199 side
chain are shown as dashed lines.
From Kiefer et al.
17
A
B
2. Imines (Schiff Bases)
As we have seen already, many enzymatic reac-
tions depend upon formation of imines, which are
commonly called Schiff bases. The two-step formation
of Schiff bases consists of addition of an amino group
to a carbonyl group to form a carbinolamine followed
by elimination of water (Eq. 13-4).
26
One group of
aldolases (Section D) have, at their active centers,
lysine side chains which form Schiff bases with the
ketone substrates prior to the principal reaction of break-
ing or forming a C C bond. Similarly, the initial reac-
tion of the aldehyde coenzyme pyridoxal phosphate
with amino acid substrates is the formation of Schiff
R C
H
O H
H
2
N R'
R C
N
OH
H
R'
R C
H
N R'
Carbinolamine
Imine or Schiff base
H
H
2
O
+
a
b
(13-4)
bases (Fig. 14-4). Indeed, the groups C = O and H
2
N are
inherently complementary and their interaction through
imine formation is extremely common.
Schiff bases often form within a fraction of a sec-
ond, but one or both steps of Eq. 13-4 may require
catalysis to achieve enzymatic velocities.
27
The reac-
tion is usually completely reversible and formation
constants are often low enough that a carbonyl com-
pound present in small amounts will not react exten-
sively with an amine unless the two are brought
together on an enzyme surface. If the amino group
is hydrogen bonded to the enzyme, the proton may
remain on the Schiff base nitrogen, enhancing the
electron-accepting properties of the C=N group. The
pK
a
values of Schiff bases with aliphatic aldehydes are
usually 1 2 units lower than those of the corresponding
primary amines. However, the local environment may
sometimes cause the pK
a
to be increased. Most Schiff
bases are reduced readily by sodium borohydride or
sodium cyanoborohydride to form secondary amines
in which the original aldehydes are bound covalently
to the original amino groups (Eq. 3-34). This provides
a method for locating sites of Schiff base formation in
enzymes. An isotopically substituted aldehyde or
amine can be employed, or an isotopic label can be
introduced, by using
2
H- or
3
H-containing sodium
borohydride in the reduction.
T199
N244
H96
H94
H119
E117
Zn
Q92
N
N
N
N
N
N
680 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
BOX 13-A ZINC
The average human ingests 10 15 mg of zinc a
day.
a
Although it is poorly absorbed, the concentra-
tions of zinc in tissues are relatively high and the
metal plays an essential role in a multitude of en-
zymes. The total zinc content of a 70-kg person is
1.4 2.3 g. A typical tissue concentration of Zn
2+
is
0.3 0.5 mM; an unusually high content of ~15 mM
is found in the prostate gland.
Zinc ion is much more tightly bound to most
organic ligands than is Mg
2+
(Table 6-9). It has a
filled 3d shell and tends to form four ionic bonds
with a tetrahedral geometry, often with nitrogen-
or sulfur-containing ligands.
b
Unlike Mg
2+
, which
interacts rapidly and reversibly with enzymes, Zn
2+
tends to be tightly bound within over 300 metallo-
enzymes.
c e
A common feature is the surrounding
of the Zn
2+
at the active center by three imidazole
groups, the fourth coordination position being free
for interaction with substrate. The second nitrogen
of the imidazole ring in many instances is hydrogen
bonded to a main chain carbonyl group of the pep-
tide, a feature that is also shared by histidines in
other metalloproteins.
The most important chemical function of Zn
2+
in enzymes is probably that of a Lewis acid providing
a concentrated center of positive charge at a nucleo-
philic site on the substrate.
f
This role for Zn
2+
is
discussed for carboxypeptidases (Fig.12-16) and
thermolysin,
g
alkaline phosphatase (Fig. 12-23),
h
RNA polymerases, DNA polymerases, carbonic
anhydrase (Fig. 13-1),
i
class II aldolases (Fig. 13-7),
some alcohol dehydrogenases (Fig. 15-5), and super-
oxide dismutases (Fig.16-22). Zinc ions in enzymes
can often be replaced by Mn
2+
, Co
2+
, and other ions
with substantial retention of catalytic activity.
f,j
In addition to its function in catalysis, zinc often
plays an important structural role, e.g., in the zinc
finger transcriptional regulators (Fig. 5-38).
k
Zinc
ions bind to insulin and stabilize its hexameric
structure (Fig. 7-18).
l
Six Zn
2+
ions are present in
the hexagonal tail plate of the T-even bacteriophage
(Box 7-C) and appear to be essential for invasion of
bacteria.
m
In carnivores, the tapetum, the reflecting
layer behind the retina of the eye of many animals,
contains crystals of the Zn
2+
cysteine complex.
Since zinc ions have no color their presence has
often been overlooked. Zinc ions will doubtless be
found in many more places within cells. Zinc is
usually the major component of the bound metals in
the metallothioneins (Box 6-E). These small 6.6-kDa
proteins which contain ~ 33% cysteine and bind as
many as six ions of Cd
2+
, Hg
2+
, Cu
2+
, or Zn
2+
per
molecule are present in all animal tissues as well
as in plants and some bacteria.
j
From a nutritional viewpoint, Cu
2+
competes
with zinc ion, as does the very toxic Cd
2+
. The latter
accumulates in the cortex of the kidney. Dietary
cadmium in concentrations less than those found in
human kidneys shortens the lives of rats and mice.
However, some marine diatoms contain a cadmium-
dependent carbonic anhydrase.
n
Although zinc
deficiency was once regarded as unlikely in humans,
it is now recognized as occurring under a variety
of circumstances
o,p
and is well-known in domestic
animals.
q
Consumption of excessive amounts of
protein as well as alcoholism, malabsorption, sickle
cell anemia, and chronic kidney disease can all be
accompanied by zinc deficiency.
a
ODell, B. L., and Campbell, B. J. (1971) Comprehensive
Biochemistry 21, 179216
b
Bock, C. W., Katz, A. K., and Glusker, J. P. (1995) J. Am. Chem.
Soc. 117, 37543765
c
Berg, J. M., and Shi, Y. (1996) Science 271, 10811085
d
Vallee, B. L., and Auld, D. S. (1993) Biochemistry 32, 64936500
e
Coleman, J. E. (1992) Ann. Rev. Biochem. 61, 897946
f
Mildvan, A. S. (1974) Ann. Rev. Biochem. 43, 357399
g
Holland, D. R., Hausrath, A. C., Juers, D., and Matthews, B. W.
(1995) Protein Sci. 4, 19551965
h
Kimura, E., Kodama, Y., Koike, T., and Shiro, M. (1995) J. Am.
Chem. Soc. 117, 83048311
i
Lesburg, C. A., and Christianson, D. W. (1995) J. Am. Chem. Soc.
117, 68386844
j
Kagi, J. H. R., Himmelhoch, S. R., Whanger, P. D., Bethune, J. L.,
and Vallee, B. L. (1974) J. Biol. Chem. 249, 35373542
k
Berg, J. M. (1990) J. Biol. Chem. 265, 65136516
l
Hill, C. P., Dauter, Z., Dodson, E. J., Dodson, G. G., and Dunn,
M. F. (1991) Biochemistry 30, 917924
m
Kozloff, L. M., and Lute, M. (1977) J. Biol. Chem. 252, 77157724
n
Lane, T. W., and Morel, F. M. M. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 46274631
o
Prasad, A. S. (1984) Fed. Proc. 43, 28292834
p
Day, H. G. (1991) FASEB J. 5, 23152316
q
Luecke, R. W. (1984) Fed. Proc. 43, 28232828
3. Stereochemistry of Addition to Trigonal
Carbon Atoms
Adducts formed by enzymatic addition of nucleo-
philes to carbonyl groups are usually chiral. For example,
addition of an amino group to the front side of the
carbonyl carbon in Eq. 13-5 (the si face as defined in
Chapter 9) creates a carbinolamine of the S configura-
tion. Addition of the amino group from behind the
plane of the paper (the re face) would lead to a carbinol-
amine of the R configuration.
681
4. Addition to CarbonCarbon Double Bonds,
Often Reversible Reactions
Most of the reactions that we will consider in this
chapter involve addition of a proton to a carbon atom
or removal of a proton attached to a carbon atom. A
frequent metabolic reaction is addition of water to a
carboncarbon double bond that is conjugated with a
carbonyl group. This transmits the polarization of the
carbonyl group to a position located two carbon atoms
further along the chain.
Because of this effect bases can add to a carboncarbon
double bond in a position to the carbonyl group as
in Eq. 13-6, step a. The product of addition of HO

is
the anion of an enol in which the negative charge is
distributed by resonance between the oxygen of the
carbonyl group and the carbon adjacent to the carbonyl.
A stable product results if a proton adds to the latter
position (Eq. 13-6, step b). Many nucleophilic groups
(Nu in Eq. 13-6) other than HO

, may add, the reaction


being known as a Michael addition. If a neutral
nucleophile Nu adds, the product may lose a proton,
or transfer it to the adjacent carbon in Eq. 13-6, step b,
to give a neutral end product. The reverse of an
addition reaction of this type is also known as an
elimination reaction.
Enzymatic reactions involving addition to a C = C
bond adjacent to a carbonyl group (or in which elimi-
nation occurs , to a carbonyl) are numerous. Except
for some enzymes acting by free radical mechanisms,
the nucleophilic group always adds at the position
suggesting that the mechanism portrayed by Eq. 13-7
is probable. It is noteworthy that frequently in a meta-
bolic sequence a carbonyl group is deliberately introduced to
R
C
C
C
O R
C
C
C
O
R
C
C
C
O
+
+

R C C C O R C
HO
C C O

R C
HO
C C O
R C
HO
C C O
HO

Enolate anion
H
(Nu)
(Nu)
(Nu)
a
b
H
+
facilitate elimination or addition at adjacent carbon atoms.
The carbonyl may be formed by oxidation of a hydroxyl
group or it may be provided by a thioester formed
with coenzyme A or with an acyl carrier protein
(Chapter 14).
Enoyl-CoA hydratase. A specific example of
the reaction in Eq. 13-6 is the addition of water to
trans-,-unsaturated CoA derivatives (Eq. 13-7). It
is catalyzed by enoyl CoA hydratase (crotonase) from
mitochondria and is a step in the oxidation of fatty
acids (Fig. 10-4 ).
The enzyme is a hexamer, actually a dimer of trimers
made up of 291-residue polypeptide chains.
28
Aceto-
acetyl-CoA is a competitive inhibitor which binds into
the active site and locates it. From the X-ray structure
of the enzymeinhibitor complex it can be deduced
that the carboxylate group of E144 abstracts a proton
from a water molecule to provide the hydroxyl ion
that binds to the position (Eq. 13-6, step a) and that
the E164 carboxyl group donates a proton to the inter-
mediate enolate anion in step b.
28
The hydroxyl group
(13-6)
R
C
H
C
H
C
O
S CoA
R
C
H
CH
2
C
O
S CoA
HO
l--Hydroxyacyl-CoA
H
2
O
Enoyl-CoA hydratase
(13-7)
C
C
H
O
NH
2
C
C
H
HO
N
H
Addition from front
(re face of the carbonyl carbon)
S configuration
(13-5)
A. Addition of R OH, RNH
2
, and R SH to Polarized Double Bonds
682 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
and the proton enter from the same side of the double
bond in a syn addition. The X-ray structure shows
that the E144 and E164 side chains are on the same
side of the double bond, accounting for this stereo-
chemistry.
28,28a
Cinnamoyl-CoA thiol esters (R = phenyl in the foregoing
structure) contain a good light-absorbing group (chro-
mophore). Binding to the protein induces distinct
shifts in the ultraviolet absorption spectrum and also
in
13
C NMR and Raman spectra. These shifts suggest
that binding induces an enhanced positive charge at
C-3 and formation of a strong hydrogen bond to the
carbonyl group.
29
The X-ray studies indicate that this
bond is to the G141 peptide nitrogen as shown. These
results favor an enolate anion intermediate as in Eq.
13-6. However, kinetic isotope effects
30
as well as studies
of proton exchange have suggested a concerted mech-
anism with water adding at the same time that a pro-
ton binds at C-2.
3031a
A closely related E. coli protein is a 79-kDa multi-
functional enzyme that catalyzes four different reac-
tions of fatty acid oxidation (Chapter 17). The amino-
terminal region contains the enoyl hydratase activity.
32
A quite different enzyme catalyzes dehydration of
thioesters of -hydroxyacids such as 3-hydroxydecanoyl-
acyl carrier protein (see Eq. 21-2) to both form and
isomerize enoyl-ACP derivatives during synthesis of
unsaturated fatty acids by E. coli. Again, a glutamate
side chain is the catalytic base but an imidazole group
of histidine has also been implicated.
33
This enzyme
is inhibited irreversibly by the N-acetylcysteamine
thioester of 3-decynoic acids (Eq. 13-8). This was one
of the first enzyme-activated inhibitors to be studied.
34
Glutathione S-transferases. Addition of gluta-
thione (Box 11-B) to a large variety of different sub-
strates containing electrophilic centers, such as that at
the position in an ,-unsaturated ketone (Eq. 13-9),
is catalyzed by the ubiquitous group of enzymes called
glutathione S-transferases. There are six classes of
R
C
C
C
S CoA
O
H
H
H
N
G141
O
H
O
H
E144
C
O
O
H
O O
E164

2
3
eukayotic glutathione S-transferasesone membrane-
associated microsomal
35
and five cytosolic.
36 40
They
play an important role in detoxifying many xenobiotics
and other relatively hydrophobic compounds by con-
verting them to more soluble compounds that can be
degraded and excreted easily (see Box 11-B). A pair of
tyrosines appear to participate in catalysis, as is indi-
cated in Eq. 13-9.
38,41
Glutathione S-transferases have
attracted attention because of their role in detoxifying
anticancer drugs. Cancer cells can become resistant to
drugs as a result of excessive synthesis of these detoxi-
fying enzymes. At the same time glutathione trans-
ferases protect human patients from drugs. In plants
these enzymes may provide protection from insecti-
cides and herbicides.
41a
Glutathione transferases of
pathogenic organisms such as schistosomes are appro-
priate targets for new drugs as well as for vaccines.
42
Nonenzymatic Michael addition of glutathione to such
compounds as 4-hydroxynonenal, a product of per-
oxidation of the polyunsaturated arachidonate (see Eq.
21-15) are also biochemically important.
Chlorobenzoyl-CoA dehalogenase. The enzymatic
release of chloride ion from 4-chloroxybenzoyl-CoA
C C CH
2
C
S
O
O
C
N
CH
3
C C C
C
S
O
C
N
CH
3
H
N
N
H
H
H
O
H
(13-8)
Y6 O
H
N
H
S
Glutathione
CH
3
O
O
Y115
H H
O
CH
2
S209
CH
3
O
S Glutathione

Soluble product
H
+
(13-9)
683
would be hard to explain by a simple nucleophilic
displacement by HO

. However, addition of HO

,
followed by elimination of Cl

, would be chemically
reasonable. Nevertheless, single-turnover studies
revealed a more complex mechanism involving forma-
tion of a covalent adduct with the enzyme.
43,44
A car-
boxylate group adds and Cl

is eliminated to form of
an oxygen ester, which is then hydrolyzed to the final
product (Eq. 13-10). Studies of mutant enzymes
45
together with X-ray crystallography
46
support this
mechanism. As with enoyl-CoA hydratase, binding of
substrate to chlorobenzoyl-CoA dehalogenase causes
alterations in ultraviolet, NMR, and Raman spectra
that can be interpreted as indicating enhanced polar-
ization of the benzoyl group.
47,48
For example, changes
in the C= O stretching frequency suggest that the bond
is elongated, presumably as a result of hydrogen
bonding to the N H of G114, which lies at the N ter-
minus of an helix and experiences the additional
polarizing effect of the helix dipole (Fig. 2-20).
48,49
This effect can be compared with that mentioned
previously for enoyl-CoA hydratase.
In fact, close sequence and structural homologies show
that chlorobenzoyl-CoA dehalogenase, enoyl-CoA
hydratase, and a variety of other hydratases, isomerases,
synthases, lyases, and hydrolases belong to a large
family of related proteins.
49a,49b,49c
Cl
C
O O
Cl
C
O S CoA
O
C
O
D145
Cl
C
O S CoA
O
C
O
D145
C
O S CoA
O
C
O
D145

ATP ADP + P
i
CoAS
C
O S

CoA
Cl
OH

H
2
O
(13-10)
5. Addition to Double Bonds Adjacent to
Carboxylate Groups
Biochemical reactions often involve addition to
C=C bonds that are not conjugated with a true carbonyl
group but with the poorer electron acceptor COO

.
While held on an enzyme a carboxylate group may
be protonated, making it a better electron acceptor.
Nevertheless, there has been some doubt as to whether
the carbanion mechanism of Eq. 13-6 holds for these
enzymes. Some experimental data suggested a quite
different mechanism, one that has been established for
the nonenzymatic hydration of alkenes. An example
is the hydration of ethylene by hot water with dilute
sulfuric acid as a catalyst (Eq. 13-11), an industrial
method of preparation of ethanol. The electrons of the
double bond form the point of attack by a proton, and
the resulting carbocation readily abstracts a hydroxyl
ion from water. Direct addition of OH

to ethylene to
form a carbanion is not favored, because there is no
adjacent carbonyl group to stabilize the negative
charge.
Fumarate hydratase. The most studied enzyme
of this group is probably the porcine mitochondrial
fumarate hydratase (fumarase; see also Chapter 9),
a tetramer of 48.5-kDa subunits
50
with a turnover
number of ~ 2 x 10
3
s
1
. It accelerates the hydration
reaction more than 10
15
-fold.
51
A similar enzyme, the
467-residue fumarase C whose three-dimensional
structure is known,
52,53
is found in cells of E. coli when
grown aerobically. The product of the fumarate hydra-
tase reaction is L-malate (S-malate). The stereospecificity
is extremely high. If the reaction is carried out in
2
H
2
O
an atom of
2
H is incorporated into the pro-R position,
i.e., the proton is added strictly from the re face of the
trigonal carbon (Eq. 13-12). To obtain L-malate the
hydroxyl must have been added from the opposite
side of the double bond. Such anti (trans) addition is
much more common in both nonenzymatic and enzy-
matic reactions than is addition of both H and OH
(or Y) from the same side (syn, cis, or adjacent addi-
tion).
54
For concerted addition it is a natural result of
stereoelectronic control. Almost all enzymatic addition
and elimination reactions involving free carboxylic
acids are anti with the proton entering from the re face,
C C
H
H
H
H
H
+
C
H
H
H
3
C
+
Carbocation
H
3
C CH
2
OH
H
2
O
H
+
(13-11)
A. Addition of R OH, RNH
2
, and R SH to Polarized Double Bonds
684 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
suggesting that there has been conservation through-
out evolution of a single basically similar mecha-
nism.
54,55
The pH dependence of the action of fumarate
hydratase indicates participation of both an acidic and
a basic group with pK
a
values of 5.8 and 7.1.
56
See
Chapter 9 for additional information. However, either
anion or carbocation mechanisms might be possible.
That the cleavage of the C H bond is not rate limiting
is suggested by the observation that malate containing
2
H in the pro-R position is dehydrated at the same rate
as ordinary malate. If the anion mechanism (Eq. 13-13)
is correct, the
2
H from the pro-R position of specifically
labeled malate might be removed rapidly, while the
loss of OH

could be slower. If so, the


2
H would be
washed out of L-malate faster than could happen
by conversion to fumarate followed by rehydration
to malate. In fact, the opposite was observed.
The hydroxyl group was lost rapidly and the
2
H more
slowly. This result suggested the carbocation ion
mechanism of Eq. 13-14.
While no primary isotope rate effect was observed,
when the hydrogen at C-2 or the pro-S hydrogen at C-3
of malate was replaced by
2
H or
3
H distinct secondary
isotope effects were seen. Thus, k(
1
H) / k(
2
H) = 1.09
for both the pro-S and the C-3 hydrogen atoms.
57
These findings appeared to support the carbocation
mechanism for the reasons considered in Chapter 12
and suggested that step b of Eq. 13-14 is rate limiting.
The relative values of V
max
for hydration of fumarate,
fluorofumarate, and difluorofumarate (104, 410, and 86
mol ml
1
min
1
mg
1
) also seemed to support the car-
bocation mechanism.
58
However, it has become clear that protons removed
from a substrate to a basic group in a protein need not
exchange rapidly with solvent (see Eq. 9-102). In fact,
the proton removed by fumarate hydratase from malate
is held by the enzyme for relatively long periods of
time. Its rate of exchange between malate and solvent
is slower than the exchange of a bound fumarate ion
on the enzyme surface with another substrate molecule
from the medium.
59
Thus, the overall rate is deter-
mined by the speed of dissociation of products from the
enzyme and we cannot yet decide whether removal of
a proton precedes or follows loss of OH

.
Two new lines of evidence suggest that proton
abstraction comes first. A careful study of both
18
O
and
2
H isotope effects
60
supports the carbanion inter-
mediate, as does the strong inhibition by anions of
3-nitropropionate and 3-nitro-2-hydroxypropionate.
61
To provide a good electron sink the carboxylate group
adjacent to the proton that is removed by fumarate
hydratase must either be actually protonated in the
enzymesubstrate (ES) complex or paired with and
hydrogen bonded to a positively charged group.
Carbanion
Fumarate
H
C C
C
l-Malate
H
+
aci-Carboxylate
resonance form
O
O
OOC
HO
C C
C
H
H
2
O
O
O
H
OOC
HO
H
H
+

(13-13)
C C
COO
H
H
OOC
*H
OH
Carbocation
l-Malate
*H
Fumarate
+
+

a
b
(13-14)
C C
H
H
N
N
H
H
C NH Protein
H
Y
or M
+
2
R

H
O
O
C C
H
H
N
N
H
H
C NH Protein +
R
H
O
O

Carbanion Guanidinium
group
aci-Carboxylate
resonance form
+
C C
COO
H
H
OOC
*H
C C
COO
H
HO
OOC
OH
*H
Hydroxyl added
in front
(si face)
H
Fumarate
l-Malate
Proton added
from behind
(re face)

(13-12)
685
The anion formed by removal of the 3-H is analogous
to the enolate anion of Eq. 13-6 and has a strong struc-
tural similarity to the readily formed anions of organic
nitro compounds. The nitronate anions may, perhaps,
be regarded as transition state inhibitors.
There is still a third possible mechanism for the
fumarate hydratase reaction. The proton and hydroxyl
groups may be added simultaneously in a concerted
reaction.
62
However, observed kinetic isotope effects
are not consistent with this mechanism.
60
In 1997 the
structure of fumarase C of E. coli was reported.
52,53
Each active site of the tetrameric enzyme is formed
using side chains from three different subunits. The
H188 imidazole is hydrogen bonded to an active site
water molecule and is backed up by the E331 carboxy-
late which forms a familiar catalytic pair. However,
these results have not clarified the exact mode of
substrate binding nor the details of the catalytic mech-
anism. Structural studies of fumarate hydratase from
yeast
53a
and the pig
53b
are also in progress.
Some other fumarate-forming reactions.
Other enzymes that catalyze elimination reactions
that produce fumarate are aspartate ammonia-lyase
(aspartase),
63
argininosuccinate lyase (Fig. 24-10,
reaction g),
64,65
and adenylosuccinate lyase
(Fig. 25-15). In every case it is NH
3
or an
amine, rather than an OH group, that is
eliminated. However, the mechanisms
probably resemble that of fumarate hydra-
tase. Sequence analysis indicated that all of
these enzymes belong to a single fumarase-
aspartase family.
64,65
The three-dimensional
structure of aspartate ammonia-lyase resem-
bles that of fumarate hydratase, but the
catalytic site lacks the essential H188 of
fumarate hydratase. However, the pK
a
val-
ues deduced from the pH dependence of
V
max
are similar to those for fumarase.
64
3-Methylaspartate lyase catalyzes the same
kind of reaction to produce ammonia plus
cis-mesaconate.
63
Its sequence is not related
to that of fumarase and it may contain a
dehydroalanine residue (Chapter 14).
66
R C
N
O
O
H

Nitronate anion
R C
N
O
O
H

C C
H
OOC
COO
CH
3

cis- Mesaconate
Enolase. A key reaction in the metabolism of
sugars is the dehydration of 2-phosphoglycerate to
formphosphoenolpyruvate (PEP), the phospho
derivative of the enolic form of pyruvic acid:
A carbanionic intermediate has often been suggested
for this enzyme.
67,68
However, despite measurements
of kinetic isotope effects and many other experiments
it has been difficult to establish a detailed mecha-
nism.
68
Enolase has a complex metal ion requirement,
68a,69
usually met by Mg
2+
and Mn
2+
. From NMR studies of
the relaxation of water protons, it was concluded that
a Mn
2+
ion coordinates two rapidly exchangeable
water molecules in the free enzyme. When substrate
binds, one of these water molecules may be immobi-
lized and may participate in an addition reaction that
forms phosphoenolpyruvate (reverse of reaction 13-15).
A tightly bound conformational metal ion is located
in the known three-dimensional structure in such a
H
2
C C
HO COO
O
H
PO
3
2
H
2
C
2-Phosphoglycerate
C
H
2
O
COO
OPO
3
2
Mg
Phosphoenolpyruvate

+
or Mn
2 + 2
(13-15)
Figure 13-2 View of the active site of yeast enolase containing a
bound molecule of 2-phospho-D-glycerate. The catalytic magnesium
ion is at the left but the conformational metal is not visible here.
The imidazole group of His 159 serves as the catalytic base and the
NH
3
+
of Lys 396 or Lys 345
73b
as the catalytic acid. From Vinarov
and Nowak.
69
K396
E295
O
O
D320
2+
Metal site I
O
O O
O

O
O
O
P
O
H
OH
H159
N
H
N
Loop L2
O
O
O
O
E211
E168

N
+
2-PGA
M

H
H
H

A. Addition of R OH, RNH


2
, and R SH to Polarized Double Bonds
686 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
way that it might function in this manner. A more
loosely bound catalytic metal ion is also essential.
69 72
Figure 13-2 shows a view of the active site of yeast
enolase occupied by a molecule of bound 2-phospho-
glycerate. Histidine 159 is probably the catalytic base
that removes the -proton to form an aci-anion which
is stabilized by interaction with the catalytic Mg
2+
ion.
A protonated lysine 396 amino group may be the
catalytic acid.
68a,69
The active site is surrounded by a
complex hydrogen-bonded network.
73
As is discussed
in Section B, a large number of other enzymes belong
to an enolase superfamily of enzymes. Among them
is glucarate dehydratase, which initiates a pathway
for catabolism of D-glucarate and galactarate in E.
coli.
73a
Pectate lyase and related enzymes. A group of
polysaccharide lyases cleave the chains of polymers
of uronic acids with 1,4 linkages such as pectins, hyal-
uronan, heparin,
74,75
and dermatan sulfate (Fig. 4-11).
These bacterial enzymes also employ an elimination
mechanism.
76
The geometry of the -linked galactur-
onic acid units of pectin is favorable for anti elimina-
tion of the 5-H and the O-glycosyl group in the 4
position (Eq. 13-16). However, the corresponding
hyaluronate lyase (hyaluronidase) acting on glucu-
ronic acid residues causes a syn elimination. These
results suggest the formation of anionic intermediates
which can eliminate a substituent from either the
equatorial or axial position of the sugar ring. Hyal-
uronate lyase from the pathogenic Streptococcus pneu-
moniae apparently utilizes an imidazole group as the
catalytic base and a tyrosine side chain as the proton
donor in the reaction.
76a,76b
Pectate lyase C from the plant pathogen Erwinia,
which causes soft-rot in many different plants, has a
parallel barrel structure (Fig. 13-3)
77
which is similar
to that of the tailspike protein shown in Fig. 2-17 and
represents what may be a very large structural family
of proteins.
78
The location of the active site is not
(13-16)
Figure 13-3 The three-dimensional structure of pectate
lyase C showing locations of amino acids substituted by
oligonucleotide-directed mutation of the cloned gene.
The green dot labeled Ca is the Ca
2+
-binding site.
79
Courtesy
of Frances Jurnak.
O
OH
C
HO
O
O OH
O
O
H
H
O
OH
HO
O
C
OH O
4
5
+
OH
O
Galacturonic acid unit of pectin
obvious from the structure but on the basis of the
effects of a large number of mutations, it is thought
to be near the Ca
2+
- binding site labeled in Fig. 13-3.
Replacement of any of three Asp or Glu and three Lys
residues in this region leads to loss of or reduction in
catalytic activity.
79
6. Aconitase and Related Iron Sulfur Proteins
Two consecutive reactions of the citric acid cycle
(Fig. 10-6), the dehydration of citrate to form cis-aconi-
tate and the rehydration in a different way to form
isocitrate (Eq. 13-17), are catalyzed by aconitase (aconi-
tate hydratase). Both reactions are completely stereo-
specific. In the first (Eq. 13-17, step a), the pro-R proton
from C-4 (stereochemical numbering) of citrate is
removed and in step c isocitrate is formed. Proton
addition is to the re face in both cases.
As with fumarate hydratase, the enzyme holds the
abstracted proton (for up to 7 x 10
5
s) long enough so
that a cis-aconitate molecule sometimes diffuses from
the enzyme and (if excess cis-aconitate is present) is
replaced by another. The result is that the new cis-
aconitate molecule sometimes receives the proton
(intermolecular proton transfer). The proton removed
687
BOX 13-B EPSP SYNTHASE AND THE HERBICIDE GLYPHOSATE
The reversible reaction of phosphoenolpyruvate
(PEP) with shikimate 3-phosphate is a step in the
synthesis of the aromatic amino acids (see Fig. 25-1).
The chemical mechanism indicated
was proposed by Leaven and Sprinson in 1964.
a
Step a is unusual
b
because it involves protonation
on the methylene carbon of PEP and addition of a
nucleophile at C-2, the opposite of the addition in
the enolase reaction (Eq. 13-15, reverse). It is likely
that formation of a cationic intermediate precedes
that of the adduct shown. However, the structure
of the adduct has been confirmed by isolation from
the active site and by synthesis
c
and it has been
observed in the active site by NMR spectroscopy.
d
The three-dimensional structure of EPSP synthase is
known.
e,m
It is inhibited by the commercial herbi-
cide glyphosate [N-(phosphonomethyl) glycine]
whose structure is somewhat similar to that of the
proposed carbocation that arises from PEP. The
inhibitor (Z)-3-fluorophosphoenolpyruvate is a
pseudosubstrate that reacts in step a to give a stable
adduct unable to go through step b to form a
product.
f h
Glyphosate was for many years viewed
as a transition-state analog but more recently has
been shown to be a tight-binding noncompetitive
inhibitor.
i
a
Levin, J. G., and Sprinson, D. B. (1964) J. Biol. Chem. 239, 1142
1150
b
Barlow, P. N., Appleyard, R. J., Wilson, B. J. O., and Evans, J.
N. S. (1989) Biochemistry 28, 79857991
c
Anderson, K. S., Sikorski, J. A., Benesi, A. J., and Johnson, K.
A. (1988) J. Am. Chem. Soc. 110, 65776579
d
Appleyard, R. J., Shuttleworth, W. A., and Evans, J. N. S.
(1994) Biochemistry 33, 68126821
e
Stallings, W. C., Abdel-Meguid, S. S., Lim, L. W., Shieh, H.-S.,
Dayringer, H. E., Leimgruber, N. K., Stegeman, R. A.,
Anderson, K. S., Sikorski, J. A., Padgette, S. R., and Kishore,
G. M. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 50465050
f
Alberg, D. G., Lauhon, C. T., Nyfeler, R., Fssler, A., and
Bartlett, P. A. (1992) J. Am. Chem. Soc. 114, 35353546
g
Walker, M. C., Jones, C. R., Somerville, R. L., and Sikorski, J.
A. (1992) J. Am. Chem. Soc. 114, 76017603
h
Ream, J. E., Yuen, H. K., Frazier, R. B., and Sikorski, J. A.
(1992) Biochemistry 31, 55285534
i
Sammons, R. D., Gruys, K. J., Anderson, K. S., Johnson, K. A.,
and Sikorski, J. A. (1995) Biochemistry 34, 64336440
j Samland, A. K., Amrhein, N., and Macheroux, P. (1999)
Biochemistry 38, 1316213169
k
Skarzynski, T., Kim, D. H., Lees, W. J., Walsh, C. T., and
Duncan, K. (1998) Biochemistry 37, 25722577
l
Jia, Y., Lu, Z., Huang, K., Herzberg, O., and Dunaway-Mariano,
D. (1999) Biochemistry 38, 1416514173
m
Lewis, J., Johnson, K. A., and Anderson, K. S. (1999) Biochemistry
38, 73727379
a
b
P
i
PEP
HO
O
COO
3
H
O P
Shikimate 3-phosphate

H
2
C C
O
COO
P H

+
HO
O
COO
O P

C
O
COO
C H
H
H
HO
O
COO
O P

C
COO
H
2
C

5-Enolpyruvoylshikimate-3-phosphate (EPSP)
Adduct
P
A related mechanism is utilized in the biosyn-
thesis of UDP-muramic acid (Eq. 20-6).
j
There is an
enolpyruvoyl adduct analogous to that of EPSP
synthase; a proposed enolpyruvoyl-enzyme adduct
with Cys 115 is not on the major path.
k,l
However,
this enzyme is not inhibited by glyphosate.
i
H
3
C C
O
COO
PO
3
2
H
2
N
CH
2
CH
2
PO
3
2
+
COO
C C
O
COO

Carbocation
PO
3
2
+
Glyphosate

H
F

(Z)-3-Fluorophosphonoenolypyruvate
A. Addition of R OH, RNH
2
, and R SH to Polarized Double Bonds
688 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
Figure 13-4 (A) Stereoscopic view of the active site of
mitochondrial aconitase with a molecule of L-isocitrate
placed by modeling next to the Fe
4
S
4
cluster. This cluster
still has a water molecule bound to one iron atom as in the
free enzyme.
83
Courtesy of C. D. Stout. (B) Interpretive
diagram.
C
C
O
O
O
O
O
H
H
Fe
S
S
C
H
CH
2
O
O
R580
H101
D100
N
H
H
N
O
H
Fe
S
Fe
Fe S
S
C424
C358
C421
S
R452
+

C
H
+

O
Ser
+
R644

O
O
D568

+
O
O

S
a
C
C
O
C O
O
H
R
C
H
S
O
O
Fe
C
O
O
3
5
B
HB
1
+
H
C
C
O
C CH
2
O
Fe
C
O
O
H
H
O
H
C
O
O
BH

Citrate
R580
cis-Aconitate

1, 5
5, 1
+ +

b
Aconitate molecule flips 180
C
C
O
C O
O
Fe
H
B
CH
2
C
O
O
C
O
O
H
H
c
C
C
O
C H
O
Fe
O
H
CH
2
C
O
O
BH
O
O

HB
+
1
5

cis-Aconitate
+

2R, 3S-Isocitrate
+

5, 1
1, 5
3
3
(13-17)
from citrate is often returned to the molecule
in Eq. 13-17, step b, but the position of
reentry is different from that of removal.
Apparently, after the initial proton removal,
the cis-aconitate that is formed flips over
so that it can be rehydrated (Eq. 13-17, step
c) with participation of the same groups
involved in dehydration but with formation
of the new product.
80,80a
Aconitase contains iron in the form of
an Fe
4
S
4
iron-sulfur cluster (Fig. 13-4).
8183
However, the enzyme is usually isolated in
a form that does not show its maximum
activity until it has been incubated with
ferrous iron (Fe
2+
). The inactive form of the
enzyme is thought to contain an Fe
3
S
4
cluster (Chapter 16) which is converted
back to the Fe
4
S
4
cluster by the incubation
A
B
689
with Fe
2+
.
84 87
From Mssbauer spectroscopy it was
deduced that the iron atom that is lost upon inactiva-
tion is the binding site for the OH group of citrate or
isocitrate and of an adjacent carboxylate.
87
Since iron
can engage in oxidationreduction processes, aconi-
tase could act by a mechanism different from others
discussed here. However, an Fe OH group could
act much as does the Zn OH of carbonic anhydrase
(Eq. 13-4). Any redox chemistry may be involved in
control of the enzyme.
X-ray studies have confirmed the proximity of the
Fe
4
S
4
cluster to bound substrate analogs.
83,88
The iso-
citrate shown in Fig. 13-4 was fitted into the active site
by modeling. It shows the water molecule bound to
the Fe
4
S
4
cluster as observed for the free enzyme.
83
Notice that this cluster is held by three cysteine side
chains (C358, C421, and C424) rather than the usual
four (Chapter 16). The fourth iron atom is free to bind
to water. When the enzyme acts on isocitrate this
water must be displaced by the one generated from
the substrate. The Fe probably acts as a polarizing
electrophile that assists in the elimination but it is
apparently the histidineaspartate pair (H101 D100)
that serves as the catalytic acid in generating the H
2
O.
What is the catalytic base? The only candidate seen in
the structure is the Ser 642 OH, dissociated to O

.
Can this be correct? Mutational analysis supports
the essential role of this side chain.
80a
The peptide
NH and guanidinium groups of R644 appear to pro-
vide an oxyanion hole (Chapter 12) that stabilizes
the negative charge.
86
Mutations also support the
role of the His-Asp pair.
88,89
Another HisAsp pair
(H167 D165) is located directly behind the bound
substrate and Fe OH
2
in Fig. 13-4A and is apparently
also essential.
88
Aconitase exists as both mitochondrial and cyto-
solic isoenzyme forms of similar structure. However,
the cytosolic isoenzyme has a second function. In its
apoenzyme form, which lacks the ironsulfur cluster,
it acts as the much-studied iron regulatory factor,
or iron-responsive element binding protein (IRE-BP).
This protein binds to a specific stem-loop structure in
the messenger RNA for proteins involved in iron
transport and storage (Chapter 28).
86,90
Other enzymes in the aconitase family include
isopropylmalate isomerase and homoaconitase
enzymes functioning in the chain elongation pathways
to leucine and lysine, both of which are pictured in
Fig. 17-18.
90
There are also ironsulfur dehydratases,
some of which may function by a mechanism similar
to that of aconitase. Among these are the two fumarate
hydratases, fumarases A and B, which are formed in
place of fumarase C by cells of E. coli growing anaero-
bically.
91,92
Also related may be bacterial L-serine and
L-threonine dehydratases. These function without
the coenzyme pyridoxal phosphate (Chapter 14)
but contain ironsulfur centers.
93 95
A lactyl-CoA
dehydratase and some related iron-sulfur enzymes
(pp. 861-862) may act via a mechanism related to that
of vitamin B
12
.
7. Addition to or Formation of Isolated
Double Bonds
Only a few examples are known in which an
enzyme induces addition to a double bond that is not
conjugated with a carbonyl or carboxyl group. Pseudo-
monads have been observed to catalyze stereospecific
hydration of oleic acid to D-10-hydroxystearate.
96
The
addition is anti and the proton enters from the re face.
8. Conjugative and Decarboxylative
Elimination Reactions
Elimination can occur if the electrophilic and
nucleophilic groups to be removed are located not on
adjacent carbon atoms but rather are separated from
each other by a pair of atoms joined by a double bond.
Such a conjugative elimination of phosphate is the last
step in the biosynthesis of chorismate (Eq. 13-18, step a).
Chorismate is converted to prephenate (see Box 9-E)
which undergoes a conjugative and decarboxylative
elimination (Eq. 13-18, step c) with loss of both water
and CO
2
to form phenylpyruvate, the immediate bio-
synthetic precursor of phenylalanine. These reactions
provide a good example of how elimination reactions
can be used to generate aromatic groups. In fact, this is the
usual method of synthesis of aromatic rings in nature.
Notice the stereochemistry of Eq. 13-18, step a.
Orbital interaction rules predict that if the elimination
is a concerted process it should be syn. The observed
anti elimination suggests a more complex mechanism
involving participation of a nucleophilic group of the
enzyme.
97
Elimination usually involves loss of a proton
together with a nucleophilic group such as OH,
NH
3
+
, phosphate, or pyrophosphate. However, as in
Eq. 13-18, step c, electrophilic groups such as COO

can replace the proton. Another example is the con-


version of mevalonic acid-5-pyrophosphate to
isopentenyl pyrophosphate (Eq. 13-19): This is a key
reaction in the biosynthesis of isoprenoid compounds
such as cholesterol and vitamin A (Chapter 22). The
phosphate ester formed in step a is a probable inter-
mediate and the reaction probably involves a carbo-
cationic intermediate generated by the loss of phosphate
prior to the decarboxylation.
A. Addition of R OH, RNH
2
, and R SH to Polarized Double Bonds
690 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
a
b
c
C
O
OOC CH
2
H COO
H
OPO
3
2
HO
H

H
H
H
HO
CH
2
C
C
COO
O
O
O

H
+
5-Enoylpyruvylshikimate-3-P
CH
2
C
COO
O

P
i
H
2
O, CO
2
Phenylpyruvate Phenylalanine
OH
O
C
OOC
CH
2
COO

Chorismate mutase
(See Box 9-E)
Chorismate
Prephenate
(13-18)
9. Isomerization Assisted by Addition
An interesting use is made of addition to a double
bond by glutathione-dependent cistrans isomerases.
76
One of them converts maleate to fumarate with a
turnover number of 300 s
1
. Similar enzymes, which
participate in bacterial breakdown of aromatic com-
pounds (Fig. 25-7), isomerize maleylacetoacetate and
maleylpyruvate to the corresponding fumaryl deriva-
tives (Eq. 13-20). The SH group of bound glutathione
is thought to add to the double bond. Rotation can
then occur in the enolic intermediate. Thiocyanate ion
catalyzes the isomerization of maleic acid nonenzy-
matically, presumably by a similar mechanism.
10. Reversibility of Addition and Elimination
Reactions
Many addition and elimination reactions, e.g., the
hydration of aldehydes and ketones, and reactions
catalyzed by lyases such as fumarate hydratase are
strictly reversible. However, biosynthetic sequences
are often nearly irreversible because of the elimination
of inorganic phosphate or pyrophosphate ions. Both
of these ions occur in low concentrations within cells
so that the reverse reaction does not tend to take place.
In decarboxylative eliminations, carbon dioxide is
produced and reversal becomes unlikely because of
the high stability of CO
2
. Further irreversibility is
introduced when the major product is an aromatic
ring, as in the formation of phenylpyruvate.
O
P
O
P
O
COO
OH
O
O O
HO CH
3
P P O
C
O
O
CH
3
O
P
O
HO
O
O CH
2
CH
3

Mevalonic acid diphosphate

P P
a
b
P
i
, CO
2
Isopentyl pyrophosphate (13-19)
COO
O O
OOC
H
G S
COO
OH O
OOC
+
Glutathione
G S

COO
O O
OOC
Maleylpyruvate

Rotation is possible in
this enolic intermediate
G SH

Fumarylpyruvate

(13-20)
691
B. Enolic Intermediates in Enzymatic
Reactions
Enzymologists have freely proposed enolate anions,
enols, and enamines as intermediates for many years.
Such intermediates have been demonstrated for some
nonenzymatic acid- or base-catalyzed reactions, but
how can enzymes form enolates at pH 7 without the
use of strong acids or bases? The microscopic pK
a
value of an -hydrogen in a ketone or aldehyde is
about 17 20.
72,98,99
It will be similar for an acyl-CoA. However, for a
carboxylic acid, protonated on oxygen, the pK
a
will be
much higher (~ 22 25) and for carboxylate anions
even higher (~ 29 32).
100
1. Mandelate Racemase and Related
Enzymes
The degradation of mandelic acid by the bacterium
Pseudomonas putida (Chapter 25) is initiated by mandalate
racemase, another (/)
8
-barrel protein.
101
X-ray
structures of bound inhibitors together with modeling
suggest that the side chain of Lys 264 is the catalytic
base that abstracts the -H from S-mandelate (Fig. 13-5)
and that the catalytic pair of His 297 and Asp 270 acts
as proton donor, or, in the reverse direction, as catalytic
B. Enolic Intermediates in Enzymatic Reactions
C C
R
1
R
2
O
H
C C
R
1
R
2
O
C C
R
1
R
2
O

pK
a
1720
(13-21)
base for deprotonation of R-mandelate.
102 104
The
enzyme is structurally a member of the enolase super-
family
105
and requires Mg
2+
for activity. The pH depen-
dence of the reaction velocity k
cat
reveals two pK
a
values
in the ES complex; ~ 6.4 and ~ 10.0. These are the same
for S- and R- isomers. How can the pK
a
values be
assigned? Do they each belong in part to His 297 and
in part to Lys 166? Do other adjacent groups also share?
In the K166R mutant the lower pK
a
is raised to 8.0.
The D270N mutant has lost all but 0.01% of its catalytic
activity.
104
The carboxylate group of the mandelate interacts
with side chains of E317, K264, and a Mg
2+
ion (Fig.
13-5). These may serve both to protonate the carboxy-
late and also to help stabilize an aci anion formed upon
dissociation of the -hydrogen (Eq. 13-22). Both pK
a
values for mandelic acid and its enolic form and the
equilibrium constants for enolization in water are
known
107
and are given beside the arrows in Eq. 13-22.
It is difficult to imagine how a base with a pK
a
near
neutrality could remove an -proton with a pK
a
of
22.0, which is 14 pK units away from the pK
a
of the
catalytic base. This corresponds to a thermodynamic
barrier (Eq. 9-97) of 14 x 5.7 = 80 kJ/ mol, making the
reaction impossibly slow. In addition to this thermo-
dynamic barrier the rates of dissociation of carbon acids
are known to be slow, presumably because of the lack
of hydrogen bond formation between the C H proton
and the catalytic base (Eq. 9-97). This intrinsic barrier
(Chapter 9) for simple ketones has been estimated
as 45 kJ/ mol for a total barrier of ~125 kJ. However,
the observed G

as estimated from Eq. 9-81 is only


~57 kJ/ mol. The enzyme must catalyze the reaction
by lowering the very high thermodynamic barrier and
perhaps also by lowering the intrinsic barrier.
72,108
Protonation of the carboxylate greatly decreases
the microscopic pK
a
for loss of the -hydrogen as a
proton to form the enolic aci acid (Eq. 13-22). Double
protonation, although depending upon a pK
a
of 8,
HN N
H297
H O O
D270

E221
E247
O
D195 O

O
O
O

K264
K166
N
H
H
H
N
H
H
C
C
O
O
H
H
O

H
O O
E317
:
Mg
2+
+
Figure 13-5 An S-mandelate ion in the
active site of mandelate racemase. Only
some of the polar groups surrounding
the active site are shown. The enzyme
has two catalytic acidbase groups.
Lysine 166 is thought to deprotonate
S-mandelate to form the aci anion, while
His 297 deprotonates R-mandelate to
form the same anion.
106
692 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
ture almost identical to that of mandelate racemase but
has incorporated an additional feature that allows
formation, by addition, of the intermediate aci-acid.
101,105
2. Isomerases
The isomerases that catalyze the simplest reactions
are tautomerases that promote the oxo-enol (keto-enol)
transformation. The widely distributed oxaloacetate
tautomerase (Eq. 13-24) is especially active in animal
tissues.
97,120
Oxaloacetate exists to a substantial extent
in the enolic form: at 38, ~ 6% enol, 13% oxo, and ~ 81%
covalent hydrate.
120,121
A mammalian phenylpyruvate
tautomerase has also been investigated.
122
The oxidation of one functional group of a mole-
cule by an adjacent group in the same molecule is a
feature of many metabolic sequences. In most cases an
enolic intermediate, formed either from a ketone as in
Eq. 13-25 or by a dehydration reaction (see Eq. 13-32),
is postulated.
Aldoseketose interconversions. A metabolically
important group of enzymes catalyze the interconver-
sion of aldose sugars with the corresponding 2-ketoses
(Table 10-1, reaction 4C). Several sugar phosphates
undergo rapid isomerization. Glucose 6-phosphate
isomerase (Eq. 13-25) appears to function in all cells
with a high efficiency.
123 125a
The 132 kDa dimeric pro-
tein from muscle converts glucose 6-phosphate to
HO
C
HO
C
H
HO
HO
C
O
C
H
HO
O
C
O
C
H
HO
HO
C
HO
C
OH
+
~8
HO
C
O
C
OH

log k
e
= 15.4
~3.4
Mandelic acid
22.0
log k
e
= 18.6
aci Acid (enol) form
B
6.6

~7.4
would reduce the pK
a
of the -hydrogen to ~ 7.4, making
it very easy to enolize
109
and solving the thermodynamic
problem. However, what forces could keep the sub-
strate molecule in this unlikely state of protonation?
Gerlt and Gassman proposed formation of a strong,
short hydrogen bond to a carboxylate oxygen.
110
Formation of such a bond would lead to an increased
positive charge on the alpha carbon, lowering the pK
a
and therefore the thermodynamic barrier. It could also
lower the intrinsic barrier,
108
for example, by permit-
ting, to some extent, the formation of a CH- - - N
hydrogen bond in the transition state.
111
Polarization
by the Mg
2+
ion may also be involved. The very strong
electrostatic forces within the active site may be suffi-
cient to explain the formation of enolate anions or enols
as intermediates in many different enzymes.
108,112
A number of other racemases and epimerases may
function by similar mechanisms. While some amino
acid racemases depend upon pyridoxal phosphate
(Chapter 14), several others function without this
coenzyme. These include racemases for aspartate,
113
glutamate,
114115a
proline, phenylalanine,
116
and diamino-
pimelate epimerase.
117
Some spiders are able to inter-
convert D and L forms of amino acid residues in intact
polypeptide chains.
118,119
Another enzyme of the mandelate pathway of
degradation of aromatic rings (Fig. 25-8) is the cis,cis-
muconate lactonizing enzyme which catalyzes the
reaction of Eq. 13-23. It has a three-dimensional struc-
(13-22)
(13-23)
C
O H H
C
C
H
CH
2
C
O
O
OH
O
OH
O
O

OOC CH
2
C COO
O
OOC C C COO
OH

Tautomerase

H
(13-24)
693
fructose 6-phosphate with a turnover number of ~ 10
3
s
1
.
Hereditary defects in this enzyme cause a variety of
problems that range from mild to very severe.
126
Mannose 6-phosphate isomerase also forms fructose
6-phosphate, while ribose 5-phosphate isomerase
127
interconverts the 5-phosphates of D-ribose and D-xylulose
in the pentose phosphate pathways (Chapter 17). Other
enzymes, most often metalloenzymes, catalyze the
isomerization of free sugars.
These enzymes vary widely in secondary and
tertiary structure.
127a
Mannose-6-phosphate isomerase
is a 45 kDa Zn
2+
-containing monomer. The larger 65 kDa
L-fucose isomerase, which also acts on D-arabinose, is
a hexameric Mn
2+
-dependent enzyme.
127a
L-Arabinose
isomerase of E. coli, which interconverts arabinose
and L-ribulose, is a hexamer of 60-kDa subunits
128
while the D-xylose isomerase of Streptomyces is a
tetramer of 43-kDa subunits.
129
The nonenzymatic
counterpart of the isomerization catalyzed by the enzyme
is the base-catalyzed Lobry deBruynAlberda van
Ekenstein transformation (Eq. 13-25).
130
In 1895, Emil Ficher proposed an enediol interme-
diate for this isomerization. As would be expected, the
enzyme-catalyzed isomerization of glucose-6-phosphate
in
2
H
2
O is accompanied by incorporation of deuterium
into the product fructose 6-phosphate at C-1. In the
reverse reaction
2
H-containing fructose 6-phosphate
was found to react at only 45% of the rate of the
1
H-
containing compound. Thus, the primary deuterium
isotope effect expected for a rate-limiting cleavage of
the C H bond was observed (see Chapter 12, Section
B,3).
When fructose 6-phosphate containing both
2
H
and
14
C in the 1 position was isomerized in the presence
of a large amount of nonlabeled fructose 6-phosphate,
the product glucose 6-phosphate contained not only
14
C but also
2
H, and the distribution indicated that the
2
H had been transferred from the C-1 position into the
C-2 position.
123
It was concluded that in over half the
B. Enolic Intermediates in Enzymatic Reactions
turnovers of the enzyme,
2
H removed from C-1 is put
back on the same molecule at C-2. This intramolecular
transfer of a proton suggests a syn transfer, the proton
being removed and put back on the same side of the
molecule. The carrier of the proton may be a histidine
side chain.
131
This result, together with the known
configuration of glucose at C-2, indicates that the
intermediate is the cis-enediol and that addition of a
proton at either C-1 or C-2 of the enediol is to the re
face. However, mannose-6-phosphate isomerase
catalyzes addition to the si face.
Catalysis of ring opening by isomerases. Glu-
cose-6-phosphate isomerase catalyzes a second reaction,
namely, the opening of the ring of the -anomer of
glucose 6-phosphate (one-half of the mutarotation
reaction; Eq. 10-88). Noltmann suggested a concerted
acidbase catalysis by two side chain groups as is
indicated in Eq. 9-90. An NMR study showed that the
isomerization of the anomer occurs at least ten times
faster than that of the . Thus, -glucose 6-phosphate
is first converted to -glucose 6-phosphate before it
can be isomerized to fructose 6-phosphate.
132
Triose phosphate isomerase. This dimeric
53-kDa enzyme interconverts the 3-phosphate esters of
glyceraldehyde and dihydroxyacetone and is the fastest
enzyme participating in glycolysis. Its molecular activity
at 25 is ~ 2800 s
1
in the direction shown in Eq. 13-26
and ~ 250 s
1
in the reverse direction (the predominant
direction in metabolism)
133
and is thought to operate
at the diffusion-controlled limit (Chapter 9).
133
Each
of the identical subunits consists of a striking ( /)
8
barrel (Fig. 13-6) with an active site at the carboxyl
ends of the strands.
134 137
Structures of the enzyme
containing bound inhibitors such as phosphoglycolo-
hydroxamate have also been determined (Fig. 13-6).
138
Triose phosphate isomerase is also one of the most
investigated of all enzymes. Not only are its catalytic
properties unusual but also there are known defects in
the human enzyme
137
and it is also a potential target
for antitrypanosomal drugs.
139
Although its high catalytic activity might favor
intramolecular transfer of the proton removed by
triose phosphate isomerase, little such transfer has
H C
C
OH
H O
C
C
OH H
OH
1
2
Glucose 6-phosphate cis-Enediol
2
H
C H OH
C O
Fructose 6-phosphate
2
H
(13-25)
Glucose
6-phosphate
isomerase
H
O
N
C
H
2
C
O
O
3
P
O

Phosphoglycolohydroxamate (PGH),
a tight-binding inhibitor (K
d
= 15M)
that resembles enediolate 1 of Eq. 13-26
2

694 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
been observed.
140
This suggested that a relatively weak
base such as a carboxylate group might serve as the
proton acceptor at the C-3 position of dihydroxyacetone
phosphate. Covalent labeling experiments and later
X-ray studies have implicated Glu 165, whose carbox-
ylate group is thought to remove the pro-R hydrogen
atom from the hydroxymethyl group of dihydroxyace-
tone phosphate as indicated in Eq. 13-26. When this
residue is replaced by Asp, most activity is lost.
141
Kinetic studies suggest that this carboxyl group has a
pK
a
of ~ 3.9 in the free enzyme. However, k
cat
is inde-
pendent of pH up to pH 10.
140
The pK
a
of the phosphate
also affects the binding (K
m
), with only the phosphate
dianion being a substrate. Study of the infrared spec-
trum of dihydroxyacetone bound to the enzyme re-
vealed a shift of the carbonyl bands at 1733 cm
1
by
about 20 cm
1
to ~ 1713 cm
1
. This might indicate a
polarization and stretching of the carbonyl group by
a positively charged histidine or lysine side chain of
the protein.
142
The His 95 side chain is appropriately
placed (Fig. 13-6) to function in this way
143
and its
replacement by glutamine decreases catalytic activity
by a factor of 400.
144
The NH
3
+
group of lysine 12,
another essential residue, is apparently needed for
substrate binding.
136,145
After product is released, an
isomerization within the enzyme is usually required
to prepare it for acceptance of a new substrate. This
may involve movement of protons between side chain
groups or a conformational change or both. In the
case of triose phosphate isomerase, a very rapid iso-
merization requiring about ~ 1 ms has been detected.
146
To investigate further the function of His 95, Lodi
and Knowles recorded the
13
C and
15
N NMR spectra of
the three histidine rings
148
both in unligated enzyme
and in the phosphoglycolohydroxamate (PGH) complex.
The results were a surprise. The His 95 resonances
did not change at all when the pH was changed from
below 5 to 9.9, and the chemical shift values (shown
above) indicate clearly that the ring is unprotonated
in both free enzyme and in the complex. The key
chemical shift values are shown on the diagram to the
OH
C
C
H
2
C
O
O
3
P
O
H
H
2

E165
O
O

O
C
C
H
2
C
O
O
3
P
O
H
2

3
2
1
Enediolate 1
O
C
C
H
2
C
O
O
3
P
O
H
2

Enediolate 2
E-165
O
O
Dihydroxyacetone
phosphate
H

H
O
C
C
H
2
C
O
O
3
P
H
2

Glyceraldehyde
3-phosphate
H
OH
a
b
c
(13-26)
15
N
C
15
N
H
H N
E97
peptide
H
O
N
C
H
2
C
O
2
O
3
P
O H

PGH
H95
213.5
204.2
138.1
137.1
121.3
122.6
Figure 13-6 Stereoscopic view
into the active site of triose phos-
phate isomerase showing side
chains of some charged residues;
PGH, a molecule of bound phos-
phoglycolohydroxamate, an
analog of the substrate enolate.
138
The peptide backbone, as an
alpha-carbon plot, is shown in
light lines.
147
The ( /)
8
-barrel
structure is often called a TIM
barrel because of its discovery
in this enzyme. Courtesy of M.
Karplus.
695 B. Enolic Intermediates in Enzymatic Reactions
left; the arrows indicate the changes upon formation
of the PGH complex. These results suggest that H95
is never protonated and that if it acts as the catalytic
acid it does so by dissociating to an imidazolate ion
(Eq. 13-27).
148
The imidazole would donate a proton to the enolate ion
and then remove a proton from the resulting enediol.
Although the proposed chemistry is unusual, it is argued
that the high pK
a
of the neutral imidazole acting as a
proton donor would be matched with the high pK
a
of
the enol, permitting rapid reaction.
148
Nevertheless,
two other possibilities exist. Theoretical calculations
support the idea that the proton transfer between the two
oxygens in the enediolate may occur without catalysis
by a proton donor
149
as indicated in step b of Eq. 13-26.
Another possibility is that a transient protonation of
H95 by the adjacent E97 carboxyl group occurs and
allows the histidine to participate in the proton transfer.
Study of kinetic isotope effects has indicated coupled
motion of protons and proton tunneling.
143
Another detail should be mentioned. The active
site of triose phosphate isomerase is formed by a series
of loops connecting the helices and strands of the
barrel. One of those loops, consisting of residues 167
176, folds over the active site after the substrate is
bound to form a hinged lid that helps to hold the
substrate in the correct orientation for reaction.
150 152
When the lid, which can be seen in Fig. 13-6, closes,
the peptide NH of G171 forms a hydrogen bond to a
phosphate oxygen atom of the substrate. This is only
one of many known enzymes with deeply buried active
sites that close in some similar fashion before a rapid
reaction occurs.
Although enzymes tend to be extremely specific
they are not always completely able to avoid side
reactions. Triose phosphate isomerase releases small
amounts of methylglyoxal (Eq. 13-28), presumably as
H
O
C
C
H
2
C
O
O
3
P
H
O
N N H
H
O
C
C
H
2
C
O
O
3
P
H
O
2
Enediol
N N

2
H
Bound enolate
intermediate
Imidazolate

(13-27)
a result of elimination of phosphate from enediolate 2
of Eq. 13-26. Deletion of four residues from the hinged
lid produced an enzyme in which this side reaction
was increased 5.5-fold.
146
Xylose isomerase and the hydride shift
mechanism. This bacterial enzyme, which isomerizes
D-xylose to D-xylulose, has an (/)
8
barrel similar to
that of triose phosphate isomerase. It is of industrial
importance because it also catalyzes isomerization of
D-glucose to the sweeter sugar D-fructose, a reaction
used in preparation of high-fructose syrup. For this
purpose the isomerase is often immobilized on an
insoluble matrix such as diethylaminoethyl cellulose.
153
This enzyme, as well as other isomerases that act on
free sugars, requires a metal ion such as Mg
2+,
Co
2+
,
or Mn
2+
. The three-dimensional structures, solved in
several different laboratories,
129,154 158
show that there
are two metal ions (ordinarily Mg
2+
) about 0.5 nm
apart and held by an array of glutamate and aspartate
side chains. One glutamate carboxylate forms short
ionic bonds to both metal ions and an essential histi-
dine is also coordinated with metal ion 2. The three-
dimensional structure is superficially similar to that of
triose phosphate isomerase but there are major differ-
ences in properties. Xylose isomerase requires metal
ions, acts on unphosphorylated non-ionic substrates,
and does not catalyze detectable exchange of protons
with the solvent. Furthermore, X-ray structures do not
show the presence of any catalytic base that could initiate
formation of an intermediate enolate ion. These facts sug-
gested an alternative isomerization mechanism, one
involving a hydride ion shift and well-known from
studies of the Cannizarro reaction.
159
Because the non-
ionic substrates and inhibitors bind weakly it has been
difficult to obtain a clear picture of events in the active
site. The substrate is the -anomer of D-xylopyranose
155
and the enzyme catalyzes the opening of the sugar
ring (step a, Eq. 13-29).
157
The details of this process
are not clear but acid-base catalysis as in Eq. 9-87 is
probable.
There is a consensus
156 158
that the open form of
O
C
C
H
2
C
O
O
3
P
H
O
H
O
C
C
H
2
C
H
O
H
O
C
C
H
3
C
H
O
Enediolate 2
P
i

2
Methylglyoxal
(13-28)
696 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
(13-30)
the sugar is bound to one of the two metal ions as shown
in Eq. 13-29 and that a bound hydroxide ion on the
second metal provides the catalytic base to remove a
proton from the OH at position 2. The isomerization
occurs with the shaded hydrogen atom being shifted
as H

to the carbon atom of the carbonyl group. During


the reaction metal ion 2 moves apart from metal ion 1
by ~ 0.09 nm,
129,157,158
another fact that has made analy-
sis of X-ray data difficult. Protonation of the alkoxide
ion at C-2 is followed by ring closure (step c, Eq. 13-29).
3-Oxosteroid isomerases. Cholesterol serves
in the animal body as a precursor of all of the steroid
hormones, including the 3-oxosteroids progesterone
and testosterone (Chapter 22). The 3-hydroxyl group
of cholesterol is first oxidized to an oxo group. This is
followed by an essentially irreversible migration of the
double bond in the 5,6-position into conjugation with
the carbonyl group (Eq. 13-30) catalyzed by a 3-oxo-
5
-
steroid isomerase. The small 125-residue enzyme from
the bacterium Pseudomonas testosteroni has been studied
by a great variety of methods.
160 164
The subunits
associate as dimers or higher oligomers. The enzyme
has a high content of nonpolar amino acid residues
and is soluble in high concentrations of ethanol. This
property is compatible with the location of the eukary-
otic enzyme in the endoplasmic reticulum. The sub-
strate binding site is a hydrophobic cavity.
Oxosteroid isomerase has a remarkably high mole-
cular activity (~ 0.75 x 10
5
s
1
). Substrates containing
2
H in the 4 position react only one-fourth as rapidly as
normal substrates. The large isotope effect suggested
that cleavage of the C H bond to form an enzyme-
stabilized enolate anion is rate limiting (Eq. 13-30,
step a).
160
The proton in the axial position at C-4 is
removed preferentially but without complete stereo-
specificty.
165
The abstracted proton must be carried by
a group on the enzyme and returned to the 6 position
of the substrate, again in an axial orientation (Eq. 13-30,
step b), a suprafacial transfer. Studies of the binding
of spin-labeled substrate analogs, molecular modeling,
and directed mutation indicate that Asp 38 is this proton
carrier.
164,166
Little exchange of the proton with solvent
is observed, presumably because of the extreme rapidity
of the isomerase action. However, competitive inhi-
bitors such as nortestosterone, whose double-bond
arrangement is that of the product in Eq. 13-30, undergo
exchange of one of the hydrogens at C-4 with the medium.
The ultraviolet absorption band of the inhibitor at 248
nm is shifted to 258 nm upon combination with the
enzyme, presumably as a result of formation of the
a
b
c
O
HO
OH
3 5
OH
1
HO
O
HO
OH
CH
2
OH
OH
-d-Xylopyranose
Hydride ion shift
d-Xylulose
O
O O
O O H
H H
H
H H
M
1
O
M
2
2
+
2
+
H
N
K182
+
H
H
H

O
O O
O
H
H
M
2
+
H H
O
H H
M
2
+

(13-29)
Stabilized enolate anion
3
5
6
a
H
+
Rate-limiting step
b
+H
+
*
O
O

Asp 38
CH
3
H *
H
H
H
O
O
O
O
C
Asp 99
Tyr 14
H
O
O
O O
C CH
3
H
H H
*
H
O
O
CH
3
H
H
H

697
enolate anion. Tyrosine 14 has been identified as the
catalytic acid, able to stabilize the enolate anion as
depicted in Eq. 13-30.
161,162
A high-resolution NMR
structure
167
and X-ray crystal structures
167a,b
have re-
vealed an adjacent Asp 99 side chain that may assist.
167c
Enzymatic isomerization of cis-aconitate to trans-
aconitate apparently also involves proton abstraction,
165
with resonance in the anion extending into the carbox-
ylic acid group. Its mechanism may be directly related
to that of the oxosteroid isomerase. However, there
are other 1,3-proton shifts in which neither a carbonyl
nor a carboxyl group is present in the substrate
(Eqs. 13-55, 13-56).
4-Oxalocrotonate tautomerase. This bacterial
enzyme, which functions in the degradation of toluene
(Chapter 25), is actually an isomerase. It catalyzes
rapid interconversion of an unconjugated unsaturated
-oxoacid such as 4-oxalocrotonate with an intermedi-
ate enol (which may leave the enzyme) and the iso-
meric conjugated oxoacid (Eq. 13-31).
168 170
A related
5-carboxymethyl-2-hydroxymuconate isomerase
catalyzes the same reaction when R = CH
2
COO

in
Eq. 13-31. It is an unusually small enzyme consisting
of only 62 amino acid residues. The pH dependence
reveals pK
a
values of 6.2 and 9.0 in the free enzyme
and 7.7 and 8.5 in the ES complex.
171
The pK
a
of 6.2 has been associated with the amino-
terminal proline 1.
172
4-Oxalocrotonate tautomerase is
one of a small group of enzymes that have been syn-
thesized nonenzymatically with both L amino acids and
as a mirror image constructed with D amino acids.
173
B. Enolic Intermediates in Enzymatic Reactions
3. Internal OxidationReduction by
Dehydration of Dihydroxyacids
When a carboxylic acid contains hydroxyl groups
in both the and positions, dehydration leads to
formation of an enol that can tautomerize to 2-oxo-3-
deoxy derivatives of the original acid (Eq. 13-32).
Thus, phosphogluconate dehydratase yields 2-oxo-3-
deoxyphosphogluconate as the product. When the
reaction is carried out in
2
H
2
O the
2
H is incorporated
with a random configuration at C-3, indicating that
the enzyme catalyzes only the dehydration and that
the tautomerization of the enol to the ketone is nonen-
zymatic.
The reaction of Eq. 13-32 initiates a unique pathway
of sugar breakdown, the EntnerDouderoff pathway
(Eq. 17-18), in certain organisms. The 6-phosphoglu-
conate is formed by oxidation of the aldehyde group
of glucose 6-phosphate. This pattern of oxidation of
a sugar to an aldonic acid followed by dehydration
according to Eq. 13-32 occurs frequently in metabolism.
A related reaction is the dehydration of 2-phosphogly-
cerate by enolase (Eq. 13-15). In this case the product
is phosphoenolpyruvate, a stabilized form of the enolic
intermediate of Eq. 13-32 (when R = H).
4. Formation and Metabolism of
Methylglyoxal (Pyruvaldehyde)
The rather toxic methylglyoxal is formed in many
organisms and within human tissues.
174
It arises in
part as a side reaction of triose phosphate isomerase
(Eq. 13-28) and also from oxidation of acetone (Eq. 17-7)
or aminoacetone, a metabolite of threonine (Chapter
24).
175
In addition, yeast and some bacteria, including
E. coli, have a methylglyoxal synthase that converts
dihydroxyacetone to methylglyoxal, apparently using
a mechanism similar to that of triose phosphate iso-
merase. It presumably forms enediolate 2 of Eq. 13-26,
which eliminates inorganic phosphate to yield methyl-
COO
H
OH
H
(R) H
COO

The enol 2-Hydroxymuconate


COO
O
H
(R) H
COO

4-Oxalocrotonate
H
H
COO
H
OH
H
COO

2-Oxo-3-hexenedioate
(R) H
H
(13-31)
O
C
R
O
OH
H
H
HO
O
C
R
O
O
H

H
COO
H
2
C
R
O

H
2
O
a
b
For 6-Phosphogluconate
R =
(CHOH)
2
CH
2
OPO
3
2
(13-32)
698 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
C
O
C H C C COO
Cleavage of CH bond
facilitated by a carbonyl group

O
Cleavage with
release of CO
2
uncertain. Glyoxylase II, which catalyzes step c of
Eq. 13-33, is an esterase.
175,180
C. Beta Cleavage and Condensation
In Chapter 12 and in the preceding sections of this
chapter we examined displacement and addition reac-
tions involving nucleophilic centers on O, N, or S.
Bonds from carbon to these atoms can usually be broken
easily by acidic or basic catalysis. The breaking and
making of C C bonds does not occur as readily and
the carbon skeletons of organic molecules often stick
together tenaciously. Yet living cells must both form
and destroy the many complex, branched carbon com-
pounds found within them.
A major mechanistic problem in cleavage or for-
mation of carboncarbon bonds is the creation of a
nucleophilic center on a carbon atom. The problem is
most often solved by using the activating influence of a
carbonyl group to generate a resonance-stabilized enolate
anion. Just as the presence of a carbonyl group facili-
tates cleavage of an adjacent C H bond, so it can also
assist the cleavage of a C C bond. The best known
reactions of this type are the aldol cleavage and the
decarboxylation of -oxo acids. The latter has been
referred to as decarboxylation and its reverse as
carboxylation. In this book these terms have been
extended to include other reactions by which bonds
between the and carbon atoms of a carbonyl com-
pound are broken or formed, and these will be referred
to as cleavage and condensation.
The condensation reactions consist of displacement
or addition reactions in which an enzyme-bound enolate
anion acts as the nucleophile. We can group these con-
densation reactions into three categories as indicated
by reaction types 5A, 5B, and 5C of Table 10-1.
1. Displacement on a Carbonyl Group
A-oxo acid, with proper catalysis, is susceptible
to hydrolysis by attack of water on the carbonyl group.
An example is the reaction catalyzed by oxaloacetate
acetylhydrolase which has been isolated from Asper-
gillus niger (Eq. 13-34). A related cleavage is catalyzed
by ribulose bisphosphate carboxylase (see Eq. 13-48).
glyoxal as in Eq. 13-28.
176,176a
Methylglyoxal is con-
verted to D-lactate by the two-enzyme glyoxalase
system (Eq. 13-33).
177
The combined action of methyl-
glyoxal synthase and the glyoxalases provides a bypass
to the usual glycolysis pathway (Fig. 10-3). Although
it does not provide energy to the cell, it releases inor-
ganic phosphate from sugar phosphates that may
accumulate under conditions of low phosphate because
the free P
i
concentration is too low to support the gly-
ceraldehyde 3-phosphate dehydrogenase reaction
(step a of Fig. 10-3).
Glyoxalase acts not only on methylglyoxal but also
on other -oxo-aldehydes. It is thought to be an im-
portant enzyme system that protects cells against these
potentially dangerous metabolites. Glyoxalase consists
of a pair of enzymes, glyoxalase I and glyoxalase II,
which catalyze the reactions of Eq. 13-33. Each sub-
unit of the 183-residue human glyoxalase I contains
one tightly bound Zn
2+
ion.
178
However, the E.coli
enzyme
178a
is inactive with Zn
2+
and maximally active
with Ni
2+
. The enzyme requires glutathione as a co-
factor, and in step a of the reaction (Eq. 13-33) the gluta-
thione adds to form a thiohemiacetal
179
which is then
isomerized in step b. During this step retention of the
abstracted proton is so complete that it was earlier
thought to function by an intramolecular hydride ion
shift as in xylose isomerase. More recent evidence
favors an enolate anion intermediate. However, the
three-dimensional structure is not related to that of
other isomerases and the exact mechanism remains
O
C
C
R
O
H
S
C
C
R
O
H
HO
G
S
C
C
R
O
G
H
OH
O
C
C
D
OH
H
d-lactate if
R = CH
3
O
Methylglyoxal if
R = CH
3

b
Glyoxalase I
Glyoxalase II
c
H
2
O
GSH
a
GSH (Glutathione)
(13-33)
699
The thiolases
181
are lyases that cleave -oxoacyl
derivatives of CoA by displacement with a thiol group
of another CoA molecule (Eq. 13-35). This is the chain
cleavage step in the oxidation sequence by which
fatty acid chains are degraded (Fig. 10-4). Biosynthetic
thiolases catalyze the condensation of two molecules
of acetyl-CoA to form acetoacetyl-CoA, (see Eq. 17-5),
a precursor to cholesterol and related compounds and
to poly--hydroxybutyrate (Box 21-D). Because acetyl-
CoA is a thioester, the reaction is usually described as
a Claisen condensation. A related reaction, involving
decarboxylation of malonyl-CoA, is a step in fatty acid
synthesis. Since the thiolases are inhibited by SH
reagents it has been suggested that a thiol group in the
enzyme reacts initially with the -carbonyl group as in
Eq. 13-35 to give an enzyme-bound S-acyl intermediate.
The acyl group is then transferred to CoA in a second
step.
A very similar reaction is catalyzed by 3-hydroxy-
3-methylglutaryl-CoA lyase (HMG-CoA lyase), which
functions in the formation of acetoacetate in the human
body (Eq. 17-5, step c) and also in the catabolism of
leucine (Fig. 24-18)
182,183
and in the synthesis of
3-hydroxy-3-methylglutaryl-CoA, the presursor
of cholesterol (Eq. 17-5, step b)
183a
2. Addition of an Enolate Anion to a Carbonyl
Group or an Imine; Aldolases
The aldol condensation (Eq. 13-36), which is also
illustrated in Box 10-D, is one of the more common
C. Beta Cleavage and Condensation
reactions by which C C bonds are formed
183b
and, in
the reverse reaction, cleaved in metabolism. Aldolases
are classified into two major types. The type II aldolases
are metal-ion dependent, the metal ion stabilizing the
intermediate enolate ion (Eq. 13-36, steps a and b).
Type I aldolases, which include the most studied
mammalian enzymes, have a more complex mechanism
involving intermediate Schiff base forms (Eq. 13-36,
steps a, b, c, d).
184
The best known members of this
group are the fructose bisphosphate aldolases
(often referred to simply as aldolases), which cleave
fructose-1,6-P
2
during glycolysis (Fig. 10-2, step e).
These enzymes have been found in all plant and
animal tissues examined and are absent only from a few
specialized bacteria. Three closely related isoenzymes
are found in vertebrates.
185,186
The much studied
rabbit muscle aldolase A is a 158-kDa protein tetramer
of identical peptide chains.
186,187
Aldolase B, which is
lacking in hereditary fructose intolerance, predominates
in liver and isoenzyme C in brain.
185
Treatment with sodium borohydride of the enzyme-
substrate complex of aldolase A and dihydroxyacetone
phosphate leads to formation of a covalent linkage
between the protein and substrate. This and other
evidence suggested a Schiff base intermediate (Eq. 13-36).
When
14
C-containing substrate was used, the borohy-
dride reduction (Eq. 3-34) labeled a lysine side chain
in the active site. The radioactive label was followed
through the sequence determination and was found
on Lys 229 in the chain of 363 amino acids.
186,188188b
The enzyme is another ( /)
8
-barrel protein and the
side chain of Lys 229 projects into the interior of the
barrel which opens at the C-terminal ends of the
strands. The conjugate base form of another lysine,
Lys 146, may represent the basic group B in Eq. 13-36,
R
C
CH
2
C
S
O O
CoA
CoA S
H
H
3
C
C
S CoA
O
R
C
S
O
CoA
Thiolase
Acetyl-CoA
E S H or
C S E R
O
S E
(13-35)
a
b
EnzymeNH
3
+
H
2
O
a
(Type I aldolase)
b
c
(Type II aldolase)
C
O
C H C
HN
C H
Enzyme
C
HN
C
Enzyme
C O
HB
C
O
C
M
2
C O
C
O
C C
Schiff base
OH
B
C
HN
C C
+
+
Enamine
OH
+
Enzyme
Stabilized enolate
+
Product Schiff base

H
2
O
d
EnzymeNH
3
+
HB
+
:B
(13-36)
OOC
C
CH
2
C
O
O O
H

CH
3
COOH

Oxaloacetate (monoanion)
HO
OOC COOH
Oxalate (monoanion)
Acetic acid

(13-34)
700 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
COO
C
CH
2
O
C
R
H OH
COO
C
CH
3
O +
C
R
H O

Pyruvate
(13-37)
step b. Another possibility is that the adjacent phosphate
group of the substrate acts as the acidbase catalyst
for this step.
189
Aldolase A has been altered by muta-
tions into a monomeric form that retains high catalytic
activity,
190
something that has not often been accom-
plished for oligomeric enzymes.
The type II aldolases are not inactivated by sodium
borohydride in the presence of substrate. A probable
function of the essential metal ion is to polarize the
carbonyl group as indicated in Eq. 13-36. In both yeast
and E. coli aldolase the Zn
2+
is held by 3 imidazole
groups.
191,191a
An arginine side chain is a conserved
residue involved in substrate binding in several class
II aldolases.
192
Some blue-green algae contain both
types of aldolase, as do the flagellates Euglena and
Chlamydomonas.
The catabolism of L-fucose by E. coli requires
cleavage of L-fuculose-1-phosphate to form dihydroxy-
acetone phosphate and D-lactaldehyde by a class II
aldolase.
193
The mechanism of chain cleavage proposed on the
basis of the structure and modeling
193
is illustrated in
Fig. 13-7.
Special aldolases of both classes cleave and form
C C bonds throughout metabolism. Several of them
act on 2-oxo-3-deoxy substrates forming pyruvate as
one product (Eq. 13-37). The aldehyde product varies.
In the EntnerDoudoroff pathway of carbohydrate
metabolism (Chapter 21) 3-deoxy-2-oxo-6-phospho-
gluconate (KDPG) is cleaved to pyruvate and 3-phos-
phoglyceraldehyde.
194
The same products arise from
the corresponding phosphogalactonate derivative.
195
The subunits of the trimeric KDPG aldolase have an
( /)
8
-barrel structure similar to that of eukaryotic
fructose 1,6-bisphosphate aldolase.
194
The 8-carbon
sugar acid KDO of bacterial cell walls (Fig. 4-26) is
cleaved by another aldolase. The catabolism of hydroxy-
proline leads to 4-hydroxy-2-oxoglutarate, which is
cleaved to pyruvate and glyoxylate.
196
In the catabo-
lism of deoxynucleotides, another aldolase converts
2-deoxyribose 5-phosphate to acetaldehyde and gly-
ceraldehyde 3-phosphate.
197
Closely related to aldolases is transaldolase, an
important enzyme in the pentose phosphate pathways
of sugar metabolism and in photosynthesis. The mech-
anism of the transaldolase reaction (Eq. 17-15) is similar
to that used by fructose-1,6-bisphosphate aldolase
with a lysine side chain forming a Schiff base and
catalytic aspartate and glutamate side chains.
198
Polycarboxylic acid synthases. Several enzymes,
including citrate synthase, the key enzyme which
catalyzes the first step of the citric acid cycle, promote
condensations of acetyl-CoA with ketones (Eq. 13-38).
An -oxo acid is most often the second substrate, and
a thioester intermediate (Eq. 13-38) undergoes hydrol-
ysis to release coenzyme A.
199
Because the substrate
acetyl-CoA is a thioester, the reaction is often described
as a Claisen condensation. The same enzyme that
catalyzes the condensation of acetyl-CoA with a ketone
also catalyzes the second step, the hydrolysis of the
CoA thioester. These polycarboxylic acid synthases
are important in biosynthesis. They carry out the
initial steps in a general chain elongation process (Fig.
17-18). While one function of the thioester group in
acetyl-CoA is to activate the methyl hydrogens toward
the aldol condensation, the subsequent hydrolysis of
the thioester linkage provides for overall irreversibility
and drives the synthetic reaction.
H
2
C O PO
3
2
C O
C OH H
C O H H
C HO H
CH
3
l-Fuculose-1-phosphate

O
O
O
O
P
2
H
2
C
O
H
C
OH
C
C
H
H
H
3
C
HO
O
H
H O
O

Y113
O
E73
Zn
2+

Figure 13-7 Interaction of the bound zinc ion of L-fuculose-


1-phosphate aldolase and catalytic side chains with the sub-
strate in the active site of the enzyme as revealed by X-ray
crystallography and modeling. See Dreyer and Schulz.
193
701 C. Beta Cleavage and Condensation
a
b
c
C C
S
O
H
H
H CoA
B
C C
S
O
H CoA
H

O C
R
C
O
O

H
2
C
C
R
C S
O
CoA

HO
OOC

H
2
C
C
R

HO
OOC
COO
BH
+
Citrate if R = CH
2
COO

l-Malate if R = H
H
2
O
CoASH
The stereochemistry of the reaction is
also illustrated in Eq. 13-38. These enzymes
may be classified by designating the face of
the carbonyl group to which the enolate
anion adds. The si face is up in Eq. 13-38.
The common citrate synthase of animal
tissues
200
and that of E. coli
201
condense with
the si face and are designated (si)-citrate
synthases. A few anaerobic bacteria use
citrate (re)-synthase having the opposite
stereochemistry.
202
Many citrate synthases
are ~100-kDa dimers
203,204
but some are
hexamers
203,205
and are allosterically inhibit-
ed by NADH. The second substrates and
products of several related reactions of
acetyl-CoA are summarized in Table 13-1.
The si-citrate synthase of pigs is a dimer
of 437-residue chains, each of which is orga-
nized into a large rigid domain and a smaller
(13-38)
TABLE 13-1
Products Arising from Reactions of Acetyl-CoA with a Second Substrate with Catalysis by a
Polycarboxylate Synthase

Glyoxylate
O C
H
COO

Oxaloacetate
O C
CH
2
COO
COO

2-Oxoglutarate
O C
CH
2
CH
2
COO
COO
Ketone substrate Product Further metabolites
Carbohydrates, etc.,
via glyoxylate pathway
2-Carbon unit from
acetyl-CoA occupies
pro-S position
Lysine via
-aminoadipic acid
Leucine
Free acetoacetate
Isoprenoid
compounds

2-Oxoisovalerate
O C
CH
COO
CH
3
CH
3
Acetoacetyl-CoA
O C
CH
2
CH
3
C
O
S CoA
C
CH
2
H
OOC
HO

COO

l-Malate
C
CH
2
CH
2
OOC
HO

COO

Citrate
COO

C
CH
2
CH
2
CH
2
OOC
HO

COO

Homocitrate
COO

C
CH
2
CH
2
HO
H
3
C
COO
C
S
O
CoA

S-3-Hydroxy-3-methylglutaryl-CoA
(HMG-CoA)
C
CH
2
CH
OOC
HO
COO

CH
3
CH
3
-Isopropylmalate
Si-Citrate synthase
702 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
Figure 13-8 Three-dimen-
sional structure of citrate
synthase. (A) Stereoscopic
alpha carbon trace of the
dimeric pig enzyme in its open
conformation.
209
A molecule
of citrate is shown in the
lower subunit. The view
is down the two-fold axis.
Courtesy of Robert Huber.
(B) Space-filling representa-
tion of the open (left) and
closed (right) forms of the
same enzyme.
200
Courtesy
of Stephen J. Remington.
flexible domain. The active site lies between the
domains.
206
When oxaloacetate binds into the cleft
the smaller domain undergoes a complex motion
that closes the enzyme tightly around the substrate
(Fig. 13-8).
200,207,208
The bound oxaloacetate is shown
in Fig. 13-9. It is surrounded by a large number of
polar side chains, including several from histidine and
arginine residues. Of these, Arg 401 and Arg 421 of
the second subunit bind the substrates two carboxyl-
ate groups. In this tight complex the imidazole of His
320 is in the correct position to protonate the carbonyl
oxygen of the oxaloacetate. The domain movement
has also brought the groups that bind the acetyl-CoA
into their proper position creating, by induced fit,
the acetyl-CoA binding site. This conformational
change also accounts for the observation of an ordered
kinetic mechanism with oxaloacetate binding before
acetyl-CoA. The imidazole of His 274 is correctly
oriented to abstract a proton from the methyl group of
acetyl-CoA to generate the intermediate enolate anion.
When oxaloacetate alone is bound into the active site
the carbonyl stretching frequency, observed by infrared
spectroscopy, is shifted from 1718 cm
1
for free oxalo-
acetate to 1697 cm
1
. This decrease of 21 cm
1
suggests
a strong polarization of the C = O bond by its interaction
with His 320 in the ground state.
211
This interaction is
seen also in the
13
C NMR spectrum, the carbonyl
resonance being shifted downfield by 6.8 ppm upon
A
B
703 C. Beta Cleavage and Condensation
binding to the protein.
212
The enolate anion mecha-
nism is also supported by kinetic isotope effects.
213
The methyl protons of dithia-acetyl-CoA are much
more reactive than those of acetyl-CoA (pK
a
~ 12.5 vs
~ 20). When citrate acts on this acetyl-CoA analog
together with oxaloacetate, the expected enolate anion
forms rapidly as is indicated by the appearance of a
306 nm absorption band. However, it condenses very
slowly with the oxaloacetate.
214
The binding equilib-
ria, kinetics, and X-ray structures of complexes for a
variety of other analogs of acetyl-CoA have also been
studied.
210,215- 216a
It appears that Asp 375 and His 274
work together to generate the enolate anion (Fig. 13-9).
NMR measurements indicate formation of an unusually
short hydrogen bond, but its signifi-
cance is uncertain.
216a,216b
Malate syn-
thase (Table 13-1) operates with a very
similar mechanism but has an entirely
different amino acid sequence and
protein fold.
216c
Other Claisen condensations are
involved in synthesis of fatty acids and
polyketides
217
(Chapter 21) and in for-
mation of 3-hydroxy-3-methylglutaryl-
CoA, the precursor to the polyprenyl
family of compounds (Chapter 22). In
these cases the acetyl group of acetyl-
CoA is transferred by a simple displace-
ment mechanism onto an SH group at
the active site of the synthase to form an
acetyl-enzyme.
218,219
The acetyl-enzyme
is the actual reactant in step b of Eq. 17-5
where this reaction, as well as that of
HMG-CoA lyase, is illustrated.
Citrate cleaving enzymes. In
eukaryotic organisms the synthesis of
citrate takes place within the mitochon-
dria, but under some circumstances
citrate is exported into the cytoplasm.
There it is cleaved by ATP-citrate
lyase. To ensure that the reaction goes
to completion, cleavage is coupled to
the hydrolysis of ATP to ADP and inor-
ganic phosphate (Eq. 13-39). The value
of G given here is extremely dependent
upon the concentration of Mg
2+
as a
result of strong chelation of Mg
2+
by
citrate.
220
The reaction sequence is com-
plex but can be understood in terms of
an initial ATP-dependent synthesis of
citryl-CoA using a mechanism similar to that in Eq.
12-47. There is evidence for both phosphoenzyme
and citryl enzyme intermediates (Eq. 13-40).
221
Native
ATP-citrate lyase is a tetramer of 110-kDa subunits.
It usually contains some phosphoserine and phospho-
threonine residues but they apparently have little
effect on activity.
222
Phosphorylation is catalyzed by
cAMP-dependent and by insulin-dependent protein
kinases.
223,224
A related reaction is the ATP-dependent
cleavage of malate to acetyl-CoA and glyoxylate. It
requires two enzymes, malyl-CoA being an intermedi-
ate.
225
Citrate
ATP
ADP + P
i
Oxaloacetate Acetyl-CoA
G' (pH 7)* = +0.9 kJ mol
-1
CoA
(13-39)
H
3
C C S CoA
S
Dithia-acetyl-CoA
R401
H
C
+
N
O
N
N
H
H
H
H
O
C

R329
N
N
N
H
H
H
H
H
C
+
O
C
C
O
O
C
D375 H274
R421
N
N
H
H
N
H
N
H
H
O
O
C
C
H
H
O
S
CoA
C
H
2
N

Figure 13-9 Active site of pig citrate synthase. (A) Stereoscopic view
with a molecule of citrate in the active site.
200
Courtesy of Stephen J.
Remington. (B) Interpretive view of the enolate anion of acetyl-CoA and
oxaloacetate bound in the active site. Based on work by Kurz et al.
210
A
B
704 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
A substrate-induced citrate lyase found in bacteria
such as E. coli and Klebsiella promotes the anaerobic
dissimilation of citrate splitting it to oxaloacetate and
acetate.
226
The large ~ 585-kDa protein from Klebsiella
225
has the composition ()
6
, where the , , and sub-
units have masses of ~ 55-, 30-, and 10-kDa, respectively.
The subunit carries an unusual covalently bound deriv-
ative of coenzyme A
227
(see also Chapter 14). The 10-kDa
subunit serves as an acyl-carrier protein, the SH
of its prosthetic group being acetylated by a separate
ATP-dependent ligase. The resulting acetyl-enzyme
undergoes an acyl exchange (analogous to a CoA-
transferase reaction; Eq. 12-50) to form a citryl enzyme
before the aldol cleavage takes place (Eq. 13-40).
The first unique enzyme of the important glyoxy-
late pathway (Chapter 17), isocitrate lyase, cleaves
isocitrate to succinate and glyoxylate (Eq. 13-41).
228
The carboxylate group that acts as electron acceptor
would presumably be protonated by the enzyme.
3. Chiral Acetates and Their Use in
Stereochemical Studies
Consider the series of enzyme-catalyzed reactions
shown in Eq. 13-42. Fumarate is hydrated in
3
H-con-
taining water to malate which is oxidized to oxaloace-
tate. Hydrolysis of the latter with oxaloacetate acetyl-
hydrolase (Eq. 13-34) in
2
H
2
O gives oxalate and chiral
(R) acetate. The identical product can be obtained by
condensing oxaloacetate with acetyl-CoA using citrate
(re)-synthetase. The resulting citrate is cleaved in
2
H
2
O
using a citrate lyase having the si specificity.
229,230
Acetate of the opposite chirality can be formed enzy-
matically beginning with [2,3-
3
H]fumarate hydrated
by fumarate hydratase in ordinary water. Chiral acetates
have also been prepared nonenzymatically,
229
and
their configuration has been established unequivocally.
During the action of both oxaloacetate acetyl-
hydrolase and citrate lyase (Eq. 13-42) inversion of
configuration occurs about the carbon atom that carries
the negative charge in the departing enolate anion.
That inversion also occurs during catalysis by citrate
(re)-synthase and other related enzymes has been
demonstrated through the use of chiral acetates.
229,230
The findings with malate synthase
199
are illustrated in
Eq. 13-38. Presumably, a basic group B of the enzyme
removes a proton to form the planar enolate anion.
The second substrate glyoxylate approaches from the
other side of the molecule and condenses as is shown.
Since any one of the three protons in either R or S chiral
acetyl-CoA might have been abstracted by base B,
several possible combinations of isotopes are possible in
the L-malate formed. One of the results of the experi-
ment using chiral (R) acetyl-CoA is illustrated in Eq.
13-43. The reader can easily tabulate the results of
removal of the
2
H or
3
H. However, notice that if the
base B: removes
2
H (D) or
3
H (T) the reaction will be
much slower because of the kinetic isotope effects
which are expected to be
H
k/
D
k 7 and
H
k/
T
k 16. A
second important fact is that the pro-R hydrogen at C-3
in malate is specifically exchanged out into water by
the action of fumarate hydratase. From the distribution
of tritium in the malate and fumarate formed using the
two chiral acetates, the inversion by malate synthase
was established. See Kyte
231
for a detailed discussion.
C COO
O
H
H
C
H
CH
2
HOOC COO
OOC
CH
2
CH
2
COO

threo-d
s
-Isocitrate
C COO
O
H

Succinate
G' (pH 7) = 8.7 kJ mol
-1

Glyoxylate

(13-41)
Acetyl-E citryl-E acetyl-E
citrate
acetate oxaloacetate
(13-40)
C C
COO
H
H
OH
3
H OOC
C C
COO
H
O
3
H OOC
OOC COO
C
HO CH
2
COO
C COO OOC
3
H H

Fumarate
3
HOH


C COO
2
H
3
H
H
R-Acetate

2
H
2
O
Oxaloacetate
acetyl-
hydrolase

R-Acetate
Citrate lyase
2
H
2
O
Oxaloacetate
Citrate
re-synthase
Acetyl-CoA
C COO
2
H
3
H
H

(13-42)
705
4. Addition of an Enolate Ion to Carbon
Dioxide and Decarboxylation
The addition of an enolate anion to CO
2
to form a
-oxoacid represents one of the commonest means of
incorporation of CO
2
into organic compounds. The
reverse reaction of decarboxylation is a major mecha-
nism of biochemical formation of CO
2
. The equilibrium
constants usually favor decarboxylation but the cleav-
age of ATP can be coupled to drive carboxylation when
it is needed, e.g., in photosynthesis.
Decarboxylation of -oxoacids. Beta-oxoacids
such as oxaloacetic acid and acetoacetic acid are unsta-
ble, their decarboxylation being catalyzed by amines,
metal ions, and other substances. Catalysis by amines
depends upon Schiff base formation,
232
while metal
ions form chelates in which the metal assists in electron
withdrawal to form an enolate anion.
233 235
Can we apply any of this information from non-
enzymatic catalysis to decarboxylating enzymes?
Some decarboxylases do form Schiff bases with their
substrates, and some are dependent on metal ions.
235
The acetone-forming fermentation of Clostridium
acetobutylicum requires large amounts of acetoacetate
decarboxylase (Eq. 13-44).
The enzyme is inactivated by borohydride in the presence
of substrate, and acid hydrolysis of the inactivated
enzyme yielded -N-isopropyllysine. Decarboxylation
occurs from a Schiff base by a mechanism analogous
to that of the aldol cleavage shown in Eq. 13-36.
236
Mechanistically related is 4-oxalocrotonate decarboxy-
lase.
236a
Linked oxidation and decarboxylation. Metabolic
pathways often make use of oxidation of a -hydroxy
acid to a -oxoacid followed by decarboxylation in
the active site of the same enzyme. An example is
conversion of L-malate to pyruvate (Eq. 13-45). The
Mg
2+
or Mn
2+
-dependent decarboxylating malic dehy-
drogenase that catalyzes the reaction is usually called
the malic enzyme. It is found in most organisms.
237 240
While a concerted decarboxylation and dehydrogena-
tion may sometimes occur,
241242
the enzymes of this
group appear usually to operate with bound oxoacid
intermediates as in Eq. 13-45.
Other reactions of this type are the oxidation of isoci-
trate to 2-oxoglutarate in the citric acid cycle (Fig. 17-4,
steps d and e),
243
oxidation of 6-phosphogluconate to
ribulose 5-phosphate (Eq. 17-12),
244
and corresponding
reactions of isopropylmalate dehydrogenase
245,246
and
tartrate dehydrogenase.
247,248
Crystallographic studies
of isocitrate dehydrogenase using both photolabile
caged isocitrate
247
and slow mutant forms
243
with
polychromatic Laue crystallography (Chapter 3) have
demonstrated the rapid formation of the anticipated
intermediate oxalosuccinate.
Phosphoenolpyruvate, a key metabolic inter-
mediate. A compound of central importance in
metabolism is the phosphate ester of the enol form of
pyruvate, commonly known simply as phosphoenol-
pyruvate (PEP).
249
It is formed in the glycolysis path-
way by dehydration of 2-phosphoglycerate (Eq. 13-15)
or by decarboxylation of oxaloacetate. Serving as a
preformed enol from which a reactive enolate anion
can be released for condensation reactions,
250,251
PEP
C. Beta Cleavage and Condensation
C C
H
H
H
O
S CoA
C
C
H
COO
OOC
H
OH

l-Malate
Bi
Malate synthase
(Eq. 13-38)
Glyoxylate
This proton is
exchanged out
by the activity
of fumarase
(13-43)
O C
O
C
C
C
O
O
O
H H
M
2


+
H
3
C
C
O
CH
2
COO

H
3
C
C
O
CH
3
Acetoacetate
CO
2
H
+
Acetone
(13-44)
CH
2
C
COO
OOC
HO H
CH
2
C
COO
OOC
O

H
3
C
C
COO
NADP
H

O
NADPH + H
+
+

+
CO
2
Bound oxaloacetate Pyruvate
(13-45)
706 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
is utilized in metabolism in many ways.
In animals and in many bacteria, PEP is formed
by decarboxylation of oxaloacetate. In this reaction,
which is catalyzed by PEP carboxykinase (PEPCK), a
molecule of GTP, ATP, or inosine triphosphate captures
and phosphorylates the enolate anion generated by the
decarboxylation (Eq. 13-46).
252
The stereochemistry is
such that CO
2
departs from the si face of the forming
enol.
253
The phospho group is transferred from GTP
with inversion at the phosphorus atom.
254
The en-
zyme requires a divalent metal ion, preferably Mn
2+
.
In fact, kinetic studies of the GTP-dependent avian
mitochondrial enzyme indicate two metal-binding
sites, one on the polyphosphate group of the bound
GTP and one on carboxylate side chains of the pro-
tein.
252,255
The three-dimensional structure of the ATP-
dependent E. coli enzyme reveals a nucleotide binding
site similar to the ATP site of adenylate kinase (Fig.
12-30).
256
A definite binding site for CO
2
is also present
in the enzyme.
257
PEPCK is also activated by low concentrations of
Fe
2+
and this activation depends upon a protein that
has been identified as glutathione peroxidase (Eq. 15-58).
By destroying H
2
O
2
the latter may allow the Fe
2+
to
prevent oxidation of an SH group on PEP carboxy-
kinase.
258
Synthesis of PEPCK is stimulated by gluca-
gon, evidently through a direct action of cAMP on
transcription of the structural gene.
259
Transcription
is also stimulated by glucocorticoids and thyroid
hormone and is inhibited by insulin.
In some organisms, such as the parasitic Ascaris
suum, PEPCK functions principally as a means of
synthesis of oxaloacetate by reaction of PEP with CO
2
and GDP. These organisms lack pyruvate kinase,
which allows for buildup of the high PEP concentra-
tion needed to drive this reaction.
260
In a similar way
PEP can be converted to oxaloacetate by PEP carboxy-
transphosphorylase, an enzyme found only in pro-
pionic acid bacteria and in Entamoeba. The reaction
(Eq. 13-47) is accompanied by synthesis of inorganic
pyrophosphate which may be cleaved to pull the
reaction in the indicated direction.
PEP + CO
2
+ P
i
PP
i
+ oxaloacetate
Oxaloacetate is also decarboxylated without phos-
phorylation of the enolate anion formed but with release
of free pyruvate. Both pyruvate kinase and PEPCK
can act as oxaloacetate decarboxylases.
261
In the important reactions discussed in the follow-
ing sections enolate ions are intermediates in carbon-
carbon bond formation. Other examples are given in
Eqs. 20-7 and 20-8, and Fig. 25-1, in which C C bonds
are formed by action of PEP as a carbon nucleophile,
e.g., in aldol-like condensations.
242a
Yet another unusual
type of reaction, involving formation of an enol ether
linkage is illustrated in Box 13-B and in Eq. 25-3.
Ribulose bisphosphate carboxylase. The major
route of incorporation of CO
2
into organic compounds
is via photosynthesis. When
14
C-labeled CO
2
enters
chloroplasts of green plants, the first organic
14
C-con-
taining compound detected is 3-phosphoglycerate.
Two molecules of this compound are formed through
the action of ribulose-1,5-bisphosphate carboxylase
(abbreviated rubisco), an enzyme present in chloro-
plasts and making up 16% of the protein of spinach
leaves. This enzyme, whose structure is illustrated in
Fig. 13-10, is thought to be the most abundant protein
on earth. Because O
2
competes with CO
2
as a substrate,
the enzyme also catalyzes an oxygenase reaction. It
is therefore often called ribulose-1,5-bisphosphate
carboxylase/ oxygenase.
262
The rubisco from most
plants is a 500 - to 560-kDa L
8
S
8
oligomer as shown in
Fig. 13-10. The large subunit is encoded in the chloro-
plast DNA. However, a family of nuclear genes en-
code the small subunits, which are synthesized as
larger precursors, with N-terminal extensions being
removed to give the mature subunits.
263,264
Two types
of small subunits occupy different positions in the
OOC C
O
CH
2
PO
3
2

Phosphoenolpyruvate (PEP)

H
2
C C
O
COO
OOC
O
P
O
P
O
P
O
O O O O O O
Guanosine

Oxaloacetate
H
2
C C
O
COO

GTP

CO
2
re Face is toward reader
PEP
GDP
PO
3
2

(13-46)
(13-47)
H

Nu
OOC C
O
CH
P

C
O
O
O
707
quasi-symmetric spinach rubisco.
264
In Euglena the
small subunit is synthesized as a polyprotein precursor
containing eight copies of the subunit.
265
Rubisco
from the hydrogen-oxidizing bacterium Alcaligenes
eutrophus has a similar quaternary structure,
266
but
the enzyme from Rhodospirillum rubrum is a simple
dimer.
267
In dinoflagellates the rubisco gene is present
in nuclear DNA rather than in the chloroplasts.
268
The carboxylation reaction catalyzed by rubisco
differs from others that we have considered in that
the carboxylated product is split by the same enzyme
(Eq. 13-48). The mechanism shown in Eq. 13-48 was
suggested by Bassham et al.
271
Ribulose bisphosphate,
for which the enzyme is absolutely specific, is first
converted to its enolic form, a 2,3-enediol. Loss of a
proton from the 3-OH group forms the endiolate anion
needed for the carboxylation in step b. The product
of this step is a -oxoacid which undergoes enzyme-
catalyzed hydrolytic cleavage (Eq. 13-48, step c; see
also Eq. 13-34). Support for this mechanism came
from the observation that 2-carboxyarabinitol bisphos-
phate (Fig. 13-11B) is a potent inhibitor, possibly a
transition state analog.
272
This inhibitor, bound into
the active site of the enzyme from spinach, is seen in
both Figs. 13-10B and 13-11A. An expanded version of
Eq. 13-48 is given in Fig. 13-12, which is based upon
modeling together with a variety of X-ray structures
including that shown in Fig. 13-11.
An essential Mg
2+
ion is held by carboxylates of
D203 and E204 and by modified K201. It also coordi-
nates three molecules of H
2
O in the free enzyme.
272a
Catalytic roles for the H294 and H327 imidazole groups
are still being elucidated.
272b
Lysine 175 protonates
C2 of the aci anion generated by CC bond cleavage
(step e, Fig. 13-12).
272c,276
Like many enzymes, rubisco
exists in two major conformational states: open and
closed.
272d
C. Beta Cleavage and Condensation
Chemical studies also support the indicated mech-
anism. For example, the -oxoacid intermediate
formed in step b of Eq. 13-48 or Fig. 13-12 has been
identified as a product released from the enzyme by
acid denaturation during steady-state turnover.
273,274
Isotopic exchange with
3
H in the solvent
275
and mea-
surement of
13
C isotope effects
277
have provided addi-
tional verification of the mechanism. The catalytic
activity of the enzyme is determined by ionizable
groups with pK
a
values of 7.1 and 8.3 in the ES com-
plex.
278
The apparent value of K
m
for total CO
2
(CO
2
+
HCO
3

) is high, 11 30 mM, but for the true substrate


CO
2
it is only 0.45 mM. In intact chloroplasts the
affinity for substrate is distinctly higher, with the K
m
for total CO
2
dropping to ~ 0.6 mM. The difference
appears to result largely from a regulatory reaction of
CO
2
in which the side chain amino group of lysine 201
of the large subunit forms a carbamate (Eq. 13-49).
Although carbamylation is spontanous, it is enhanced
by an ATP-dependent process catalyzed by rubisco
activase.
279
The carbamylation converts the side
chain of Lys 201 into a negatively charged group that
binds to an essential divalent metal, usually Mg
2+
, in
the active center
282
as is shown in Fig. 13-11, A and B.
The nature of the reaction is uncertain. One possibility
is that the activase is a chaperonin.
280
It appears to
assist the enzyme in removing inhibitory sugars that
arise by side reactions in the active site.
281
Rubisco is
also regulated by the level of a natural inhibitor which
has been identified as 2-carboxyarabinitol 1-phosphate.
This is the same as the inhibitor shown in Fig. 13-11
but with one less phosphate group and consequent
weaker binding.
273a
In most plants photosynthesis is also strongly
inhibited by O
2
. This observation led to the discovery
that O
2
competes directly for CO
2
at the active site of
rubisco in a process called photorespiration. Chloro-
plasts inhibited by oxygen produce glycolate in large
amounts
282a
as a result of the reaction of the intermedi-
ate enediolate ion formed in step b of Eq. 13-48 with O
2
a
b
c
C
C
C
CH
2
OPO
3
2
CH
2
OPO
3
2
HO
H
OH
H
O
1
2
3

C
C
C
CH
2
OPO
3
2
CH
2
OPO
3
2
O
OH
H
HO
O C O

C
C
C
CH
2
OPO
3
2
CH
2
OPO
3
2
O
OH
H
HO
OOC

H
2
O

C
COO
HO
H
CH
2
OPO
3
2

Ribulose 1,5-bisphosphate Enediolate


d-3-Phosphoglycerate
(13-48)
N C
O
O
Lys 201
N C
O
O
Lys 201 Mg
2
H

H
NH
2
Lys 201
Mg
2
Inactive form
CO
2

+
+
Active form
(13-49)
708 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
Figure 13-10 Stereoscopic view of
ribulose bisphosphate carboxylase
(rubisco) from spinach. (A) The sym-
metric L
8
S
8
molecule contains eight 475-
residue large subunits (in two shades of
gray) and eight 123-residue small sub-
units (lighter gray in upper half of
image). (B) One L
2
S
2
substructure
containing two active sites shared
between adjacent large subunits with
the bound inhibitor 2-carboxy-D-arabini-
tol 1,5-bisphosphate.
269
In the upper LS
unit the S subunit (top) is light and the L
subunit is dark. Courtesy of Inger
Andersson. Similar structures have
been determined for enzymes from
tobacco
269a
and from the cyanobacterium
Synechoccus.
270
Figure 13-11 (A) Overview of the active site of spinach rubisco showing bound 2-carboxy-D-arabinitol 1,5-bisphosphate and
Mg
2+
and residues within hydrogen-bonding distance of these ligands. The hydroxyl groups at C2 and C3 of the inhibitor are
in cis conformation.
269
Courtesy of Inger Andersson. (B) Structure of the inhibitor 2-carboxy-D-arabinitol 1,5-bisphosphate. A
part of the carbamylated lysine 201 and the essential metal ion are also shown.
O
O
O
O
P
2
1
HO
O
OH
OH H
H
O
M
O
N H
K201
4
5
O
O
O
O
P
2
2
3
2
O

+
A
B
A B
709
Figure 13-12 Proposed mechanism of action of ribulose bisphosphate carboxylase (rubisco). This is an abbreviated version
of the mechanism as presented by Taylor and Andersson.
276
The binding of ribulose 1,5-bisphosphate occurs after carbamyla-
tion of lysine 201 and binding of a magnesium ion. Formation of an enediolate intermediate in step a is probably catalyzed by
the carbamate group as indicated. Removal of a proton from the 3-OH, perhaps by H294, and addition of a proton to form an
OH group at C2 are also necessary. The CO
2
may bind to the Mg
2+
and be polarized by interaction with other side chains
prior to reaction. Carboxylation occurs by addition of the enediolate to CO
2
in step b. The hydration of the resulting 3-oxo
group (step c) is necessary for cleavage of the C C bond in step d. The participation of the newly formed carboxylate as an aci
anion coordinated to Mg
2+
is presumably involved. Protonation, with stereochemical inversion, is thought to involve K175, as
shown. The two product molecules dissociate in step e.
E204
C
O
O

OH
2
Mg
2
O
O
C

O
O
H
H
C
C
O
H294
HN
N
C
O
K201
HN
1
2
3
4
5
+
D203
OH
Carbamylated
lysine 201, one
resonance form
E204
C
O
O

O
Mg
2
O
O
C

O
O
C
C
O
H294
HN
N
C
HO
OPO
3
2
K201
HN
+
D203
OH
H

O
C
H
3
N
K334
b
c
H294
abstracts
proton
+
a
CO
2
binds

E204
C
O
O

O
Mg
2
O
O
C

O
O
C
C
O
C
HO
K201
HN
+
D203
O
2
O

+
C
OPO
3
2
OPO
3
2
H
NH
H
K175
+
d
OPO
3
2
OPO
3
2
+
+
+
OPO
3
2
Enediolate and bound CO
2
E204
C
O
O

O
Mg
2
O
O
C

O
O
C
C
O
C
HO
K201
HN
OPO
3
2
+
D203
O
C
2
1
3
O H
H327
HN
N
H
+

E204
C
O
O

O
Mg
2
O
O
C

O
O
C
C
O
C
HO
K201
HN
OPO
3
2
+
D203
OH
O
C
Hydration of
C-3 carbonyl
2
1
3
O
H
H
H327
HN
N

OH
OH
e
Two molecules of
3-Phosphoglycerate
H
H H
H
C. Beta Cleavage and Condensation
710 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
intermediate of Eq. 13-50. The resulting dicarbonyl
bisphosphate can rearrange to give a carboxytetritol
bisphosphate (Eq. 13-51).
284
Carbon dioxide or bicarbonate ion? An impor-
tant question in the consideration of carboxylation and
decarboxylation reactions is whether the reactant or the
product is CO
2
or HCO
3

. An approach to answering
the question was suggested by Krebs and Roughton,
286
who pointed out that the attainment of equilibrium
between free CO
2
and HCO
3

(Eq. 13-52) may require


several seconds. If an enzyme produces CO
2
as a
product and progress of the reaction is followed mano-
metrically, the pressure will rise higher than the equi-
librium value as the CO
2
is evolved. Later when
substrate is exhausted and the CO
2
equilibrates with
bicarbonate, the pressure will fall again. The addition
of carbonic anhydrase, which catalyzes Eq. 13-52, step
a, abolishes the overshoot. If bicarbonate is the
primary product of a decarboxylation, there is a lag
in the appearance of free CO
2
.
A second approach is to use [
18
O]bicarbonate and
to follow the incorporation of
18
O into a carboxylated
substrate. If CO
2
is the primary substrate only two
labeled oxygen atoms enter the compound, whereas
if HCO
3

is the reactant three are incorporated.


287
A
third technique is measurement of the rate of incorpo-
ration of CO
2
or bicarbonate in the carboxylated prod-
uct. Over a short interval of time, e.g., 1 min, different
kinetics will be observed for the incorporation of CO
2
and of bicarbonate.
288
Using these methods, it was
established that the product formed in Eq. 13-46 and
the reactant in Eq. 13-47 is CO
2
. However, the carboxyla-
tion enzymes considered in the next section use bicar-
bonate as the substrate.
C
C
O
O
C
CH
2
OPO
3
2
OH
H
H CH
2
OPO
3
2
C
C
CH
2
OPO
3
2
OH

+
Deoxypentodiulose-bisphosphate

H
HO
2-Carboxytetritol-1,4-bisphosphate
CH
2
OPO
3
2
HO
OOC

(13-51)
a b
CO
2
+
H
2
O H
2
CO
3
H
+
HCO
3
+

(13-52)
(Eq. 13-50, step a). The peroxide formed in this way
breaks up under the hydrolytic action of the enzyme
to form phosphoglycolate and 3-phosphoglycerate
(Eq. 13-50, step b).
Molecular oxygen usually reacts rapidly with only
those organic substrates, such as dihydroflavins, that
are able to form stable free radicals. However, the
endiolate anion of Eq. 13-50 may be able to donate a
single electron to O
2
to form a superoxideorganic
radical pair prior to formation of the peroxide (see also
Eq. 15-30). Similar oxygenase side reactions have been
observed for a variety of other enzymes that utilize
carbanion mechanisms.
283
The reaction of rubisco
with O
2
is of both theoretical and practical interest, the
latter because of its significance in lowering the yield
in photosynthesis (Chapter 23).
The simpler dimeric rubisco from Rhodospirillum is
very inefficient in carboxylation and catalyzes much
more oxygenation than do rubiscos of higher plants.
283a
Mutant enzymes that have impaired carboxylase and
enhanced oxygenase are also known.
284,284a
The small subunits of rubisco may help suppress
undesirable side reactions.
285
For example, the follow-
ing deoxypentodiulose phosphate can be formed by
elimination from the second intermediate of Eq. 13-48.
Some side products of the normal oxgenase reaction
arise by elimination of a peroxide ion from the first
O
O
C
H
C
HO CH
2
OPO
3
2
O C
CH
2
OPO
3
2
H
OH
O
O
C
H
C
HO CH
2
OPO
3
2
O C
CH
2
OPO
3
2
H
OH
H
C
C
CH
2
OPO
3
2
H
OH
O
O
+

C
CH
2
OPO
3
2
O O
OH

3-Phosphoglycerate Phosphoglycolate

a
b
H
2
O
(13-50)
C
C
O
O
C
CH
2
OPO
3
2
OH
CH
3

H
711
5. Incorporation of Bicarbonate into Carboxyl
Groups
An important enzyme with a biosynthetic function
in many bacteria and in all higher plants is PEP car-
boxylase,
289
which catalyzes the reaction of Eq. 13-53.
This enzyme, in effect, accomplishes the reverse of Eq.
13-46 by converting the three-carbon PEP, by reaction
with bicarbonate, into the four-carbon oxaloacetate.
The latter is needed for priming of the citric acid
cycle and for biosynthesis of such amino acids as
aspartate and glutamate. That the enzyme functions
in this way is indicated by the fact that mutants of
Salmonella defective in the enzyme do not grow unless
oxaloacetate or some other intermediate in the citric
acid cycle is added to the medium. The enzyme from
S. typhimurium is a 400-kDa tetramer with complex
regulatory properties. The corresponding enzyme
from spinach has 12 subunits and 12 bound Mn
2+
ions. The enzyme also has a special function in the
C
4
plants,
290
in which it is a component of a carbon
dioxide concentrating system (Chapter 23).
When [
18
O]bicarbonate is a substrate, two labeled
oxygen atoms enter the oxaloacetate, while the third
appears in P
i
. A concerted, cyclic mechanism could
explain these results. However, study of kinetic iso-
tope effects,
291
use of a substrate with a chiral thio-
phospho group,
292
and additional
18
O exchange
studies
293
have ruled out this possibility. A transient
carboxyl phosphate (Eq. 13-53) is evidently an inter-
mediate.
294,295
The incorporation of the
18
O from bicar-
bonate into phosphate is indicated by the asterisks.
The carboxyl group enters on the si face of PEP. How-
ever, there is another possibility.
295,296
The carboxyl
phosphate, while in the active site adjacent to the
enolate anion, may eliminate phosphate, the enolate
ion adding to the resulting CO
2
to form the final
product. According to this mechanism the group
transfer potential of the phosopho group in PEP is
C. Beta Cleavage and Condensation
utilized to concentrate the CO
2
and to localize it next
to the enolate anion generated by the same process.
PEP carboxylase is lacking from animal tissues
and fungi. In these creatures PEP is converted to
pyruvate, which is then carboxylated to oxaloacetate
with coupled cleavage of ATP by the action of pyruvate
carboxylase (Eq. 14-3), an enzyme that not only utilizes
bicarbonate ion but also contains biotin. However,
there are mechanistic similarities between its action
and that of PEP carboxylase.
PEP mutase and the synthesis of phosphonates.
The lipids of some organisms, such as Tetrahymena,
contain aminoethylphosphonate, a compound with
a C P bond (Chapter 8). There are also many other
naturally occurring phosphono compounds and huge
quantities of synthetic phosphonates, present in deter-
gents, herbicides, and insecticides, are metabolized by
bacteria.
297
Here we will consider only one step in the
biosynthesis of phosphonates, the conversion of PEP
into phosphonopyruvate (Eq. 13-54), a reaction cata-
lyzed by PEP mutase. The phospho group is moved
from the oxygen atom of PEP to the methylene carbon
atom. When attached to carbon the phospho group
is designated phosphono. The reaction occurs with
retention of the configuration around phosphorus
and a phosphoenzyme intermediate seems to be in-
volved
298,299
as shown in Eq. 13-54. The equilibrium in
this reaction strongly favors PEP. One further product
formed from phosphonopyruvate is phosphonoethyl-
amine, a component of phosphonolipids.
300
It is easy
to imagine a synthetic route involving transamination
followed by decarboxylation. Another mutase, similar
to PEP mutase, shifts the carboxyphospho group

OOC PO
2

exactly as in Eq. 13-54 as part of the


biosynthetic pathway of a natural herbicide formed
by Streptomyces hygroscopicus.
301,302
Soil bacteria con-
tain specialized enzymes that catalyze the hydrolytic
cleavage of P C bonds.
302a
C
CH
2
OOC O P
O
OH
O
OOC
H
2
C
COO
O
C
H
2
C
O OOC
C
OH
O
O
P
O
O
OH
O C
O
OH


P
O
O
OH
*
*
Carboxyl phosphate

*
O

*
PEP
Oxaloacetate
a
b
*
*
* *
(13-53)
C
CH
2
OOC O P
O
O
OH

Y Enzyme
C
O
OOC CH
2
P
O
O
Y Enzyme
HO
OOC
C
CH
2
P
O O
O
OH

Y Enzyme
Phosphonopyruvate

PEP
a
b
Reverse reaction ( )
is strongly favored
(13-54)
712 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
fact that the enzyme is strongly inhibited by the cation
of 2-(diethylamino) ethyl pyrophosphate, an analog of
a probable carbocation intermediate. Cysteine and
glutamate side chains are essential.
307
Reaction type 6B of Table 10-1 is allylic rearrange-
ment with simultaneous condensation with another
molecule. The reaction, which is catalyzed by prenyl-
transferases,
307a
occurs during the polymerization
of polyprenyl compounds (Fig. 22-1,Eqs. 22-2, 22-3).
Experimental evidence favors a carbocation mechanism
for all of these reactions.
308,309
See Chapter 22.
Reaction type 6C (Table 10-1) occurs during the
biosynthesis of leucine and valine (Fig. 24-17). The
rearrangement is often compared with the nonenzy-
matic acid-catalyzed pinacolpinacolone rearrangement
in which a similar shift of an alkyl group takes place
(Eq. 13-57). The enzyme-catalyzed rearrangement
presumably gives the structure drawn in brackets in
Table 10-1. The same enzyme always catalyzes reduction
with NADH to the diol, the Mg
2+
-dependent enzyme
being called acetohydroxy acid isomeroreductase.
Rearrangement has never been observed without the
accompanying reduction.
310313
More complex rear-
rangements that occur during biosynthesis of sterols
are described in Chapter 22.
H
3
C C
CH
3
OH
C
CH
3
OH
CH
3
Pinacol
H
3
C C
CH
3
OH
C
CH
3
O
CH
3
H
+
H
H
3
C C
CH
3
O
C
CH
3
CH
3
+
H
H
3
C C
O
C
CH
3
CH
3
CH
3
Pinacolone
H
2
O
H
+
H
+
(13-57)
D. Some Isomerization and Rearrangement
Reactions
A few metabolic reactions do not fit into any of
the categories discussed so far and apparently do not
depend upon a coenzyme. Some of these involve
transfer of alkyl groups or of hydrogen atoms from
one carbon to another. The hydrogen atoms move by
direct transfer without exchange with the medium.
All of the reactions could involve carbocations but
there is often more than one mechanistic possibility.
A simple 1,3-proton shift is shown in Table 10-1 as
reaction type 6A. An example is the isomerization of
oleic acid to trans-
10
-octadecenoic acid (Eq. 13-55)
catalyzed by a soluble enzyme from a pseudomonad.
303
A second example is isomerization of isopentenyl
diphosphate to dimethylallyl diphosphate (Eq.
13-56).
304 307
The stereochemistry has been investigated
using the
3
H-labeled compound shown in Eq. 13-56.
The pro-R proton is lost from C-2 and a proton is added
to the re face at C-4. When the reaction was carried
out in
2
H
2
O a chiral methyl group was produced as
shown.
304
A concerted proton addition and abstraction
is also possible, the observed trans stereochemistry
being expected for such a mechanism. However, the
C C
H
C
H
H
H
H
C C
CH
2
Oleic acid
(cis-
9
-octadecenoic acid)
10
trans-
10
-Octadecenoic acid
H
+
9
H
(13-55)
1
H
C
C
C
CH
2
O P P
CH
3
H H
R
3
H
T
Isopentenyl diphosphate
1
H
C
C
C
CH
2
O P P
CH
3
Dimethylallyl diphosphate
3
H
2
H
H
2
H
+
H
+
(13-56)
713
50. Sacchettini, J. C., Meininger, T., Rodrick, S.,
and Banaszak, L. J. (1986) J. Biol. Chem. 261,
1518315185
51. Bearne, S. L., and Wolfenden, R. (1995) J. Am.
Chem. Soc. 117, 95889589
52. Weaver, T., and Banaszak, L. (1996) Biochemis-
try 35, 1395513965
53. Weaver, T., Lees, M., and Banaszak, L. (1997)
Protein Sci. 6, 834842
53a. Weaver, T., Lees, M., Zaitsev, V., Zaitseva, I.,
Duke, E., Lindley, P., McSweeny, S., Svensson,
A., Keruchenko, J., Keruchenko, I., Gladilin,
K., and Banaszak, L. (1998) J. Mol. Biol. 280,
431442
53b. Beeckmans, S., and Van Driessche, E. (1998)
J. Biol. Chem. 273, 3166131669
54. Mohrig, J. R., Moerke, K. A., Cloutier, D. L.,
Lane, B. D., Person, E. C., and Onasch, T. B.
(1995) Science 269, 527529
55. Rose, I. A. (1972) CRC Critical Review of
Biochemistry 1, 3357
56. Brant, D. A., Barnett, L. B., and Alberty, R. A.
(1963) J. Am. Chem. Soc. 85, 22042209
57. Schmidt, D. E., Jr., Nigh, W. G., Tanzer, C.,
and Richards, J. H. (1969) J. Am. Chem. Soc. 91,
58495854
58. Nigh, W. G., and Richards, J. H. (1969) J. Am.
Chem. Soc. 91, 58475848
59. Hansen, J. N., Dinovo, E. C., and Boyer, P. D.
(1969) J. Biol. Chem. 244, 62706279
60. Blanchard, J. S., and Cleland, W. W. (1980)
Biochemistry 19, 45064513
61. Porter, D. J. T., and Bright, H. J. (1980) J. Biol.
Chem. 255, 47724780
62. Jones, V. T., Lowe, G., and Potter, B. V. L.
(1980) Eur. J. Biochem. 108, 433437
63. Botting, N. P., and Gani, D. (1992) Biochemistry
31, 15091520
64. Shi, W., Dunbar, J., Jayasekera, M. M. K., Viola,
R. E., and Farber, G. K. (1997) Biochemistry 36,
91369144
65. Jayasekera, M. M. K., Shi, W., Farber, G. K.,
and Viola, R. E. (1997) Biochemistry 36, 9145
9150
66. Goda, S. K., Minton, N. P., Botting, N. P., and
Gani, D. (1992) Biochemistry 31, 1074710756
67. Weiss, P. M., Boerner, R. J., and Cleland, W. W.
(1987) J. Am. Chem. Soc. 109, 72017202
68. Anderson, S. R., Anderson, V. E., and Knowles,
J. R. (1994) Biochemistry 33, 1054510555
68a. Vinarov, D. A., and Nowak, T. (1998) Biochem-
istry 37, 1523815246
69. Vinarov, D. A., and Nowak, T. (1999) Biochem-
istry 38, 1213812149
70. Zhang, E., Hatada, M., Brewer, J. M., and
Lebioda, L. (1994) Biochemistry 33, 62956300
71. Wedekind, J. E., Reed, G. H., and Rayment, I.
(1995) Biochemistry 34, 43254330
72. Duquerroy, S., Camus, C., and Janin, J. (1995)
Biochemistry 34, 1251312523
73. Larsen, T. M., Wedekind, J. E., Rayment, I.,
and Reed, G. H. (1996) Biochemistry 35, 4349
4358
73a. Hubbard, B. K., Koch, M., Palmer, D. R. J.,
Babbitt, P. C., and Gerlt, J. A. (1998) Biochemis-
try 37, 1436914375
73b. Thompson, T. B., Garrett, J. B., Taylor, E. A.,
Meganathan, R., Gerlt, J. A., and Rayment, I.
(2000) Biochemistry 39, 1066210676
74. Linhardt, R. J., Turnbull, J. E., Wang, H. M.,
Loganathan, D., and Gallagher, J. T. (1990)
Biochemistry 29, 26112617
75. Desai, U. R., Wang, H.-M., and Linhardt, R. J.
(1993) Biochemistry 32, 81408145
76. Kiss, J. (1974) Adv. Carbohydrate Chem.
Biochem. 29, 229303
76a. Li, S., Kelly, S. J., Lamani, E., Ferraroni, M.,
and Jedrzejas, M. J. (2000) EMBO J. 19, 1228
1240
1. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, N. Y. (p. 465)
2. Sorensen, P. E., and Jencks, W. P. (1987) J. Am.
Chem. Soc. 109, 46754690
3. Kallen, R. G., and Jencks, W. P. (1966) J. Biol.
Chem. 241, 58455850, 58515863
4. Edsall, J. T., and Wyman, J. (1958) Biophysical
Chemistry, Vol. I, Academic Press, New York
(p. 550ff)
5. Fernley, R. T. (1988) Trends Biochem. Sci. 13,
356359
6. Sly, W. S., and Hu, P. Y. (1995) Ann. Rev.
Biochem. 64, 375401
7. Hazen, S. A., Waheed, A., Sly, W. S., LaNoue,
K. F., and Lynch, C. J. (1996) FASEB J. 10,
481490
8. Heck, R. W., Boriack-Sjodin, P. A., Qian, M.,
Tu, C., Christianson, D. W., Laipis, P. J., and
Silverman, D. N. (1996) Biochemistry 35,
1160511611
9. Maren, T. H. (1985) N. Engl. J. Med. 313, 179
181
10. Stams, T., Nair, S. K., Okuyama, T., Waheed,
A., Sly, W. S., and Christianson, D. W. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1358913594
11. Guilloton, M. B., Korte, J. J., Lamblin, A. F.,
Fuchs, J. A., and Anderson, P. M. (1992) J. Biol.
Chem. 267, 37313734
12. Kisker, C., Schindelin, H., Alber, B. E., Ferry, J.
G., and Rees, D. C. (1996) EMBO J. 15, 2323
2330
13. Rowlett, R. S., Chance, M. R., Wirt, M. D.,
Sidelinger, D. E., Royal, J. R., Woodroffe, M.,
Wang, Y.-F. A., Saha, R. P., and Lam, M. G.
(1994) Biochemistry 33, 1396713976
13a. Kimber, M. S., and Pai, E. F. (2000) EMBO J.
19, 1407418
14. Miyamoto, H., Miyashita, T., Okushima, M.,
Nakano, S., Morita, T., and Matsushiro, A.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 96579660
15. Hkansson, K., Carlsson, M., Svensson, L. A.,
and Liljas, A. (1992) J. Mol. Biol. 227, 11921204
16. Kumar, V., and Kannan, K. K. (1994) J. Mol.
Biol. 241, 226232
17. Kiefer, L. L., Paterno, S. A., and Fierke, C. A.
(1995) J. Am. Chem. Soc. 117, 68316837
17a. Lesburg, C. A., Huang, C.-c, Christianson, D.
W., and Fierke, C. A. (1997) Biochemistry 36,
1578015791
18. Liang, J.-Y., and Lipscomb, W. N. (1987)
Biochemistry 26, 52935301
19. Pocker, Y., and Janjic, N. (1989) J. Am. Chem.
Soc. 111, 731733
19a. Earnhardt, J. N., Qian, M., Tu, C., Lakkis, M.
M., Bergenhem, N. C. H., Laipis, P. J., Tashian,
R. E., and Silverman, D. N. (1998) Biochemistry
37, 1083710845
19b. Denisov, V. P., Jonsson, B.-H., and Halle, B.
(1999) J. Am. Chem. Soc. 121, 23272328
20. Paneth, P., and OLeary, M. H. (1985) J. Am.
Chem. Soc. 107, 73817384
21. Scolnick, L. R., and Christianson, D. W. (1996)
Biochemistry 35, 1642916434
22. Toba, S., Colombo, G., and Merz, K. M., Jr.
(1999) J. Am. Chem. Soc. 121, 22902302
23. Merz, K. M., Jr., and Banci, L. (1997) J. Am.
Chem. Soc. 119, 863871
24. Briganti, F., Mangani, S., Orioli, P., Scozzafa-
va, A., Vernaglione, G., and Supuran, C. T.
(1997) Biochemistry 36, 1038410392
25. Ren, X., Tu, C., Laipis, P. J., and Silverman, D.
N. (1995) Biochemistry 34, 84928498
26. Malatesta, V., and Cocivera, M. (1978) Journal
of Organic Chemistry 43, 17371742
27. Jencks, W. P. (1987) Catalysis in Chemistry and
Enzymology, Dover, Mineola, New York
28. Engel, C. K., Mathieu, M., Zeelen, J. P.,
Hiltunen, J. K., and Wierenga, R. K. (1996)
EMBO J. 15, 51355145
References
28a. Engel, C. K., Kiema, T. R., Hiltunen, J. K., and
Wierenga, R. K. (1998) J. Mol. Biol. 275, 847
859
29. DOrdine, R. L., Tonge, P. J., Carey, P. R., and
Anderson, V. E. (1994) Biochemistry 33, 12635
12643
30. Bahnson, B. J., and Anderson, V. E. (1991)
Biochemistry 30, 58945906
31. DOrdine, R. L., Bahnson, B. J., Tonge, P. J.,
and Anderson, V. E. (1994) Biochemistry 33,
1473314742
31a. Hofstein, H. A., Feng, Y., Anderson, V. E., and
Tonge, P. J. (1999) Biochemistry 38, 95089516
32. Yang, S.-Y., He, X.-Y., and Schulz, H. (1995)
Biochemistry 34, 64416447
33. Annand, R. R., Kozlowski, J. F., Davisson, V.
J., and Schwab, J. (1993) J. Am. Chem. Soc. 115,
10881094
34. Brock, D. J. H., Kass, L. R., and Bloch, K.
(1967) J. Biol. Chem. 242, 44324440
35. Hebert, H., Schmidt-Krey, I., Morgenstern, R.,
Murata, K., Hirai, T., Mitsuoka, K., and
Fujiyoshi, Y. (1997) J. Mol. Biol. 271, 751758
36. Hu, L., Borleske, B. L., and Colman, R. F.
(1997) Protein Sci. 6, 4352
37. Rushmore, T. H., and Pickett, C. B. (1993) J.
Biol. Chem. 268, 1147511478
38. Ji, X., Johnson, W. W., Sesay, M. A., Dickert, L.,
Prasad, S. M., Ammon, H. L., Armstrong, R.
N., and Gilliland, G. L. (1994) Biochemistry 33,
10431052
39. Oakley, A. J., Rossjohn, J., Lo Bello, M.,
Caccuri, A. M., Federici, G., and Parker, M. W.
(1997) Biochemistry 36, 576585
40. Reinemer, P., Prade, L., Hof, P., Neuefeind, T.,
Huber, R., Zettl, R., Palme, K., Schell, J.,
Koelln, I., Bartunik, H. D., and Bieseler, B.
(1996) J. Mol. Biol. 255, 289309
41. Xiao, G., Liu, S., Ji, X., Johnson, W. W., Chen,
J., Parsons, J. F., Stevens, W. J., Gilliland, G. L.,
and Armstrong, R. N. (1996) Biochemistry 35,
47534765
41a. Neuefeind, T., Huber, R., Reinemer, P.,
Knblein, J., Prade, L., Mann, K., and Bieseler,
B. (1997) J. Mol. Biol. 274, 577587
42. McTigue, M. A., Williams, D. R., and Tainer, J.
A. (1995) J. Mol. Biol. 246, 2127
43. Yang, G., Liang, P.-H., and Dunaway-
Mariano, D. (1994) Biochemistry 33, 83278331
44. Crooks, G. P., Xu, L., Barkley, R. M., and
Copley, S. D. (1995) J. Am. Chem. Soc. 117,
1079110798
45. Yang, G., Liu, R.-Q., Taylor, K. L., Xiang, H.,
Price, J., and Dunaway-Mariano, D. (1996)
Biochemistry 36, 1087910885
46. Benning, M. M., Taylor, K. L., Liu, R.-Q., Yang,
G., Xiang, H., Wesenberg, G., Dunaway-
Mariano, D., and Holden, H. M. (1996)
Biochemistry 35, 81038109
47. Taylor, K. L., Liu, R.-Q., Liang, P.-H., Price, J.,
Dunaway-Mariano, D., Tonge, P. J., Clarkson,
J., and Carey, P. R. (1995) Biochemistry 34,
1388113888
48. Clarkson, J., Tonge, P. J., Taylor, K. L.,
Dunaway-Mariano, D., and Carey, P. R. (1997)
Biochemistry 36, 1019210199
49. Taylor, K. L., Xiang, H., Liu, R.-Q., Yang, G.,
and Dunaway-Mariano, D. (1997) Biochemistry
36, 13491361
49a. Benning, M. M., Haller, T., Gerlt, J. A., and
Holden, H. M. (2000) Biochemistry 39, 4630
4639
49b. Xiang, H., Luo, L., Taylor, K. L., and Dun-
away-Mariano, D. (1999) Biochemistry 38,
76387652
49c. Babbitt, P. C., and Gerlt, J. A. (1997) J. Biol.
Chem. 272, 3059130594
714 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
References
76b. Ponnuraj, K., and Jedrzejas, M. J. (2000) J. Mol.
Biol. 299, 885895
77. Yoder, M. D., Keen, N. T., and Jurnak, F.
(1993) Science 260, 15031507
78. Cohen, F. E. (1993) Science 260, 14441445
79. Kita, N., Boyd, C. M., Garrett, M. R., Jurnak,
F., and Keen, N. T. (1996) J. Biol. Chem. 271,
2652926535
80. Kuo, D. J., and Rose, I. A. (1987) Biochemistry
26, 75897596
80a. Lloyd, S. J., Lauble, H., Prasad, G. S., and
Stout, C. D. (1999) Protein Sci. 8, 26552662
81. Werst, M. M., Kennedy, M. C., Houseman, A.
L. P., Beinert, H., and Hoffman, B. M. (1990)
Biochemistry 29, 1053310540
82. Kilpatrick, L. K., Kennedy, M. C., Beinert, H.,
Czernuszewicz, R. S., Qiu, D., and Spiro, T. G.
(1994) J. Am. Chem. Soc. 116, 40534061
83. Lauble, H., Kennedy, M. C., Beinert, H., and
Stout, C. D. (1992) Biochemistry 31, 27352748
84. Kennedy, M. C., Emptage, M. H., and Beinert,
H. (1984) J. Biol. Chem. 259, 31453151
85. Kennedy, M. C., and Beinert, H. (1988) J. Biol.
Chem. 263, 81948198
86. Beinert, H., and Kennedy, M. C. (1993) FASEB
J. 7, 14421448
87. Kent, T. A., Emptage, M. H., Merkle, H.,
Kennedy, M. C., Beinert, H., and Mnck, E.
(1985) J. Biol. Chem. 260, 68716881
88. Lauble, H., Kennedy, M. C., Beinert, H., and
Stout, C. D. (1994) J. Mol. Biol. 237, 437451
89. Zheng, L., Kennedy, M. C., Beinert, H., and
Zalkin, H. (1992) J. Biol. Chem. 267, 78957903
90. Gruer, M. J., Artymiuk, P. J., and Guest, J. R.
(1997) Trends Biochem. Sci. 22, 36
91. Flint, D. H. (1993) Biochemistry 32, 799805
92. Flint, D. H., and McKay, R. G. (1994) J. Am.
Chem. Soc. 116, 55345539
93. Hofmeister, A. E. M., Grabowski, R., Linder,
D., and Buckel, W. (1993) Eur. J. Biochem. 215,
341349
94. Grabowski, R., Hofmeister, A. E. M., and
Buckel, W. (1993) Trends Biochem. Sci. 18, 297
300
95. Hofmeister, A. E. M., Berger, S., and Buckel,
W. (1992) Eur. J. Biochem. 205, 743749
96. Schroepfer, G. J., Jr. (1966) J. Biol. Chem. 241,
54415447
97. Hill, R. K., and Newkome, G. R. (1969) J. Am.
Chem. Soc. 91, 58935894
98. Chiang, Y., and Kresge, A. J. (1991) Science
253, 395400
99. Vellom, D. C., Radic, Z., Li, Y., Pickering, N.
A., Camp, S., and Taylor, P. (1993) Biochemistry
32, 1217
100. Gerlt, J. A., and Gassman, P. G. (1993)
Biochemistry 32, 1194311952
101. Petsko, G. A., Kenyon, G. L., Gerlt, J. A.,
Ringe, D., and Kozarich, J. W. (1993) Trends
Biochem. Sci. 18, 372376
102. Neidhart, D. J., Howell, P. L., Petsko, G. A.,
Powers, V. M., Li, R., Kenyon, G. L., and
Gerlt, J. A. (1991) Biochemistry 30, 92649273
103. St. Maurice, M., and Bearne, S. L. (2000)
Biochemistry 39, 1332413335
104. Schafer, S. L., Barrett, W. C., Kallarakal, A. T.,
Mitra, B., Kozarich, J. W., Gerlt, J. A., Clifton,
J. G., Petsko, G. A., and Kenyon, G. L. (1996)
Biochemistry 35, 56625669
105. Babbitt, P. C., Hasson, M. S., Wedekind, J. E.,
Palmer, D. R. J., Barrett, W. C., Reed, G. H.,
Rayment, I., Ringe, D., Kenyon, G. L., and
Gerlt, J. A. (1996) Biochemistry 35, 1648916501
106. Babbitt, P. C., Mrachko, G. T., Hasson, M. S.,
Huisman, G. W., Kolter, R., Ringe, D., Petsko,
G. A., Kenyon, G. L., and Gerlt, J. A. (1995)
Science 267, 11591161
107. Chiang, Y., Kresge, A. J., Pruszynski, P.,
Schepp, N. P., and Wirz, J. (1990) Angew.
Chem. Int. Ed. Engl. 29, 792794
108. Guthrie, J. P., and Kluger, R. (1993) J. Am.
Chem. Soc. 115, 1156911572
109. Gerlt, J. A., Kozarich, J. W., Kenyon, G. L., and
Gassman, P. G. (1991) J. Am. Chem. Soc. 113,
96679669
110. Gerlt, J. A., and Gassman, P. G. (1993) J. Am.
Chem. Soc. 115, 1155211568
111. Guthrie, J. P. (1997) J. Am. Chem. Soc. 119,
11511152
112. Tobin, J. B., and Frey, P. A. (1996) J. Am. Chem.
Soc. 118, 1225312260
113. Yamauchi, T., Choi, S.-Y., Okada, H., Yohda,
M., Kumagai, H., Esaki, N., and Soda, K.
(1992) J. Biol. Chem. 267, 1836118364
114. Glavas, S., and Tanner, M. E. (1999) Biochemis-
try 38, 41064113
115. Ho, H.-T., Falk, P. J., Ervin, K. M., Krishnan, B.
S., Discotto, L. F., Dougherty, T. J., and Pucci,
M. J. (1995) Biochemistry 34, 24642470
115a. Hwang, K. Y., Cho, C.-S., Kim, S. S., Sung, H.-
C., Yu, Y. G., and Cho, Y. (1999) Nature Struct.
Biol. 6, 422426
116. Stein, T., Kluge, B., Vater, J., Franke, P., Otto,
A., and Wittmann-Liebold, B. (1995) Biochem-
istry 34, 46334642
117. Wiseman, J. S., and Nichols, J. S. (1984) J. Biol.
Chem. 259, 89078914
118. Shikata, Y., Watanabe, T., Teramoto, T., Inoue,
A., Kawakami, Y., Nishizawa, Y., Katayama,
K., and Kuwada, M. (1995) J. Biol. Chem. 270,
1671916723
119. Heck, S. D., Faraci, W. S., Kelbaugh, P. R.,
Saccomano, N. A., Thadeio, P. F., and
Volkmann, R. A. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 40364039
120. Johnson, J. D., Creighton, D. J., and Lambert,
M. R. (1986) J. Biol. Chem. 261, 45354541
121. Cooper, A. J. L., Gines, J. Z., and Meister, A.
(1983) Chem. Rev. 83, 321358
122. Pirrung, M. C., Chen, J., Rowley, E. G., and
McPhail, A. T. (1993) J. Am. Chem. Soc. 115,
71037110
123. Noltmann, E. A. (1972) in The Enzymes, 3rd
ed., Vol. 6 (Boyer, P. D., ed), pp. 271354,
Academic Press, New York
124. McGee, D. M., Hathaway, G. H., Palmieri, R.
H., and Noltmann, E. A. (1980) J. Mol. Biol.
142, 2942
125. Mushegian, A. R., and Koonin, E. V. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1026810273
125a. Jeffery, C. J., Bahnson, B. J., Chien, W., Ringe,
D., and Petsko, G. A. (2000) Biochemistry 39,
955964
126. Tanaka, K. R., and Paglia, D. E. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease (Scriver, C. R., Beaudet, A. L., Sly, W.
S., and Valle, D., eds), pp. 34853511,
McGraw-Hill, New York
127. Woodruff, W. W., III, and Wolfenden, R.
(1979) J. Biol. Chem. 254, 58665867
127a. Seemann, J. E., and Schulz, G. E. (1997) J. Mol.
Biol. 273, 256268
128. Wallace, L. J., Eiserling, F. A., and Wilcox, G.
(1978) J. Biol. Chem. 253, 37173720
129. Bogumil, R., Kappl, R., Httermann, J., and
Witzel, H. (1997) Biochemistry 36, 23452352
130. Speck, J. C., Jr. (1958) Adv. Carbohydrate Chem.
13, 63103
131. Gibson, D. R., Gracy, R. W., and Hartman, F.
C. (1980) J. Biol. Chem. 255, 93699374
132. Balaban, R. S., and Ferretti, J. A. (1983) Proc.
Natl. Acad. Sci. U.S.A. 80, 12411245
133. Blacklow, S. C., Raines, R. T., Lim, W. A.,
Zamore, P. D., and Knowles, J. R. (1988)
Biochemistry 27, 11581167
134. Muirhead, H. (1983) Trends Biochem. Sci. 8,
326330
135. Lolis, E., Alber, T., Davenport, R. C., Rose, D.,
Hartman, F. C., and Petsko, G. A. (1990)
Biochemistry 29, 66096618
136. Joseph-McCarthy, D., Lolis, E., Komives, E.
A., and Petsko, G. A. (1994) Biochemistry 33,
28152823
137. Mande, S. C., Mainfroid, V., Kalk, K. H.,
Goraj, K., Martial, J. A., and Hol, W. G. J.
(1994) Protein Sci. 3, 810821
138. Davenport, R. C., Bash, P. A., Seaton, B. A.,
Karplus, M., Petsko, G. A., and Ringe, D.
(1991) Biochemistry 30, 58215826
139. Verlinde, C. L. M. J., Witmans, C. J., Pijning,
T., Kalk, K. H., Hol, W. G. J., Callens, M., and
Opperdoes, F. R. (1992) Protein Sci. 1, 15781584
140. Rose, I. A., Fung, W.-J., and Warms, J. V. B.
(1990) Biochemistry 29, 43124317
140a. Harris, T. K., Cole, R. N., Comer, F. I., and
Mildvan, A. S. (1998) Biochemistry 37, 16828
16838
141. Joseph-McCarthy, D., Rost, L. E., Komives, E.
A., and Petsko, G. A. (1994) Biochemistry 33,
28242829
142. Belasco, J. G., and Knowles, J. R. (1980)
Biochemistry 19, 472477
143. Alston, W. C., II, Kanska, M., and Murray, C.
J. (1996) Biochemistry 35, 1287312881
144. Nickbarg, E. B., Davenport, R. C., Petsko, G.
A., and Knowles, J. R. (1988) Biochemistry 27,
59485960
145. Lodi, P. J., Chang, L. C., Knowles, J. R., and
Komives, E. A. (1994) Biochemistry 33, 2809
2814
146. Raines, R. T., and Knowles, J. R. (1987)
Biochemistry 26, 70147020
147. Bash, P. A., Field, M. J., Davenport, R. C.,
Petsko, G. A., Ringe, D., and Karplus, M.
(1991) Biochemistry 30, 58265832
148. Lodi, P. J., and Knowles, J. R. (1991) Biochemis-
try 30, 69486956
149. Alagona, G., Ghio, C., and Kollman, P. A.
(1995) J. Am. Chem. Soc. 117, 98559862
150. Pompliano, D. L., Peyman, A., and Knowles,
J. R. (1990) Biochemistry 29, 31863194
151. Yksel, K. ., Sun, A.-Q., Gracy, R. W., and
Schnackerz, K. D. (1994) J. Biol. Chem. 269,
50055008
152. Williams, J. C., and McDermott, A. E. (1995)
Biochemistry 34, 83098319
153. Carrell, H. L., Rubin, B. H., Hurley, T. J., and
Glusker, J. P. (1984) J. Biol. Chem. 259, 3230
3236
154. Farber, G. K., Glasfeld, A., Tiraby, G., Ringe,
D., and Petsko, G. A. (1989) Biochemistry 28,
72897297
155. Collyer, C. A., Goldberg, J. D., Viehmann, H.,
Blow, D. M., Ramsden, N. G., Fleet, G. W. J.,
Montgomery, F. J., and Grice, P. (1992)
Biochemistry 31, 1221112218
156. Jenkins, J., Janin, J., Rey, F., Chiadmi, M., van
Tilbeurgh, H., Lasters, I., De Maeyer, M., Van
Belle, D., Wodak, S. J., Lauwereys, M.,
Stanssens, P., Mrabet, N. T., Snauwaert, J.,
Matthyssens, G., and Lambeir, A.-M. (1992)
Biochemistry 31, 54495458
157. Whitaker, R. D., Cho, Y., Cha, J., Carrell, H. L.,
Glusker, J. P., Karplus, P. A., and Batt, C. A.
(1995) J. Biol. Chem. 270, 2289522906
158. Allen, K. N., Lavie, A., Petsko, G. A., and
Ringe, D. (1995) Biochemistry 34, 37423749
159. Hall, S. S., Doweyko, A. M., and Jordan, F.
(1978) J. Am. Chem. Soc. 100, 5934
160. Xue, L., Talalay, P., and Mildvan, A. S. (1991)
Biochemistry 30, 1085810865
161. Brooks, B., and Benisek, W. F. (1994) Biochem-
istry 33, 26822687
162. Austin, J. C., Zhao, Q., Jordan, T., Talalay, P.,
Mildvan, A. S., and Spiro, T. G. (1995)
Biochemistry 34, 44414447
715
214. Wlassics, I. D., and Anderson, V. E. (1989)
Biochemistry 28, 16271633
215. Usher, K. C., Remington, S. J., Martin, D. P.,
and Drueckhammer, D. G. (1994) Biochemistry
33, 77537759
215a. Kurz, L. C., Nakra, T., Stein, R., Plungkhen,
W., Riley, M., Hsu, F., and Drysdale, G. R.
(1998) Biochemistry 37, 97249737
216. Schwartz, B., Drueckhammer, D. G., Usher, K.
C., and Remington, S. J. (1995) Biochemistry 34,
1545915466
216a. Gu, Z., Drueckhammer, D. G., Kurz, L., Liu,
K., Martin, D. P., and McDermott, A. (1999)
Biochemistry 38, 80228031
216b. Mulholland, A. J., Lyne, P. D., and Karplus,
M. (2000) J. Am. Chem. Soc. 122, 534535
216c. Howard, B. R., Endrizzi, J. A., and Reming-
ton, S. J. (2000) Biochemistry 39, 31563168
217. Smith, S. (1994) FASEB J. 8, 12481259
218. Misra, I., Narasimhan, C., and Miziorko, H.
M. (1993) J. Biol. Chem. 268, 1212912135
219. Misra, I., and Miziorko, H. M. (1996) Biochem-
istry 35, 96109616
220. Guynn, R. W., and Veech, R. L. (1979) J. Biol.
Chem. 254, 16911698
221. Linn, T. C., and Srere, P. A. (1979) J. Biol.
Chem. 254, 16911698
222. Ranganathan, N. S., Linn, T. C., and Srere, P.
A. (1982) J. Biol. Chem. 257, 698702
223. Elshourbagy, N. A., Near, J. C., Kmetz, P. J.,
Sathe, G. M., Southan, C., Strickler, J. E.,
Gross, M., Young, J. F., Wells, T. N. C., and
Groot, P. H. E. (1990) J. Biol. Chem. 265, 1430
1435
224. Pentyala, S. N., and Benjamin, W. B. (1995)
Biochemistry 34, 1096110969
225. Hersch, L. B. (1973) J. Biol. Chem. 248, 7295
7303
226. Nilekani, S., and SivaRaman, C. (1983)
Biochemistry 22, 46574663
227. Oppenheimer, N. J., Singh, M., Sweeley, C. C.,
Sung, S.-J., and Srere, P. A. (1979) J. Biol. Chem.
254, 10001002
228. Ko, Y. H., Vanni, P., Munske, G. R., and
McFadden, B. A. (1991) Biochemistry 30, 7451
7456
229. Lenz, H., Buckel, W., Wunderwald, P.,
Biedermann, G., Buschmeier, V., Eggerer, H.,
Cornforth, J. W., Redmond, J. W., and Mallaby,
R. (1971) Eur. J. Biochem. 24, 207215
230. Retey, J., Luthy, J., and Arigoni, D. (1970)
Nature (London) 226, 519521
231. Kyte, J. (1995) Mechanism in Protein Chemistry,
Garland Publ., New York (pp. 293313)
232. Leussing, D. L., and Raghavan, N. V. (1980)
J. Am. Chem. Soc. 102, 56355643
233. Steinberger, R., and Westheimer, F. H. (1951)
J. Am. Chem. Soc. 73, 429435
234. Kubala, G., and Martell, A. E. (1982) J. Am.
Chem. Soc. 104, 66026609
235. Waldrop, G. L., Braxton, B. F., Urbauer, J. L.,
Cleland, W. W., and Kiick, D. M. (1994)
Biochemistry 33, 52625267
236. Highbarger, L. A., Gerlt, J. A., and Kenyon, G.
L. (1996) Biochemistry 35, 4146
236a. Stanley, T. M., Johnson, W. H., Jr., Burks, E. A.,
Whitman, C. P., Hwang, C.-C., and Cook, P. F.
(2000) Biochemistry 39, 718726
237. Loeber, G., Infante, A. A., Maurer-Fogy, I.,
Krystek, E., and Dworkin, M. B. (1991) J. Biol.
Chem. 266, 30163021
238. Chou, W.-Y., Liu, M.-Y., Huang, S.-M., and
Chang, G.-G. (1996) Biochemistry 35, 98739879
239. Winning, B. M., Bourguignon, J., and Leaver,
C. J. (1994) J. Biol. Chem. 269, 47804786
240. Wei, C.-H., Chou, W.-Y., and Chang, G.-G.
(1995) Biochemistry 34, 79497954
241. Karsten, W. E., and Cook, P. F. (1994) Biochem-
istry 33, 20962103
163. Zhao, Q., Li, Y.-K., Mildvan, A. S., and Talalay,
P. (1995) Biochemistry 34, 65626572
164. Hawkinson, D. C., Pollack, R. M., and Ambulos,
N. P., Jr. (1994) Biochemistry 33, 1217212183
165. Viger, A., Coustal, S., and Marquet, A. (1983)
J. Am. Chem. Soc. 103, 451458
166. Holman, C. M., and Benisek, W. F. (1994)
Biochemistry 33, 26722681
167. Wu, Z. R., Ebrahimian, S., Zawrotny, M. E.,
Thornburg, L. D., Perez-Alvarado, G. C.,
Brothers, P., Pollack, R. M., and Summers, M.
F. (1997) Science 276, 415418
167a. Kim, S. W., Cha, S.-S., Cho, H.-S., Kim, J.-S.,
Ha, N.-C., Cho, M.-J., Joo, S., Kim, K. K., Choi,
K. Y., and Oh, B.-H. (1997) Biochemistry 36,
1403014036
167b. Choi, G., Ha, N.-C., Kim, S. W., Kim, D.-H.,
Park, S., Oh, B.-H., and Choi, K. Y. (2000)
Biochemistry 39, 903909
167c. Thornburg, L. D., Hnot, F., Bash, D. P.,
Hawkinson, D. C., Bartel, S. D., and Pollack,
R. M. (1998) Biochemistry 37, 1049910506
168. Chen, L. H., Kenyon, G. L., Curtin, F.,
Harayama, S., Bembenek, M. E., Hajipour, G.,
and Whitman, C. P. (1992) J. Biol. Chem. 267,
1771617721
169. Subramanya, H. S., Roper, D. I., Dauter, Z.,
Dodson, E. J., Davies, G. J., Wilson, K. S., and
Wigley, D. B. (1996) Biochemistry 35, 792802
170. Taylor, A. B., Czerwinski, R. M., Johnson, W.
H., Jr., Whitman, C. P., and Hackert, M. L.
(1998) Biochemistry 37, 1469214700
171. Stivers, J. T., Abeygunawardana, C., Mildvan,
A. S., Hajipour, G., and Whitman, C. P. (1996)
Biochemistry 35, 814823
172. Czerwinski, R. M., Harris, T. K., Johnson, W.
H., Jr., Legler, P. M., Stivers, J. T., Mildvan, A.
S., and Whitman, C. P. (1999) Biochemistry 38,
1235812366
173. Fitzgerald, M. C., Chernushevich, I., Stand-
ing, K. G., Kent, S. B. H., and Whitman, C. P.
(1995) J. Am. Chem. Soc. 117, 1107511080
174. Lo, T. W. C., Westwood, M. E., McLellan, A.
C., Selwood, T., and Thornalley, P. J. (1994) J.
Biol. Chem. 269, 3229932305
175. Ridderstrm, M., Saccucci, F., Hellman, U.,
Bergman, T., Principato, G., and Mannervik,
B. (1996) J. Biol. Chem. 271, 319323
176. Richard, J. P. (1991) Biochemistry 30, 45814585
176a. Saadat, D., and Harrison, D. H. T. (2000)
Biochemistry 39, 29502960
177. Lan, Y., Lu, T., Lovett, P. S., and Creighton, D.
J. (1995) J. Biol. Chem. 270, 1295712960
178. Cameron, A. D., Ridderstrm, M., Olin, B.,
Kavarana, M. J., Creighton, D. J., and
Mannervik, B. (1999) Biochemistry 38, 13480
13490
178a. He, M. M., Clugston, S. L., Honek, J. F., and
Matthews, B. W. (2000) Biochemistry 39, 8719
8727
179. Rae, C., ODonoghue, S. I., Bubb, W. A., and
Kuchel, P. W. (1994) Biochemistry 33, 35483559
180. Bito, A., Haider, M., Hadler, I., and Breiten-
bach, M. (1997) J. Biol. Chem. 272, 2150921519
181. Mathieu, M., Modis, Y., Zeelen, J. P., Engel, C.
K., Abagyan, R. A., Ahlberg, A., Rasmussen,
B., Lamzin, V. S., Kunau, W. H., and Wieren-
ga, R. K. (1997) J. Mol. Biol. 273, 714728
181a. Modis, Y., and Wierenga, R. K. (2000) J. Mol.
Biol. 297, 11711182
182. Roberts, J. R., Narasimhan, C., and Miziorko,
H. M. (1995) J. Biol. Chem. 270, 1731117316
183. Narasimhan, C., Roberts, J. R., and Miziorko,
H. M. (1995) Biochemistry 34, 99309935
183a. Vinarov, D. A., and Miziorko, H. M. (2000)
Biochemistry 39, 33603368
183b. Richard, J. P., and Nagorski, R. W. (1999) J.
Am. Chem. Soc. 121, 47634770
184. Marsh, J. J., and Lebherz, H. G. (1992) Trends
Biochem. Sci. 17, 110113
185. Cox, T. M. (1994) FASEB J. 8, 6271
186. Morris, A. J., and Tolan, D. R. (1994) Biochem-
istry 33, 1229112297
187. Sygusch, J., Beaudry, D., and Allaire, M. (1987)
Proc. Natl. Acad. Sci. U.S.A. 84, 78467850
188. Lai, C. Y., and Oshima, T. (1971) Arch.
Biochem. Biophys. 144, 363
188a. Choi, K. H., Mazurkie, A. S., Morris, A. J.,
Utheza, D., Tolan, D. R., and Allen, K. N.
(1999) Biochemistry 38, 1265512664
188b. Dalby, A., Dauter, Z., and Littlechild, J. A.
(1999) Protein Sci. 8, 291297
189. Periana, R. A., Motiu-DeGrood, R., Chiang, Y.,
and Hupe, D. J. (1980) J. Am. Chem. Soc. 102,
39233927
190. Beernink, P. T., and Tolan, D. R. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 53745379
191. Kadonaga, J. T., and Knowles, J. R. (1983)
Biochemistry 22, 130136
191a. Hall, D. R., Leonard, G. A., Reed, C. D., Watt,
C. I., Berry, A., and Hunter, W. N. (1999) J.
Mol. Biol. 287, 383394
192. Qamar, S., Marsh, K., and Berry, A. (1996)
Protein Sci. 5, 154161
193. Dreyer, M. K., and Schulz, G. E. (1996) J. Mol.
Biol. 259, 458466
194. Lebioda, L., Hatada, M. H., Tulinsky, A., and
Mavridis, I. M. (1982) J. Mol. Biol. 162, 445
458
195. Meloche, H. P. (1981) Trends Biochem. Sci. 6,
3841
196. Dekker, E. E., and Kitson, R. P. (1992) J. Biol.
Chem. 267, 1050710514
197. Wong, C.-H., Garcia-Junceda, E., Chen, L.,
Blanco, O., Gijsen, H. J. M., and Steensma, D.
H. (1995) J. Am. Chem. Soc. 117, 33333339
198. Jia, J., Schrken, U., Lindqvist, Y., Sprenger,
G. A., and Schneider, G. (1997) Protein Sci. 6,
119124
199. Higgins, M. J. P., Kornblatt, J. A., and Rudney,
H. (1972) in The Enzymes, 3rd ed., Vol. 7
(Boyer, P. D., ed), pp. 407434, Academic
Press, New York
200. Wiegand, G., and Remington, S. J. (1986) Ann.
Rev. Biophys. Biophys. Chem. 15, 97117
201. Anderson, D. H., and Duckworth, H. W.
(1988) J. Biol. Chem. 263, 21632169
202. Wiegand, G., Remington, S., Deisenhofer, J.,
and Huber, R. (1984) J. Mol. Biol. 174, 205219
203. Rault-Leonardon, M., Atkinson, M. A. L.,
Slaughter, C. A., Moomaw, C. R., and Srere, P.
A. (1995) Biochemistry 34, 257263
204. Russell, R. J. M., Ferguson, J. M. C., Hough, D.
W., Danson, M. J., and Taylor, G. L. (1997)
Biochemistry 36, 99839994
205. Pereira, F. D., Donald, L. J., Hosfield, D. J.,
and Duckworth, H. W. (1994) J. Biol. Chem.
269, 412417
206. Karpusas, M., Holland, D., and Remington, S.
J. (1991) Biochemistry 30, 60246031
207. Chothia, C., and Lesk, A. M. (1985) Trends
Biochem. Sci. 10, 116118
208. Evans, C. T., Kurz, L. C., Remington, S. J., and
Srere, P. A. (1996) Biochemistry 35, 1066110672
209. Remington, S., Wiegand, G., and Huber, R.
(1982) J. Mol. Biol. 158, 111 152
210. Kurz, L. C., Roble, J. H., Nakra, T., Drysdale,
G. R., Buzan, J. M., Schwartz, B., and Drueck-
hammer, D. G. (1997) Biochemistry 36, 3981
3990
211. Kurz, L. C., and Drysdale, G. R. (1987)
Biochemistry 26, 26232627
212. Kurz, L. C., Ackerman, J. J. H., and Drysdale,
G. R. (1985) Biochemistry 24, 452457
213. Clark, J. D., OKeefe, S. J., and Knowles, J. R.
(1988) Biochemistry 27, 59615971
References
716 Chapter 13. Enzymatic Addition, Elimination, Condensation, and Isomerization: Roles for Enolate and Carbocation Intermediates
241a. Liu, D., Karsten, W. E., and Cook, P. F. (2000)
Biochemistry 39, 1195511960
242. Edens, W. A., Urbauer, J. L., and Cleland, W.
W. (1997) Biochemistry 36, 11411147
242a. Gruys, K. J., and Sikorski, J. A. (1998) in
Comprehensive Biological Catalysis. A Mechanis-
tic Reference, Vol. I (Sinnott, M., ed), pp. 273
291, Academic Press, San Diego, California
243. Bolduc, J. M., Dyer, D. H., Scott, W. G., Singer,
P., Sweet, R. M., Koshland, D. E., Jr., and
Stoddard, B. L. (1995) Science 268, 13121318
244. Berdis, A. J., and Cook, P. F. (1993) Biochemis-
try 32, 20412046
245. Dean, A. M., and Dvorak, L. (1995) Protein Sci.
4, 21562167
246. Wallon, G., Kryger, G., Lovett, S. T., Oshima,
T., Ringe, D., and Petsko, G. A. (1997) J. Mol.
Biol. 266, 10161031
247. Brubaker, M. J., Dyer, D. H., Stoddard, B., and
Koshland, D. E., Jr. (1996) Biochemistry 35,
28542864
248. Tipton, P. A. (1993) Biochemistry 32, 28222827
249. Kalckar, H. M. (1985) Trends Biochem. Sci. 10,
132133
250. Peliska, J. A., and OLeary, M. H. (1991) J. Am.
Chem. Soc. 113, 18411842
251. Seeholzer, S. H., Jaworowski, A., and Rose, I.
A. (1991) Biochemistry 30, 727732
252. Matte, A., Tari, L. W., Goldie, H., and Delbaere,
L. T. J. (1997) J. Biol. Chem. 272, 81058108
253. Hwang, S. H., and Nowak, T. (1986) Biochem-
istry 25, 55905595
254. Konopka, J. M., Lardy, H. A., and Frey, P. A.
(1986) Biochemistry 25, 55715575
255. Hlavaty, J. J., and Nowak, T. (1997) Biochemis-
try 36, 33893403
256. Matte, A., Goldie, H., Sweet, R. M., and
Delbaere, L. T. J. (1996) J. Mol. Biol. 256, 126143
257. Arnelle, D. R., and OLeary, M. H. (1992)
Biochemistry 31, 43634368
258. Punekar, N. S., and Lardy, H. A. (1987) J. Biol.
Chem. 262, 67146719
259. Roseler, W. J., Vandenbark, G. R., and Hanson,
R. H. (1989) J. Biol. Chem. 264, 96579664
260. Rohrer, S. D., Saz, H. J., and Nowak, T. (1986)
J. Biol. Chem. 261, 1304913055
261. Hebdo, C. A., and Nowak, T. (1982) J. Biol.
Chem. 257, 55035514
262. Hartman, F. C., and Harpel, M. R. (1994) Ann.
Rev. Biochem. 63, 197234
263. Wasmann, C. C., Reiss, B., and Bohnert, H. J.
(1988) J. Biol. Chem. 263, 617619
264. Shibata, N., Inoue, T., Fukuhara, K., Nagara,
Y., Kitagawa, R., Harada, S., Kasai, N.,
Uemura, K., Kato, K., Yokota, A., and Kai, Y.
(1996) J. Biol. Chem. 271, 2644926452
265. Tessier, L. H., Paulus, F., Keller, M., Vial, C.,
and Imbault, P. (1995) J. Mol. Biol. 245, 2233
266. Holzenburg, A., Mayer, F., Harauz, G., van
Heel, M., Tokuoka, R., Ishida, T., Harata, K.,
Pal, G. P., and Saenger, W. (1987) Nature
(London) 325, 730732
267. Erijman, L., Lorimer, G. H., and Weber, G.
(1993) Biochemistry 32, 51875195
268. Morse, D., Salois, P., Markovic, P., and
Hastings, J. W. (1995) Science 268, 16221624
269. Andersson, I. (1996) J. Mol. Biol. 259, 160174
269a. Schreuder, H. A., Knight, S., Curmi, P. M. G.,
Andersson, I., Cascio, D., Sweet, R. M.,
Brndn, C.-I., and Eisenberg, D. (1993)
Protein Sci. 2, 11361146
270. Newman, J., and Gutteridge, S. (1993) J. Biol.
Chem. 268, 2587625886
271. Bassham, J. A., Benson, A. A., Kay, L. D.,
Harris, A. Z., Wilson, A. T., and Calvin, M.
(1954) J. Am. Chem. Soc. 76, 17601770
272. Siegel, M. I., and Lane, M. D. (1973) J. Biol.
Chem. 248, 54865498
References
272a. Taylor, T. C., and Andersson, I. (1996) Nature
Struct. Biol. 3, 95101
272b. Harpel, M. R., Larimer, F. W., and Hartman, F.
C. (1998) Protein Sci. 7, 730738
272c. Harpel, M. R., and Hartman, F. C. (1996)
Biochemistry 35, 1386513870
272d. Duff, A. P., Andrews, T. J., and Curmi, R. M.
G. (2000) J. Mol. Biol. 298, 903916
273. Pierce, J., Andrews, T. J., and Lorimer, G. H.
(1986) J. Biol. Chem. 261, 1024810256
273a. Berry, J. A., Lorimer, G. H., Pierce, J., Seeman,
J. R., Meeks, J., and Freas, S. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 734738
274. Jaworowski, A., Hartman, F. C., and Rose, I.
A. (1984) J. Biol. Chem. 259, 67836789
275. Saver, B. G., and Knowles, J. R. (1982)
Biochemistry 21, 53985403
276. Taylor, T. C., and Andersson, I. (1997) J. Mol.
Biol. 265, 432444
277. Roeske, C. A., and OLeary, M. H. (1984)
Biochemistry 23, 62756284
278. Van Dyke, D. E., and Schloss, J. V. (1986)
Biochemistry 25, 51455156
279. van de Loo, F. J., and Salvucci, M. E. (1996)
Biochemistry 35, 81438148
280. de Jimnez, E. S., Medrano, L., and Martnez-
Barajas, E. (1995) Biochemistry 34, 28262831
281. Larson, E. M., OBrien, C. M., Zhu, G.,
Spreitzer, R. J., and Portis, A. R., Jr. (1997) J.
Biol. Chem. 272, 1703317037
282. Mueller, D. D., Schmidt, A., Pappan, K. L.,
McKay, R. A., and Schaefer, J. (1995) Biochem-
istry 34, 55975603
282a. Ogren, W. L. (1984) Ann. Rev. Plant Physiol. 35,
415442
283. Tse, J. M.-T., and Schloss, J. V. (1993) Biochem-
istry 32, 1039810403
283a. Sugawara, H., Yamamoto, H., Shibata, N.,
Inoue, T., Okada, S., Miyake, C., Yokota, A.,
and Kai, Y. (1999) J. Biol. Chem. 274, 15655
15661
284. Harpel, M. R., Serpersu, E. H., Lamerdin, J.
A., Huang, Z.-H., Gage, D. A., and Hartman,
F. C. (1995) Biochemistry 34, 1129611306
284a. Gutteridge, S., Rhoades, D. F., and Herrmann,
C. (1993) J. Biol. Chem. 268, 78187824
285. Morell, M. K., Wilkin, J.-M., Kane, H. J., and
Andrews, T. J. (1997) J. Biol. Chem. 272, 5445
5451
286. Krebs, H. A., and Roughton, F. J. W. (1948)
Biochem. J. 43, 550555
287. Kaziro, Y., Hass, L. F., Boyer, P. D., and Ochoa,
S. (1962) J. Biol. Chem. 237, 14601468
288. Cooper, T. G., Tchen, T. T., Wood, H. G., and
Benedict, C. R. (1968) J. Biol. Chem. 243, 3857
3863
289. Gonzlez, D. H., and Andreo, C. S. (1988)
Biochemistry 27, 177183
290. Harpster, M. H., and Taylor, W. C. (1986) J.
Biol. Chem. 261, 61326136
291. OLeary, M. H., Rife, J. E., and Slater, J. D.
(1981) Biochemistry 20, 73087314
292. Hansen, D. E., and Knowles, J. R. (1982) J.
Biol. Chem. 257, 1479514798
293. Fujita, N., Izui, K., Nishino, I., and Katsuki, H.
(1984) Biochemistry 23, 17741779
294. Gonzlez, D. H., and Andreo, C. S. (1989)
Trends Biochem. Sci. 14, 2427
295. Knowles, J. R. (1989) Ann. Rev. Biochem. 58,
195221
296. Janc, J. W., Urbauer, J. L., OLeary, M. H., and
Cleland, W. W. (1992) Biochemistry 31, 6432
6440
297. Lee, S.-L., Hepburn, T. W., Swartz, W. H.,
Ammon, H. L., Mariano, P. S., and Dunaway-
Matiano, D. (1992) J. Am. Chem. Soc. 114,
73467354
298. McQueney, M. S., Lee, S.-l, Bowman, E.,
Mariano, P. S., and Dunaway-Mariano, D.
(1989) J. Am. Chem. Soc. 111, 68856887
299. Seidel, H. M., and Knowles, J. R. (1994)
Biochemistry 33, 56415646
300. Bowman, E., McQueney, M., Barry, R. J., and
Dunaway-Mariano, D. (1988) J. Am. Chem.
Soc. 110, 55755576
301. Hidaka, T., and Seto, H. (1989) J. Am. Chem.
Soc. 111, 80128013
302. Freeman, S., Pollack, S. J., and Knowles, J. R.
(1992) J. Am. Chem. Soc. 114, 377378
302a. Morais, M. C., Zhang, W., Baker, A. S., Zhang,
G., Dunaway-Mariano, D., and Allen, K. N.
(2000) Biochemistry 39, 1038510396
303. Mortimer, C. E., and Niehaus, W. G., Jr. (1974)
J. Biol. Chem. 249, 28332842
304. Clifford, K., Cornforth, J. W., Mallaby, R., and
Phillips, G. T. (1971) J. Chem. Soc. Chem.
Commun., 15991600
305. Reardan, J. E., and Abeles, R. H. (1986)
Biochemistry 25, 56095616
306. Poulter, C. D., Muehlbacker, M., and Davis, D.
R. (1989) J. Am. Chem. Soc. 111, 37403742
307. Street, I. P., Coffman, H. R., Baker, J. A., and
Poulter, C. D. (1994) Biochemistry 33, 42124217
307a. Oh, S. K., Han, K. H., Ryu, S. B., and Kang, H.
(2000) J. Biol. Chem. 275, 1848218488
308. Cane, D. E., Abell, C., Harrison, P. H.,
Hubbard, B. R., Kane, C. T., Lattman, R.,
Oliver, J. S., and Weiner, S. W. (1991) Philos
Trans R Soc Lond B Biol Sci 332, 123129
309. Cane, D. E., Pawlak, J. L., and Horak, R. M.
(1990) Biochemistry 29, 54765490
310. Aulabaugh, A., and Schloss, J. V. (1990)
Biochemistry 29, 28242830
311. Biou, V., Dumas, R., Cohen-Addad, C., Douce,
R., Job, D., and Pebay-Peyroula, E. (1997)
EMBO J. 16, 34053415
312. Halgand, F., Dumas, R., Biou, V., Andrieu, J.-
P., Thomazeau, K., Gagnon, J., Douce, R., and
Forest, E. (1999) Biochemistry 38, 60256034
313. Proust-De Martin, F., Dumas, R., and Field,
M. J. (2000) J. Am. Chem. Soc. 122, 76887697
717
Study Questions
CH
2
OH
HC OH
C O
(HC OH)
2
CH
2
OP
1. Discuss the role of carbonyl groups in facilitating
reactions of metabolism.
2. Write a step-by-step sequence showing the chemical
mechanisms involved in the action of a type I
aldolase that catalyzes cleavage of fructose 1,6-
bisphosphate. The enzyme is inactivated by sodium
borohydride in the presence of the substrate.
Explain this inactivation.
3. Some methylotrophic bacteria dehydrogenate
methanol to formaldehyde. The latter undergoes
an aldol condensation to form a hexulose-6-phos-
phate:
Write a reasonable cycle of biochemical reactions
(utilizing this compound) by which three molecules
of methanol can yield one molecule of glyceralde-
hyde 3-phosphate, a compound that can be either
catabolized for energy or utilized for biosynthesis.
4. Malate is formed in the glyoxylate pathway by
reaction of glyoxylate with Acetyl-CoA. Indicate
the chemical mechanism of the reactions involved
and structure of an intermediate species.
5. Acetyl-CoA is condensed with acetoacetyl-CoA to
give an intermediate which is eventually converted
to mevalonate.
a) Show the sequence of reactions leading from
acetyl-CoA to mevalonate with the structures
of important intermediates.
b) Mevalonate is an important intermediate on
the pathway leading to what class of compounds?
6. a) Show the structures of the reactants for the
3-hydroxy-3-methylglutaryl-CoA synthase
reaction.
b) Free coenzyme A is liberated in the above
reaction. From which of the two reactants did
it come? Explain the metabolic significance of
the liberation of free CoA.
7. Phosphoenolpyruvate carboxykinase catalyzes the
following reaction:
Oxaloacetate + GTP CO
2
+
phosphoenolpyruvate + GDP
Illustrate the probable mechanism of the reaction.
8. Write a complete step-by-step mechanism for the
action of ATP-citrate lyase which catalyzes the
following reaction (Eq. 13-39):
ATP + citrate + CoA SH Acetyl CoA +
oxaloacetate + ADP + P
i
9. Malonyl-CoA synthetase forms its product from
free malonate and MgATP. Both phospho-enzyme
and malonyl-enzyme intermediates have been
detected. Suggest a sequential mechanism of
action. See Kim, Y. S., and Lee, J. K. (1986) J. Biol.
Chem. 261, 1629516297.
10. The carboxylation of acetone with HCO
3

to form
acetoacetate (the reverse of Eq. 13-44) is not a
thermodynamically spontaneous process (G
~17 kJ mol
1
). An anaerobic strain of Xanthobacter
couples this reaction to the cleavage of ATP to
AMP + 2 P
i
. Suggest possible mechanisms. See
Sluis, M. K., and Ensign, S. A. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 84568461.
11. The reaction of Eq. 10-11 begins with a hydroxyla-
tion, which can be viewed as insertion of an oxygen
atom into a CH bond. Draw the probable inter-
mediate formed in this reaction and indicate how
it can be converted to the final products.
12. Cyanase from E. coli decomposes cyanate (NCO

)
to ammonia and CO
2
. Isotopic labeling studies
show that the second oxygen atom in the product
CO
2
comes not from H
2
O but from a second sub-
strate, bicarbonate ion. The correct equation for
the enzymatic process is:
NCO

+ H
+
+ HCO
3

NH
2
COO

+ CO
2
Propose a sequence of steps for this enzymatic
reaction, which has been investigated by Ander-
son and coworkers. Johnson, W. V., and Ander-
son, P. M. (1987) J. Biol. Chem. 262, 90219025 and
Anderson, P. M., Korte, J. J., Holcomb, T. A., Cho,
Y.-g, Son, C.-m, and Sung, Y.-c. (1994) J. Biol. Chem.
269, 1503615045.
718 Chapter 14. Coenzymes: Natures Special Reagents
Contents
719
720
723
724
725
725
726
727
729
729
730
730
731
733
734
736
736
737
737
740
741
741
744
745
745
746
747
747
749
750
750
750
751
753
754
755
755
758
763
A. ATP and the Nucleotide Handles
B. Coenzyme A and Phosphopantetheine
C. Biotin and the Formation of Carboxyl Groups
from Bicarbonate
1. Biotin-Containing Enzymes
2. The Mechanism of Biotin Action
Carboxybiotin
Carboxyphosphate
The carboxylation step
3. Control Mechanisms
4. Pumping Ions with the Help of Biotin
D. Thiamin Diphosphate
1. Chemical Properties of Thiamin
2. Catalytic Mechanisms
3. Structures of Thiamin-Dependent Enzymes
4. The Variety of Enzymatic Reactions Involving
Thiamin
5. Oxidative Decarboxylation and 2-Acetylthiamin
Diphosphate
6. Thiamin Coenzymes in Nerve Action
E. Pyridoxal Phosphate
1. Nonenzymatic Models
2. A General Mechanism of Action of PLP
3. The Variety of PLP-Dependent Reactions
Loss of the -Hydrogen (Group a)
Decarboxylation (Group b)
Side chain cleavage (Group c)
Ketimine intermediate as electron acceptor
(Group d)
Glycogen phosphorylase
4. Pyridoxamine Phophate as a Coenzyme
5. Stereochemistry of PLP-Requiring Enzymes
6. Seeing Changes in the Optical Properties of the
Coenzyme
Imine groups in free enzymes
Absorption bands at 500 nm
7. Atomic Structures
8. Constructing a Detailed Picture of the Action of a
PLP Enzyme
F. Pyruvoyl Groups and Other Unusual Electrophilic
Centers
1. Decarboxylases
2. Proline and Glycine Reductases
3. Dehydroalanine and Histidine and Phenylalanine
Ammonia-Lyases
References
Study Questions
721
728
738
756
725
735
743
753
Boxes
Box 14-A Discovery of the Vitamins
Box 14-B The Biotin-Binding Proteins Avidin and
Streptavidin
Box 14-C The Vitamin B
6
Family: Pyridoxine,
Pyridoxal, and Pyridoxamine
Box 14-D Dietary Requirements for B Vitamins
Tables
Table 14-1 Enzymes Containing Bound Biotin
Table 14-2 Enzymes Dependent upon Thiamin
Diphosphate as a Coenzyme
Table 14-3 Some Enzymes That Require Pyridoxal
Phosphate as a Coenzyme
Table 14-4 Some Pyruvoyl Enzymes
C
H3C
O
O
O
NH3
H
C
H3N
O
O
H3N
CH3
C O
O
+

S
N
N
O
+
+

H
H
Biotin
Alanine
CO2
Pimeloyl-CoA
8-Amino-
7-oxo-
nonanoate
a
b
c
d
The vitamin biotin is formed in nature (left) by condensation of L-alanine with pimeloyl-CoA to form
8-amino-7-oxononanoate (AON). This compound is seen at the upper left of the center structure
joined as a Schiff base with the coenzyme pyridoxal phosphate (PLP). This is a product complex of
the enzyme AON synthase (see Webster et al., Biochemistry 39, 516-528, 2000) Courtesy of D. Alexeev,
R. L. Baxter, and L. Sawyer. Biotin synthesis requires three other enzymes (steps b, c, d). Step b is
catalyzed by a PLP-dependent transaminase. At the left is thiamin diphosphate, in the form of its
2-(1-hydroxyethyl) derivative, an intermediate in the enzyme pyruvate decarboxylase (Dobritzsch
et al., J. Biol. Chem. 273, 20196-20204, 1998). Courtesy of Guoguang Lu. Thiamin diphosphate functions
in all living organisms to cleave CC bonds adjacent to C=O groups.
719
Coenzymes: Natures Special Reagents
14
Most of the reactions discussed in Chapters 12 and
13 are catalyzed by enzymes that contain only those
functional groups found in the side chains of the
constituent amino acids. Coenzymes are nonprotein
molecules that function as essential parts of enzymes.
Coenzymes often serve as special reagents needed
for reactions that would be difficult or impossible
using only simple acidbase catalysis. In many instances,
they also serve as carriers, alternating catalysts that
accept and donate chemical groups, hydrogen atoms,
or electrons. Coenzymes will be considered here in
three groups:
1. Compounds of high group transfer potential such
as ATP and GTP that function in energy coupling
within cells. Because it is cleaved and then disso-
ciates from the enzyme to which it is bound, ATP
may be regarded as a substrate rather than a
coenzyme. However, as a phosphorylated form
of ADP it may also be viewed as a carrier of high-
energy phospho groups.
2. Compounds, often derivatives of vitamins that,
while in the active site of the enzyme, alter the
structure of a substrate in a way that permits it to
react more readily. Coenzyme A, pyridoxal phos-
phate, thiamin diphosphate, and vitamin B
12
coenzymes fall into this group.
3. Oxidative coenzymes with structures of precisely
determined oxidationreduction potential. Ex-
amples are NAD
+
, NADP
+
, FAD, and lipoic acid.
They serve as carriers of hydrogen atoms or of
electrons. Some of these coenzymes, such as
NAD
+
and NADP
+
, can usually dissociate rapidly
and reversibly from the enzymes with which they
function. Others, including FAD, are much more
tightly bound and rarely if ever dissociate from
the protein catalyst. Heme groups are covalently
linked to proteins such as cytochrome c and
cannot be dissociated without destroying the
enzyme. Very tightly bound coenzyme groups
are often called prosthetic groups, but there is
no sharp line that divides prosthetic groups from
the loosely bound coenzymes. For example, NAD
+
is bound weakly to some proteins but tightly to
others. Oxidative coenzymes are discussed in
Chapter 15.
A. ATP and the Nucleotide Handles
The role of ATP in driving biosynthetic reactions
has been considered in Chapter 12, where attention was
focused on the polyphosphate group which undergoes
cleavage. What about the adenosine end? Here is a
shapely structure borrowed from the nucleic acids.
What is it doing as a carrier of phospho groups? At
least part of the answer seems to be that the adenosine
monophosphate (AMP) portion of the molecule is a
handle which can be grasped by catalytic proteins.
For some enzymes, such as acetyl-CoA synthetase
(Eq. 12-45), the handle is important because the inter-
mediate acyl adenylate must remain bound to the
protein. Without the large adenosine group, there
would be little for the protein to hold onto.
C
H3C
O
O
O
NH3
H
C
H3N
O
O
H3N
CH3
C O
O
+

S
N
N
O
+
+

H
H
Biotin
Alanine
CO
2
Pimeloyl-CoA
8-Amino-
7-oxo-
nonanoate
a
b
c
d
720 Chapter 14. Coenzymes: Natures Special Reagents
AMP is only one of several handles to which
nature attaches phospho groups to form di- and tri-
phosphate derivatives. Like AMP, the other handles
are nucleotides, the monomer units of nucleic acids.
Thus, one enzyme requiring a polyphosphate as an
energy source selects ATP, another CTP, or GTP. The
nucleotide handles not only carry polyphosphate
groups but also are present in other coenzymes, such
as CoA, NAD
+
, NADP
+
, and FAD. In addition, they
serve as carriers for small organic molecules. For
example, uridine diphosphate glucose (UDP-Glc)
Chapter 10, is a carrier of active glucosyl groups im-
portant in sugar metabolism and cytidine diphos-
phate choline is an intermediate in synthesis of
phospholipids.
Recalling that acetyl adenylate (acetyl-AMP) is
an intermediate in synthesis of acetyl-CoA, and com-
paring the biosynthesis of sugars, phospholipids, and
acetyl-CoA, we see that in each case the enzyme in-
volved requires a different nucleotide handle. The
handle may provide a means of recognition which can
help an enzyme to pick the right bit of raw material
out of the sea of molecules surrounding it. Figure 5-6
shows the shapes of the four purine and pyrimidine
bases forming the most common nucleotide handles.
The distinctive differences both in shape and in hydro-
gen bond patterns are obvious. In binding to proteins,
the hydrogen bond-forming groups in the purine and
pyrimidine bases sometimes interact with precisely
positioned groups in the protein. However, in some
enzymes the handle is not precisely bound. This
seems to be the case for adenine, which makes surpris-
ingly few hydrogen bonds to proteins.
1
Hydroxyl
groups of the ribose or deoxyribose ring also often
form hydrogen bonds and the negatively charged
oxygen atoms of the 5'-phosphate may interact with
positively charged protein side chains.
B. Coenzyme A and Phosphopantetheine
The existence of a special coenzyme required in
biological acetylation was recognized by Fritz Lipmann
in 1945.
23a
The joining of acetyl groups to other
N
N
N
N
NH
2
CH
2
O
P

O O
O P
O
O

O
O
OH OH
P

O
O

O
The AMP handle in ATP
molecules is a commonplace reaction within living
cells, one example being the formation of the neuro-
transmitter acetylcholine. In the laboratory acetyla-
tion is carried out with reactive compounds such as
acetic anhydride or acetyl chloride.
Lipmann wondered what nature used in their place.
His approach in seeking the biological active acetate
is one that has been used successfully in solving many
biochemical problems. He first set up a test system to
examine the ability of extracts prepared from fresh
liver tissue to catalyze the acetylation of sulfanilamide
(Eq. 14-2). A specific color test was available for quan-
titative determination of very small amounts of the
product. The rate of acetylation of sulfanilamide under
standard conditions was taken as a measure of the
activity of the biochemical acetylation system. Lipmann
soon discovered that the reaction required ATP and
that the ATP was cleaved to ADP concurrently with
the formation of acetyl-sulfanilamide. He also found
that dialysis or ultrafiltration rendered the liver extract
almost inactive in acetylation. Apparently some essen-
tial material passed out through the semipermeable
dialysis membrane. When the dialysate or ultrafiltrate
was concentrated and added back, acetylation activity
was restored. The unknown material was not destroyed
by boiling, and Lipmann postulated that it was a new
coenzyme which he called coenzyme A (CoA). Now
the test system was used to estimate the amount of the
(14-1)
NH
2
S
NH
2
O O
Sulfanilamide
HN
S
NH
2
O O
C
O
CH
3
acetylation
(14-2)
H
3
C C
O
O
Acetylating
agent
+
HO
CH
2
CH
2
N
CH
3
CH
3
CH
3
+
CH
2
CH
2
N
CH
3
CH
3
CH
3
Acetylcholine
721
BOX 14-A DISCOVERY OF THE VITAMINS
Several mysterious and often fatal diseases
which resulted from vitamin deficiencies were
prevalent until the past century. Sailors on long sea
voyages were often the victims. In the Orient, the
disease beriberi was rampant and millions died of
its strange paralysis called polyneuritis. In 1840,
George Budd predicted that the disease was caused
by the lack of some organic compound that would
be discovered in a not too distant future.
a
In 1893,
C. Eijkman, a Dutch physician working in Indonesia,
observed paralysis in chicks fed white rice consumed
by the local populace. He found that the paralysis
could be relieved promptly by feeding an extract of
rice polishings. This was one of the pieces of evidence
that led the Polish biochemist Casimir Funk to for-
mulate the vitamine theory in about 1912. Funk
suggested that the diseases beriberi, pellagra, rickets,
and scurvy resulted from lack in the diet of four
different vital nutrients. He imagined them all to be
amines, hence the name vitamine.
In the same year in England, F. G. Hopkins
announced that he had fed rats on purified diets
and discovered that amazingly small amounts of
accessory growth factors, which could be obtained
from milk, were necessary for normal growth.
b
By
1915, E. V. McCollum and M. Davis at the University
of Wisconsin had recognized that rat growth depended
on not one but at least two accessory factors. The
first, soluble in fatty solvents, they called A, and the
other, soluble in water, they designated B. Factor B
cured beriberi in chicks. Later, when it was shown
that vitamine A was not an amine, the e was
dropped and vitamin became a general term.
Progress in isolation of the vitamins was slow,
principally because of a lack of interest. According
to R. R. Williams, when he started his work on isola-
tion of the antiberiberi factor in 1910 most people
were convinced that his efforts were doomed to
failure, so ingrained was Pasteurs idea that diseases
were caused only by bacteria. In 1926, Jansen isolated
a small amount of thiamin, but it was not until 1933
that Williams, working almost without financial
support, succeeded in preparing a large amount
of the crystalline compound from rice polishings.
Characterization and synthesis followed rapidly.
c,d
It was soon apparent that the new vitamin alone
would not satisfy the dietary need of rats for the B
factor. A second thermostable factor (B
2
) was required
in addition to thiamin (B
1
), which was labile and
easily destroyed by heating. When it became clear
that factor B
2
contained more than one component,
it was called vitamin B complex. There was some
confusion until relatively specific animal tests for
each one of the members had been devised.
Riboflavin was found to be most responsible for the
stimulation of rat growth, while vitamin B
6
was
needed to prevent a facial dermatitis or rat pellagra.
Pantothenic acid was especially effective in curing
a chick dermatitis, while nicotinamide was required
to cure human pellagra. Biotin was required for
growth of yeast.
The antiscurvy (antiscorbutic) activity was
called vitamin C, and when its structure became
known it was called ascorbic acid. The fat-soluble
factor preventing rickets was designated vitamin D.
By 1922, it was recognized that another fat-soluble
factor, vitamin E, is essential for full-term pregnancy
in the rat. In the early 1930s vitamin K and the
essential fatty acids were added to the list of fat-
soluble vitamins. Study of the human blood disor-
ders tropical macrocytic anemia and pernicious
anemia led to recognition of two more water-soluble
vitamins, folic acid and vitamin B
12
. The latter is
required in minute amounts and was not isolated
until 1948. Have all the vitamins been discovered?
Rats can be reared on an almost completely synthetic
diet. However, there is the possibility that for good
health humans require some as yet undiscovered
compounds in our diet. Furthermore, it is quite
likely that we receive some essential nutrients that
we cannot synthesize from bacteria in our intestinal
tracts. An example may be the pyrroloquinoline
quinone (PQQ).
e
Why do we need vitamins? Early clues came
in 1935 when nicotinamide was found in NAD
+
by
H. von Euler and associates and in NADP
+
by War-
burg and Christian. Two years later, K. Lohman and
P. Schuster isolated pure cocarboxylase, a dialyz-
able material required for decarboxylation of pyru-
vate by an enzyme from yeast. It was shown to be
thiamin diphosphate (Fig. 15-3). Most of the water-
soluble vitamins are converted into coenzymes or
are covalently bound into active sites of enzymes.
Some lipid-soluble vitamins have similar functions
but others, such as vitamin D and some metabolites
of vitamin A, act more like hormones, binding to
receptors that control gene expression or other
aspects of metabolism.
a
Hughes, R. E. (1973) Medical History 17, 127134
b
Harris, L. D. (1937) Vitamins in Theory and Practice, Cambridge
Univ. Press, London and New York
c
Karlson, P. (1984) Trends Biochem. Sci. 9, 536537
d
Williams, R. R., and Spies, T. D. (1938) Vitamin B
1
and Its Use in
Medicine, Macmillan, New York
e
Killgore, J., Smidt, C., Duich, L., Romero-Chapman, N., Tinker,
D., Reiser, K., Melko, M., Hyde, D., and Rucker, R. B. (1989)
Science 245, 850852
B. Coenzyme A and Phosphopantetheine
722 Chapter 14. Coenzymes: Natures Special Reagents
coenzyme in a given volume of dialysate or in any
other sample. When small amounts of CoA were
supplied to the test system, only partial restoration of
the acetylation activity was observed and the amount
of restoration was proportional to the amount of CoA.
With test system in hand to monitor various fraction-
ation methods, Lipmann soon isolated the new coen-
zyme in pure form from yeast and liver.
Coenzyme A (Fig. 14-1) is a surprisingly complex
molecule. The handle is AMP with an extra phospho
group on its 3'-hydroxyl. The phosphate of the 5'-carbon
is linked in anhydride (pyrophosphate) linkage to
another phosphoric acid, which is in turn esterified
with pantoic acid. Pantoic acid is linked to -alanine
and the latter to -mercaptoethylamine through
amide linkages, the reactive SH group being attached
to a long (1.9-nm) semi-flexible chain. Coenzyme A
can be cleaved by hydrolysis to pantetheine (Fig. 14-1),
pantetheine 4'-phosphate, and pantothenic acid.
These three compounds are all growth factors.
Pantothenic acid is a vitamin, which is essential to
human life. Its name is derived from a Greek root that
reflects its universal occurrence in living things. The
bacteriumLactobacillus bulgaricus, which converts milk
to yogurt, needs the more complex pantetheine for
growth. It finds a ready supply of pantetheine in milk
and has lost its ability to synthesize this compound.
However, it can convert pantetheine to CoA. Pantetheine
4'-phosphate is required for growth of Acetobacter
suboxydans. While it is not a dietary essential for most
organisms, it is found in a covalently bound form in
several enzymes.
While CoA was discovered as the acetylation
coenzyme, it has a far more general function. It is
required, in the form of acetyl-CoA, to catalyze the
synthesis of citrate in the citric acid cycle. It is essential
to the oxidation of fatty acids and carries propionyl
and other acyl groups in a great variety of other meta-
bolic reactions. About 4% of all known enzymes require
CoA or one of its esters as a substrate.
4
Coenzyme A has two distinctly different biochemical
functions, which have already been considered briefly
in Chapters 12 and 13 and can be summarized as
follows:
These functions depend, to a considerable extent, on
the fact that the properties of the carbonyl group of a
thioester are closely similar to those of an isolated
carbonyl group in a ketone.
5
Synthesis of fatty acids in bacteria requires a small
acyl carrier protein (ACP) whose functions are
similar to those of CoA. However, it contains pante-
2. Activation of a hydrogen
atom adjacent to the
carbonyl of a thioester
R C
O
1. Activation of
acyl group, ,
toward transfer by
nucleophilic displacment
R C
O
S CoA
Y

C C
O
S CoA
H
(Leaves as H
+
)
HO CH
2
C
C C
O
N H
2
C
CH
2
COOH
OH
CH
3
CH
3
H
H
Pantothenic acid
Pantoic acid
-Alanine
Figure 14-1 Coenzyme A, an acyl-activating coenzyme
containing the vitamin pantothenic acid.
-Mercapto-
ethylamine
-Alanine
d-Pantoic
acid
The
vitamin
pantothenic
acid
1.9 nm
Reactive SH group at
end of long flexible chain
Pantetheine
4'-phosphate,
bound
prosthetic
group of
acyl carrier
protein
(ACP)
HS
AMP
with extra
phospho
group on
3'-OH
NH
NH
O
P
O
O
O
H
OH
H
3
C
H
3
C
4'
P
O
O

O
5'
O
N
N
N
N
NH
3
OH O
P

O
OH
O
3'
O
O

723
theine 4'-phosphate covalently bonded through phos-
phodiester linkage to a serine side chain. In E. coli this
is at position 36 in the 77-residue protein.
6,7
Here the
nucleotide handle of CoA has been replaced with a
larger and more complex protein which can interact in
specific ways with the multiprotein fatty acid synthase
complex described in Chapter 21. In higher organisms
ACP is usually not a separate protein but a domain in
a large synthase molecule. Bound phosphopantetheine
is also found in enzymes involved in synthesis of pep-
tide antibiotics (Chapter 29)
8,9
and polyketides (Chap-
ter 21).
10,11
It is also present in subunits of cytochrome
oxidase and of ATP synthase of Neurospora but appears
to play only a structural role, being needed for proper
assembly of these multimeric proteins.
12
In a citrate-
cleaving enzyme, and in a bacterial malonate decar-
boxylase, phosphopantethine is attached to a serine
side chain as 2-(5-phosphoribosyl)-3-dephospho-
CoA.
1314a
Attachment of phosphopantetheine to proteins is
catalyzed by a phosphotransferase that utilizes CoA as
the donor. A phosphodiesterase removes the phospho-
pantetheine, providing a turnover cycle.
1515b
A variety
of synthetic analogs have been made.
4,16
The reactive
center of CoA and phosphopantetheine is the SH group,
which is carried on a flexible arm that consists in part
of the -alanine portion of pantothenic acid. A mystery
is why pantoic acid, a small odd-shaped molecule that
the human body cannot make, is so essential for life.
The hydroxyl group is a potential reactive site and the
two methyl groups may enter into formation of a tri-
alkyl lock (p. 485), part of a sophisticated elbow or
shoulder for the SH-bearing arm.
When it binds to citrate synthase acetyl-CoA appears
to bind only after the enzyme has undergone a confor-
mational change that closes the enzyme around its
other substrate oxaloacetate (Fig. 13-8). The adenine
ring of the long CoA handle is tightly bonded to the
protein through hydrophobic interactions and hydrogen-
bond recognition interactions. Additional specific
hydrophobic interactions allow the enzyme to hold the
acetyl group in a precise position where it can be acted
upon by the catalytic groups. In the case of CoA trans-
ferase (Eq. 12-50) smaller thiols can replace CoA but
with much lower catalytic rates. The acyl-CoA deriva-
tives are weakly bound, the expected intrinsic binding
energy of the pantetheine portion of the molecule
apparently being used to increase k
cat
.
17
From a study
of kinetics and equilibria it was concluded that the
binding energy of the interaction of the nucleotide
portion of coenzyme A with the enzyme is utilized
to increase the rate of formation and to stabilize the
covalently linked E-CoA (see Eq. 12-50). On the other
hand, binding of the pantoic acid part of the molecule
decreases the stability of the transition state for break-
up of the complex by ~ 40 kJ/mol, which corresponds
to a 10
7
-fold increase in the reaction rate.
16
C. Biotin and the Formation of Carboxyl
Groups from Bicarbonate
By 1901 it was recognized that yeast required for
its growth an unknown material which was called
bios. This was eventually found to be a mixture of
pantothenic acid, inositol, and a third component
which was named biotin. Biotin was also recognized
as a factor promoting growth and respiration of the
clover root nodule organism Rhizobium trifolii and as
vitamin H, a material that prevented dermatitis and
paralysis in rats that were fed large amounts of un-
cooked egg white. Isolation of the pure vitamin was
a heroic task accomplished by Kgl in 1935. In one
preparation 250 kg of dried egg yolk yielded only
1.1 mg of crystalline biotin.
18
Biotin contains three chiral centers and therefore
has eight stereoisomers.
18,19
Of these, only one, the
dextrorotatory (+)-biotin, is biologically active.
19,20
The vitamin is readily oxidized to the sulfoxide and sul-
fone. The sulfoxide can be reduced back to biotin by a
molybdenum-containing reductase in some bacteria
(see also Chapter 16, Section H).
20a
Biotin is synthesized
from pimeloyl-CoA (see chapter banner, p. 719 and
Eq. 24-39). Four enzymes are required. Two of them, a
C. Biotin and the Formation of Carboxyl Groups from Bicarbonate
S
N
N
O
H
HOOC
1
2 3
4 5
1
2
3
H
H
(+)-Biotin
6
7
8
9
10
P
O
O

O
O
Adenine
O HO
O
Phosphopantetheine
O
CH
2
O
P
O CH
2
Ser
O O

4'-
3'

OH HO
3'
5''
2'
2'- (5''-Phophoribosyl) -3'-dephospho-CoA
724 Chapter 14. Coenzymes: Natures Special Reagents
synthase that catalyzes step a (banner) and a transami-
nase that catalyzes step b, contain the coenzyme pyri-
doxal phosphate (PLP).
20b
The final step is insertion of
a sulfur atom from an iron-sulfur center.
1. Biotin-Containing Enzymes
Within cells the biotin is covalently bonded to
proteins, its double ring being attached to a 1.6-nm
flexible arm. The first clue to this fact was obtained
from isolation of a biotin-containing material biocytin,
-N-biotinyl-L-lysine, from autolysates (self-digests) of
rapidly growing yeast.
21
It was subsequently shown
that the lysine residue of the biocytin was originally
present in proteins at the active sites of biotin-containing
enzymes usually within the sequence AMKM
22
or
VMKM.
23
Other conserved features also mark the
attachment site.
24
Biotin acts as a carboxyl group carrier in a series
of carboxylation reactions, a function originally sug-
gested by the fact that aspartate partially replaces
biotin in promoting the growth of the yeast Torula
cremonis. Aspartate was known to arise by transami-
nation from oxaloacetate, which in turn could be
formed by carboxylation of pyruvate. Subsequent
studies showed that biotin was needed for an enzy-
matic ATP-dependent reaction of pyruvate with bicar-
bonate ion to form oxaloacetate (Eq. 14-3). This is a
carboxylation coupled to the hydrolysis of ATP.
In addition to pyruvate carboxylase, other biotin-
requiring enzymes act on acetyl-CoA, propionyl-CoA,
and -methylcrotonyl-CoA, using HCO
3

to add
carboxyl groups at the sites indicated by the arrows in
the accompanying structures. Because of the presence
of the C = C double bond conjugated with the carbonyl
group, the carboxylation of -methylcrotonyl-CoA is
electronically analogous to carboxylation.
Human cells, as well as those of other higher eukary-
otes, carry genes for all four of these enzymes.
25 29
Acetyl-CoA carboxylase is a cytosolic enzyme needed
for synthesis of fatty acids but the other three enzymes
enter mitochondria when they function. The biotin-
dependent carboxylases, which are listed in Table 14-1,
have a variety of molecular sizes and subunits but show
much evolutionary conservation in their sequences
and chemical mechanisms.
22,25
In higher eukaryotes
all of the catalytic apparatus of the enzymes is present
in single large 190 - to 200-kDa subunits. The 251-kDa
subunit of yeast acetyl-CoA carboxylase consists of
2337 amino acid residues.
30
That of rats contains 2345
31
and that of the alga Cyclotella 2089.
32
Human cytosolic
acetyl-Co carboxylase has 2347 residues while a mito-
chondria-associated form has an extra 136 residues,
most of them in a hydrophobic N-terminal extension.
32a,b
Plants have two forms of the enzyme, one cytosolic
and one located in plastids. In wheat, they have 2260
and 2311 residues, respectively.
32c,32d
Animal and
fungal pyruvate carboxylases are also large ~ 500-kDa
tetramers.
32e
The yeast enzyme consists of 1178-residue
monomers.
33
In contrast, the 560-kDa human propionyl-
CoA carboxylase is an
4

4
tetramer.
34,35
In bacteria and in at least some plant chloroplasts,
36,37
acetyl-CoA carboxylase consists of three different
kinds of subunit and four different peptide chains.
The much studied E. coli enzyme is composed of a
156-residue biotin carboxyl carrier protein,
38
a 449-
residue biotin carboxylase, whose three-dimensional
structure in known,
39,39a
and a carboxyltransferase
subunit consisting of 304 ()- and 319 ()- residue
chains. These all associate as a dimer of the three
subunits (eight peptide chains).
40 42
Biotin becomes attached to the proper -amino
groups at the active centers of biotin enzymes by the
action of biotin holoenzyme synthetase (biotinyl
NH HN
S
C
N NH
O
C
HN
O
C O
O
H
Biocytin
Biotin
Peptide
chain
~
1
.6
n
m
(14-3)
H
3
C C
O
S CoA H
2
C C
O
S CoA
C C
O
S CoA
CH
3
Propionyl-CoA
CH H
3
C
Acetyl-CoA
CH
3
-Methylcrotonyl-CoA
CH
3
C COO

O
CH
2
C COO

OOC
HCO
3

Pyruvate carboxylase
ATP
ADP + P
i
Oxaloacetate
725
protein ligase), which utilizes ATP to form an interme-
diate biotinyl-AMP.
43 47a
Hereditary deficiency of this
enzyme has been observed in a few children and has
been treated by administration of extra biotin.
48
The E.
coli biotin holoenzyme synthetase, whose three-dimen-
sional structure is known, has a dual function. It is also
a repressor of transcription of the biotin biosynthetic
operon.
45
Intracellular degradation of biotin-contain-
ing proteins yields biotin-containing oligopeptides as
well as biocytin. These are acted on by biotinidase
to release free biotin.
49,50
The action of this enzyme in
recycling biotin may be a controlling factor in the rate
of formation of new biotin-dependent enzymes.
2. The Mechanism of Biotin Action
It may seem surprising that a coenzyme is needed
for these carboxylation reactions. However, unless
the cleavage of ATP were coupled to the reactions, the
equilibria would lie far in the direction of decarboxyla-
tion. For example, the measured apparent equilibrium
constant K for conversion of propionyl-CoA to S methyl-
malonyl-CoA at pH 8.1 and 28C
51
is given by Eq. 14-4.
The function of biotin is to mediate the coupling of
ATP cleavage to the carboxylation, making the overall
reaction exergonic. This is accomplished by a two-stage
process in which a carboxybiotin intermediate is
formed (Eq. 14-5). There is one known biotin-containing
enzyme that does not utilize ATP. Propionic acid bacteria
contain a carboxyltransferase which transfers a carb-
oxyl group reversibly from methylmalonyl-CoA to
pyruvate to form oxaloacetate and propionyl-CoA
(see Fig. 17-10). This huge enzyme consists of a central
hexameric core of large 12S subunits to which six 5S
dimeric subunits and twelve 123-residue biotinylated
peptides are attached.
25,52
No ATP is needed because
free HCO
3

is not a substrate. However, biotin serves


as the carboxyl group carrier in this enzyme too.
Carboxybiotin. The structure of biotin suggested
that bicarbonate might be incorporated reversibly into
its position 2'. However, this proved not to be true and
it remained for F. Lynen and associates to obtain a clue
from a model reaction. They showed that purified
-methylcrotonyl-CoA carboxylase promoted the
carboxylation of free biotin with bicarbonate (H
14
CO
3

)
and ATP. While the carboxylated biotin was labile,
treatment with diazomethane (Eq. 14-6) gave a stable
dimethyl ester of N-1'-carboxybiotin.
53,54
The co-
valently bound biotin at active sites of enzymes was
also successfully labeled with
14
CO
2.
Treatment of
the labeled enzymes with diazomethane followed by
hydrolysis with trypsin and pepsin gave authentic
N-1'-carboxybiocytin. It was now clear that the cleav-
age of ATP is required to couple the CO
2
from HCO
3

to the biotin to form carboxybiotin. The enzyme must


C. Biotin and the Formation of Carboxyl Groups from Bicarbonate
=

K
G
[ ][ ][ ]
[ ][ ][ ]
.
ADP P methylmalonyl CoA
ATP HCO propionyl CoA
= 5.7
kJ mol
i
3
4 36
1
(14-4)
(14-5)
TABLE 14-1
Enzymes Containing Bound Biotin
1. Catalyzing beta carboxylation using HCO
3

with coupled
cleavage of ATP to ADP + P
i
Acetyl-CoA carboxylase
Propionyl-CoA carboxylase
Pyruvate carboxylase
-Methylcrotonyl-CoA carboxylase
( carboxylation)
2. Carboxyl group transfer without cleavage of ATP
Carboxyltransferase of Propionobacterium
3. Biotin-dependent Na
+
pumps
Oxaloacetate decarboxylase
Methylmalonyl-CoA decarboxylase
Glutaconyl-CoA decarboxylase
4. Other
Malonate decarboxylase
Urea carboxylase
C
O
C H
C
O
C COO

b
ATP
ADP + P
i
HCO
3

a
Biotin (covalently bound)
Carboxybiotin
C
O
C C
CH
3
H
H
C
O
C C
CH
3
H
COO

or
b
726 Chapter 14. Coenzymes: Natures Special Reagents
then transfer the carboxyl group from carboxybiotin
to the substrate that is to be carboxylated. Enzymatic
transfer of a carboxyl group from chemically synthe-
sized carboxybiotin onto specific substrates confirmed
the proposed mechanism.
55
The biotin carboxyl carrier subunit of E. coli
acetyl-CoA carboxylase contains the covalently bound
biotin.
55a,b
The larger biotin carboxylase subunit cata-
lyzes the ATP-dependent attachment of CO
2
to the
biotin and the carboxyltransferase subunit catalyzes
the final transcarboxylation step (Eq. 14-5, step b) by
which acetyl-CoA is converted into malonyl-CoA.
The biotin, which is attached to the carrier protein, is
presumably able to move by means of its flexible arm
from a site on the carboxylase to a site on the transcar-
boxylase.
Carboxyphosphate. During the initial carboxyla-
tion step
18
O from labeled bicarbonate enters the P
i
that is split from ATP. This suggested transient forma-
tion of carboxyphosphate by nucleophilic attack of
HCO
3

on ATP (Eq. 14-7). The carboxyl group of this


reactive mixed anhydride
56
could then be transferred
to biotin. This mechanism is supported by the fact
that biotin carboxylase catalyzes the transfer of a
(14-6)
phospho group to ADP from carbamoyl phosphate,
an analog of carboxyphosphate in a reaction that is
analogous to the reverse of that in Eq. 14-7 and also
by a slow bicarbonate-dependent ATPase activity that
does not depend upon biotin.
57,58,58a
The simplest mechanism for transfer of the carboxyl
group of carboxyphosphate to biotin would appear to
be nucleophilic displacement of the phosphate leaving
group by N1' of biotin. The enzyme could presumably
first catalyze removal of the N1' hydrogen to form a
ureido anion.
59
Another reasonable possibility would
be for the terminal phospho group of ATP to be trans-
ferred to biotin to form an O-phosphate
60,61
(Eq. 14-8, step a) which could react with bicarbonate as
in Eq. 14-8, step b. Cleavage of the enol phosphate by
attack of HCO
3

would simultaneously create a nucleo-


philic center at N1' and carboxyphosphate ready to
react with N1'. However, this could not easily explain
the ATPase activity in the absence of biotin.
(14-7)
NH N
O
1'
Ureido anion

(14-8)
S
NH HN
O
C
O
OH
S
NH N
O
C
O
OCH
3
14
C
H
3
CO
O
1
3
ATP
ADP + P
i
H
14
CO
3

CH
3
N
2
NH N
O
14
C
O

O
N-1-Carboxybiotin
H O C
O
O

HO C
O
O
ATP
PO
3

ADP
Carboxyphosphate
b
a
NH N
O
C
ATP ADP
NH N
O
Bicarbonate
C
O
HO
Carboxybiotin
P
i
NH HN
O
O
O

HO
P
O
O

HO
727
Either of the foregoing mechanisms requires that
the ureido anion of biotin attack the rather unreactive
carbon atom of carboxyphosphate. Another alternative,
which is analogous to that suggested for PEP carboxy-
lase (Eq. 13-53) is for carboxyphosphate to eliminate
inorganic phosphate to give the more electrophilic CO
2
(Eq. 14-9, step a). The very basic inorganic phosphate
trianion PO
4
3
that is eliminated could remove the
proton from N1' of biotin to create the biotin ureido
anion (step b), which could then add to CO
2
(step c).
22
Have we checked all of the possibilities for the
mechanism of biotin carboxylation? Kruger and asso-
ciates
62,63
suggested that biotin, as a ureido anion,
might add to bicarbonate to form a highly unstable
intermediate which, however, could be phosphorylated
by ATP (Eq. 14-10, steps a and b). This intermediate
could undergo elimination of inorganic phosphate
C. Biotin and the Formation of Carboxyl Groups from Bicarbonate
(14-9)
(step c), driving the reaction to completion. The
observed transfer of isotope from
18
O-containing
bicarbonate into ADP would be observed.
The carboxylation step. Once formed, the
carboxybiotin head group could swing to the carb-
oxyltransferase site where transfer of the carboxyl
group into the final product takes place. This might
occur either by nucleophilic attack of an enolate anion
on the carbonyl carbon (Eq. 14-11) or on CO
2
generated
by reversal of the reactions of Eq. 14-9, steps b and c.
When pyruvate with a chiral methyl group is
carboxylated by pyruvate carboxylase the configuration
at C-3 is retained. The carboxyl enters from the 2-si
side, the same side from which the proton (marked H*)
was removed to form the enolate anion (Eq. 14-12).
Comparable stereochemistry has been established for
other biotin-dependent enzymes.
64,65
These enzymes do not catalyze any proton exchange
at C-3 of pyruvate or at C-2 of an acyl-CoA unless the
biotin is first carboxylated. This suggested that removal
of the proton to the biotin oxygen and carboxylation
might be synchronous. However,
13
C and
2
H kinetic
isotope effects and studies of
3
H exchange
66
support
the existence of a discrete enolate anion intermediate
as shown in Eq. 14-11.
65,67
This mechanism is also
consistent with the observation that propionyl-CoA
(14-11)
C C
COO

O
H
H
H C C
COO

H
H
Pyruvate 2-Si face is facing viewer
2
C C
COO

OOC
H
H
*
(14-12)
NH N
O

NH N
O
C

O
HO
HO
P OH
O

O
O
ADP
C
HO
O

O
NH N
O

C O
O
HO
2
O
3
P
NH N
O
Biotin ureido anion
C
O
HO
ADP
P
i
ATP
H
+
H
Carboxybiotin
b
a
c
(14-10)
Biotin
NH N
O
C
O
HO
CH
2
S
O

CoA
CH
2
S
O
CoA COOH
Enolate anion
from acetyl-CoA
M
2+
or H
+
Malonyl-CoA
NH N
O
Biotin
NH N
O
C
O
O O
P
O
O

H
O

P
O
O

Carboxybiotin
H
+
C
O
O
Carboxyl-phosphate
b
a
c
728 Chapter 14. Coenzymes: Natures Special Reagents
BOX 14-B THE BIOTIN-BINDING PROTEINS AVIDIN AND STREPTAVIDIN
A biochemical curiosity is the presence in egg
white of the glycoprotein avidin.
a,b
Each 68-kDa
subunit of this tetrameric protein binds one molecule
of biotin tenaciously with K
f
~ 10
15
M
1
. Natures
purpose in placing this unusual protein in egg white
is uncertain. Perhaps it is a storage form of biotin,
but it is more likely an antibiotic that depletes the
environment of biotin. A closely similar protein
streptavidin is secreted into the culture medium by
Streptomyces avidinii.
c
Its sequence is homologous to
that of avidin. It has a similar binding constant for
biotin and the two proteins have similar three-dimen-
sional structures.
a,d j
Biotin binds at one end of a
barrel formed from antiparallel strands and is held
by multiple hydrogen bonds and a conformational
alteration that allows a peptide loop to close over
the bound vitamin.
Historically, avidin was important to the discov-
ery of biotin. The bonding between avidin and
biotin is so tight that inclusion of raw egg white in
the diet of animals is sufficient to cause a severe biotin
deficiency. Avidin has also been an important tool
to enzymologists interested in biotin-containing
enzymes. Avidin invariably inhibits these enzymes
and inhibition by avidin is diagnostic of a biotin-
containing protein. Recently, avidin and streptavidin
have found widespread application in affinity
chromatography, in immunoassays, and in the
staining of cells and tissues.
d,k p
These uses are all
based on the ability to attach biotin covalently to
side chain groups of proteins, polysaccharides, and
other substances. The carboxyl group of the biotin
arm, which lies at the surface of the complex with
avidin or streptovidin, can be converted to any of a
series of reactive derivatives. For example, p-nitro-
phenyl or N-hydroxysuccinimide esters of biotin can
be used to attach biotin to amino groups of proteins
to form biotinylated proteins.
Other reactive derivatives can be used to attach
biotin to phenolic, thiol, or carbonyl groups.
The affinity of avidin or streptavidin for the
resulting biotinylated materials is still very high.
This fact has permitted the application of this biotin
avidin technology to numerous aspects of research
and diagnostic medicine. For example, a specific
antibody (IgG) can be utilized for immunoassay of a
hormone or other ligand. A second antibody, specific
for the IgGligand complex, can be produced in a
biotinylated form. A steptavidin complex of a biotin-
ylated enzyme such as alkaline phosphatase, -galac-
tosidase, or horseradish peroxidase, for which a sensitive
colorimetric assay is available, is allowed to react
with the biotinanti-IgGIgGligand complex. The
streptavidin now releases enzyme in proportion to
the amount of ligand originally present. Since the
biotinylated anti IgG and streptavidinbiotinylated
enzyme can be stored as a stable mixture, the assay
is simple and fast.
Avidin technology can also be applied to the
isolation of proteins and other materials from cells.
Because the irreversibility of the binding of biotin
may be a problem, photocleavable biotin derivatives
have been developed.
q
In the following structure,
the biotin derivative has been joined to a protein
(as in the first equation in this box) and is ready for
separation, perhaps on a column containing immo-
bilized avidin or streptavidin. After separation the
biotin together with the linker and photocleavable
group are cut off by a short irradiation with ultra-
Ligand
IgGL
IgG (specific)
Anti IgG*
Biotin
*mouse
*rabbit
*goat
BiotinAnti IgGIgGL
Streptavidinbiotinylated enzyme*
Color
Streptavidinbiotinanti-IgGIgGL
+
Free biotinylated enzyme (active)
Stable
mixture
e.g.
*Alkaline phophatase
*-Galactosidase
*Horseradish peroxidase
O
O
O
O
N O
C
O
Biotin
N HO
H
2
N Protein
Biotin C
O
N Protein
H
Biotinylated protein
729
BOX 14-B (continued)
violet light, leaving the protein in a free form.
a
Livnah, O., Bayer, E. A., Wilshek, M., and Sussman, J. L. (1993)
Proc. Natl. Acad. Sci. U.S.A. 90, 50765080
b
Pugliese, L., Coda, A., Malcovati, M., and Bolognesi, M. (1993)
J. Mol. Biol. 231, 698710
c
Meslar, H. W., Camper, S. A., and White, H. B., III. (1978) J. Biol.
Chem. 253, 69796982
Spacer
Photocleavable group
h
Biotin
C O
N H
C O
N H
CH
2
CH
NO
2
O
H
3
C C
O
N Protein
H
d
Wilchek, M., and Bayer, E. A. (1989) Trends Biochem. Sci. 14,
408412
e
Punekar, N. S., and Lardy, H. A. (1987) J. Biol. Chem. 262, 6714
6719
f
Henderson, W. A., Phler, A., Smith, J. L., Satow, Y., Merritt, E.
A., and Phizackerley, R. P. (1989) Proc. Natl. Acad. Sci. U.S.A.
86, 21902194
g
Weber, P. C., Pantoliano, M. W., and Thompson, L. D. (1992)
Biochemistry 31, 93509354
h
Schmidt, T. G. M., Koepke, J., Frank, R., and Skerra, A. (1996)
J. Mol. Biol. 255, 753766
i
Weber, P. C., Ohlendorf, D. H., Wendoloski, J. J., and Salemme,
F. R. (1989) Science 243, 8588
j
Sano, T., and Cantor, C. R. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 31803184
k
Bayer, E. A., and Wilchek, M. (1980) Meth. Biochem. Anal. 26,
146
l
Childs, G. V., Naor, Z., Hazum, F., Tibolt, R., Westlund, K. N.,
and Hancock, M. B. (1983) J. Histochem. Cytochem. 31, 1422
1425
m
Wilchek, M., and Bayer, E. A., eds. (1990) Methods in Enzymol-
ogy, Vol. 184, Academic Press, San Diego, California
n
Savage, M. D., Mattson, G., Desai, S., Nielander, G. W.,
Morgensen, S., and Conklin, E. J. (1992) Avidin-Biotin Chemis-
try: A Handbook, Pierce, Rockford, Illinois
o
Donnelson, J. E., and Wu, R. (1972) J. Biol. Chem. 247, 4661
4668
p
Laundon, C. H., and Griffith, J. D. (1987) Biochemistry 26, 3759
3762
q
Olejnik, J., Sonar, S., Krzymaska-Olejnik, E., and Rothschild,
K. J. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 75907594
carboxylase and transcarboxylase both catalyze elimi-
nation of HF from -fluoropropionyl-CoA to form the
unsaturated acrylyl-CoA.
67
The elimination presum-
ably occurs via an enolate anion intermediate as in
Eq. 13-28.
A bound divalent metal ion, usually Mn
2+
, is
required in the transcarboxylation step. A possible
function is to assist in enolization of the carboxyl
acceptor. However, measurement of the effect of the
bound Mn
2+
on
13
C relaxation times in the substrate
for pyruvate carboxylase indicated a distance of ~0.7
nm between the carbonyl carbon and the Mn
2+
, too
great for direct coordination of the metal to the carbonyl
oxygen.
68
Another possibility is that the metal binds
to the carbonyl of biotin as indicated in Eq. 14-11.
Pyruvate carboxylase utilizes two divalent metal ions
and at least one monovalent cation.
68a
What is the role of the sulfur atom in biotin? Per-
haps it interacts with CO
2
, helping to hold it in a correct
orientation for reaction. Perhaps it helps to keep the
ureido ring of biotin planar, or perhaps it has no special
function.
69
3. Control Mechanisms
Most pyruvate carboxylases of animal and of yeast
are allosterically activated by acetyl-CoA, but those of
bacteria are usually not. The enzyme from chicken
liver has almost no activity in the absence of acetyl-CoA,
which appears to increase greatly the rate of formation
of carboxyphosphate and to slow the side reaction by
which carboxyphosphate is hydrolyzed to bicarbonate
and phosphate.
70
The acetyl-CoA carboxylases of rat
or chicken liver aggregate in the presence of citrate to
form ~ 8000-kDa rods. Citrate is an allosteric activator
for this enzyme but it acts only on a phosphorylated
form and the primary control mechanism.
71,72
This
enzyme in plants is a target for a group of herbicides
that are selectively toxic to grasses.
73
4. Pumping Ions with the Help of Biotin
Biotin-dependent decarboxylases act as sodium
ion pumps in Klebsiella
74
and in various anaerobes.
22,75
For example, oxaloacetate is converted to pyruvate and
bound carboxybiotin.
74,74a
The latter is decarboxylated
730 Chapter 14. Coenzymes: Natures Special Reagents
to CO
2
at the same time that two Na
+
ions are transported
from the inside to the outside of the cell. The function
of this pump, like that of the Na
+
, K
+
-ATPase (Fig. 8-25)
is to provide an electrochemical gradient that drives
the transport of other ions and molecules through the
membrane. Similar ion pumps are operated by decar-
boxylation of methylmalonyl-CoA
76
and glutaconyl-
CoA.
77
Yeast (Saccharomyces cerevisiae) cannot make
biotin and requires an unusually large amount of the
vitamin when urea, allantoin, allantoic acid, and certain
other compounds are supplied as the sole source of
nitrogen for growth. The reason is that in this organism
urea must first be carboxylated by the biotin-containing
urea carboxylase
78
(see Eq. 24-25) before it can be
hydrolyzed to NH
3
and CO
2
.
D. Thiamin Diphosphate
In Chapter 13 we considered the breaking of a
bond between two carbon atoms, one of which is also
bonded to a carbonyl group. These cleavages are
catalyzed by simple acidic and basic groups of the
protein side chains. On the other hand, the decarboxy-
lation of 2-oxo acids (Eq. 14-13) and the cleavage and
formation of -hydroxyketones (Eq. 14-14) depend
upon thiamin diphosphate (TDP).
79 85
These reactions
represent a second important method of making and
breaking carboncarbon bonds which we will designate
condensation and cleavage. The common
feature of all thiamin-catalyzed reactions is that the
bond broken (or formed) is immediately adjacent to the
carbonyl group, not one carbon removed, as in cleavage
reactions. No simple acidbase catalyzed mechanisms
can be written; hence the need for a coenzyme.
1. Chemical Properties of Thiamin
The weakly basic portion of thiamin or of its coen-
zyme forms is protonated at low pH, largely on N-1 of
the pyrimidine ring.
86 88
The pK
a
value is ~ 4.9. In basic
solution, thiamin reacts in two steps with an opening
of the thiazole ring (Eq. 14-15) to give the anion of a
thiol form which may be crystallized as the sodium
salt.
79,84
This reaction, like the competing reaction
described in Eq. 7-19, and which leads to a yellow
unstable form of the thiamin anion, is an example of a
cooperative two-proton dissociation with linked
structural changes. A very low concentration of the
intermediate pseudobase is present during the titra-
tion. This property, which is unusual among small
molecules, was instrumental in leading Williams et al.
to the correct structure for the vitamin.
89
A still unan-
swered question is, What biological significance is
associated with these reactions? Perhaps the thiol form
depicted in Eq. 14-15 or the yellow form (Eq. 7-19)
becomes attached to active sites of some proteins
through disulfide linkages.
Thiamin is unstable at high pH
90,91
and is destroyed
by the cooking of foods under mildly basic conditions.
The thiol form undergoes hydrolysis and oxidation
by air to a disulfide. The tricyclic form (Eq. 7-19) is
oxidized to thiochrome, a fluorescent compound
R C COO

O
R CH
O
+ H
+
+ CO
2
(14-13)
R C C
HO
R C
O
C R
O
R
O
H
H
H +
(14-14)
S
N
H
CH
2
CH
2
CH
2
O
P
O
P
O
CH
3
OH
O O

N
N H
3
C
H
2
N
+
H
+
Protonation occurs
here with pK
a
~ 4.9
1
2
3
4
5
1'
2'
3'
4'
5'
6'
This hydrogen
dissociates as
H
+
during
catalysis
Thiamin diphosphate
(14-15)
S
N
R
C
2
H
4
OH
S
N
R
C
2
H
4
OH
H
HO

S
C
N
R
C
2
H
4
OH
+
+ OH

H
O
Thiazolium form
Intermediate pseudobase
Thiol form
H
+
CH
3
CH
3
CH
3
731
whose formation from thiamin by treatment with alka-
line hexacyanoferrate (III) (ferricyanide; Eq. 14-16) is
the basis of a much used fluorimetric assay.
Treatment of thiamin with boiling 5N HCl deaminates
it to the hydroxy analogue oxythiamin, a potent antag-
onist. Pyrithiamin, another competitor containing
in place of the thiazolium ring, is very toxic especially
to the nervous system.
In a solution of sodium sulfite at pH 5, thiamin is
cleaved by what appears to be a nucleophilic displace-
ment reaction on the methylene group to give the free
thiazole and a sulfonic acid.
In fact the mechanism of the reaction is more complex
and is evidently initiated by addition of a nucleophile
Y, such as

OH or bisulfite, followed by elimination of


the thiazole (Eq. 14-18).
A similar cleavage is catalyzed by thiamin-degrad-
ing enzymes known as thiaminases which are found in
a number of bacteria, marine organisms, and plants.
In a bacterial thiaminase, group Y of Eq. 14-18 is a
cysteine SH.
92,92a
Thiamin is synthesized in bacteria, fungi, and
plants from 1-deoxyxylulose 5-phosphate (Eq. 25-21),
which is also an intermediate in the nonmevalonate
pathway of polyprenyl synthesis. However, thiamin
diphosphate is a coenzyme for synthesis of this inter-
mediate (p. 736), suggesting that an alternative path-
way must also exist. Each of the two rings of thiamin
is formed separately as the esters 4-amino-5-hydroxy-
methylpyrimidine diphosphate and 4-methyl-5-(-
hydroxyethyl) thiazole monophosphate. These
precursors are joined with displacement of pyrophos-
phate to form thiamin monophosphate.
92b
In eukary-
otes this is hydrolyzed to thiamin, then converted to
thiamin diphosphate by transfer of a diphospho group
from ATP.
92b,c
In bacteria thiamin monophosphate is
converted to the diphosphate by ATP and thiamin
monophosphate kinase.
92b
2. Catalytic Mechanisms
The first real clue to the mechanism of thiamin-
dependent cleavage came in about 1950 when Mizuhara
showed that at pH 8.4 thiamin catalyzes the nonenzy-
matic conversion of pyruvate into acetoin (Eq. 14-19).
93
Following Mizuharas lead, Breslow investigated the
same reaction using the then new NMR method.
94
He made the surprising discovery that the hydrogen
atom in the 2 position of the thiazolium ring, between
the sulfur and the nitrogen atoms, exchanged easily
with deuterium of
2
H
2
O. The pK
a
of this proton has
been estimated as ~ 18, low enough to permit rapid
N
N
N HN
S
CH
3
CH
2
CH
2
OH
N
N
N N
S
CH
3
CH
2
CH
2
OH
Tricyclic form of thiamin
Thiochrome
Fe(CN)
6
3
OH

(14-16)
(14-17)
N
N
NH
2
CH
2
S
N
C
2
H
4
OH
CH
3
Y
H
N
N
NH
Y
H
CH
2
H
+
+
H
H

OSO
2
H
Thiazole
Adduct
Products
(14-18)
D. Thiamine Diphosphate
N
N
NH
2
CH
2
SO
3

S N
H
3
C C
2
H
4
OH
HSO
3

H
+
Thiamin
N
CH
2
CH
2
OH
+
H
3
C
732 Chapter 14. Coenzymes: Natures Special Reagents
(14-20)
S
N
C C

O
O
CH
3
OH
CH
2
S
C
N
C
H
3
C
HO
CH
2
+
H
S
N
C
CH
2
+
CH
3
H
O H
-Lactylthiamin
diphosphate.
Intermediate
arising from
pyruvate
CH
3
C
H O
CO
2
Enamine
+
Thiazolium
dipolar ion
b
a
(14-21)
dissociation and replacement with
2
H.
84,95,96
The resulting thiazolium
dipolar ion (or ylid ) formed by this
dissociation (Eq. 14-20, step a) is stabi-
lized by the electrostatic interaction
of the adjacent positive and negative
charges. Breslow suggested that this
dipolar ion is the key intermediate in
reactions of thiamin-dependent en-
zymes.
94,97
The anionic center of the
dipolar ion can react with a substrate
such as an 2-oxo acid or 2-oxo alcohol
by addition to the carbonyl group
(Eq. 14-20, step b or 14-20, step b).
The resulting adducts are able to
undergo cleavage readily, as indicated
by the arrows showing the electron
flow toward the =N
+
group.
Below the structures of the adducts
in Eq. 14-20 are those of a 2-oxo acid
and a -ketol with arrows indicating
the electron flow in decarboxylation
and in the aldol cleavage. The similari-
ties to the thiamin-dependent cleavage
reaction are especially striking if one
remembers that in some aldolases and
decarboxylases the substrate carbonyl
group is first converted to an N-proto-
nated Schiff base before the bond cleavage.
We see that the essence of the action of thiamin diphos-
phate as a coenzyme is to convert the substrate into a form
in which electron flow can occur from the bond to be broken
into the structure of the coenzyme. Because of this alter-
ation in structure, a bond breaking reaction that would
not otherwise have been possible occurs readily. To
complete the catalytic cycle, the electron flow has to be
reversed again. The thiamin-bound cleavage product
(an enamine) from either of the adducts in Eq. 14-20
can be reconverted to the thiazolium dipolar ion and
an aldehyde as shown in step b of Eq. 14-21 for decar-
boxylation of pyruvate to acetaldehyde.
The adducts -lactylthiamin and -lactylthiamin
diphosphate have both been synthesized.
84,98 100
As
long as -lactylthiamin is kept as a dry solid or at low
pH, it is stable. However, it decarboxylates readily in
neutral solution (Eq. 14-21). Decarboxylation is much
CH
3
C
O
COO

CH
3
C
O
C
OH
CH
3
2 H
+
H
2 CO
2
2
(14-19)
a
Acidic
hydrogen
pK
a
~ 18
Adducts
formed by
thiamin and
substrates
Compare with
-oxo acid
and -oxo-
alcohol which
undergo easy
cleavage

N
N H
3
C
CH
2
NH
2
N CH
3
H
S
CH
2
CH
2
O
N
S
CH
2
R C COO

O
R C C R'
O OH
H
N
S
CH
2
C
R
HO
C
O

O
N
S
CH
2
C
R
HO
C
O
R'
H
H
O
C
O

C
C
O
O
C
C
C
O
+ +
Thiazolium
dipolar ion (ylid)
H
+
-Lactylthiamin
PP
+
b
b
733
more rapid in methanol, a fact that was predicted
by Lienhard and associates.
101
They suggested that
decarboxylation is easier in a solvent of low polarity
because the transition state has a lower polarity than
does lactylthiamin. An enzyme could assist the reac-
tion by providing a relatively nonpolar environment.
The crystal structures of thiamin-dependent en-
zymes (see next section) as well as modeling
102,103
suggest that lactylthiamin pyrophosphate has the
conformation shown in Eq. 14-21. If so, it would be
formed by the addition of the ylid to the carbonyl of
pyruvate in accord with stereoelectronic principles,
and the carboxylate group would also be in the correct
orientation for elimination to form the enamine in
Eq. 14-21, step b.
8283a
A transient 380- to 440-nm
absorption band arising during the action of pyruvate
decarboxylase has been attributed to the enamine.
What is the role of the pyrimidine portion of the
coenzyme in these reactions? The pyrimidine ring has
a large inductive effect on the basicity of the thiazolium
nitrogen and may increase the rate of dissociation of the
C-2 proton somewhat.
104
More significant is the fact
that the NH
2
group is properly placed to function as a
basic catalyst in the generation of the thiazolium dipolar
ion. However, the amino group of thiamin is not very
basic (pK
a
~ 4.9) and the site of protonation at low pH
is largely N-1 of the pyrimidine ring. In the protonated
form the NH
2
group is even less basic because of
electron withdrawal into the ring. Studies of thiamin
analogs suggested another possibility. Schellenberger
105
found pyruvate decarboxylase inactive when TDP was
substituted by analogs with modified aminopyrimidine
rings, e.g., with methylated or dimethylated amino
groups or with N1 of the ring replaced by carbon (an
aminopyridyl analog). More recently the experiment
has been repeated with additional enzymes
106
and
X-ray studies have shown that the analogs bind into
the active site of transketolase in a normal way.
107
Of
the compounds studied only an aminopyridyl analog of
TDP having a nitrogen atom at 1' (but CH at 3') had
substantial catalytic activity. Jordan and Mariam showed
that N-1'-methylthiamin is a superior catalyst in non-
enzymatic catalysis.
108
These results are consistent
with the speculative scheme illustrated in the following
drawing from the first edition of this book.
109
The NH
2
group of the N1-protonated aminopyrimidine has lost
a proton to form a normally minor tautomer in which the
resulting imino group would be quite basic. Assisted
by a basic group from the protein, it could abstract the
proton from the thiazolium ring to form the ylid.
Crystallographic studies show that the catalytic base
(:B-protein) is the carboxylate group of a conserved
glutamate side chain (E59' in Fig. 14-2). Kern et al.
used NMR spectroscopy of thiamin diphosphate
present in native and mutant pyruvate decarboxylase
and transketolase to monitor the exchange rates of the
C2-H proton of the thiazolium ring.
109a
The results
confirmed the importance of the conserved glutamate
side chain for dissociation of the C2-H proton. Partici-
pation of other catalytic groups from the enzyme may
also be important. However, these groups are not con-
served in the whole family of enzymes. For example,
glutamine 122, which is within hydrogen-bonding
distance of both the substrate and thiamin amino
group, is replaced by histidine in transketolase.
110
A
variety of kinetic studies involving mutants,
111,111a
alternative substrates,
112,113
and isotope effects in
substrates
114 116
and solvent
117,118
have not yet resolved
the details of the proton transfers that occur within the
active site.
3. Structures of Thiamin-Dependent Enzymes
By 1998, X-ray structures had been determined for
four thiamin diphosphate-dependent enzymes: (1) a
bacterial pyruvate oxidase,
119,120
(2) yeast and bacterial
pyruvate decarboxylases,
121122c
(3) transketolase,
110,123,124
and (4) benzoylformate decarboxylase.
124a
The reactions
catalyzed by these enzymes are all quite different, as
are the sequences of the proteins. However, the thiamin
diphosphate is bound in a similar way in all of them.
A conserved pattern of hydrogen bonds holds the
diphosphate group to the protein and also provides
ligands to a metal ion. This is normally Mg
2+
, which
is held in nearly perfect octahedral coordination by
two phosphate oxygen atoms, a conserved aspartate
carboxylate, a conserved asparagine amide, and a
water molecule. The thiamin rings are in a less polar
region. The amino group of the pyrimidine is adjacent
to the 2 CH of the thiazole and N1' of the pyrimidine
is apparently protonated and hydrogen bonded to the
carboxylate group of a conserved glutamate side
N
N
N
N
S
H
H
H
3
C
H
C
O
R
R'
+
Substrate
B-protein
D. Thiamine Diphosphate
734 Chapter 14. Coenzymes: Natures Special Reagents
P
O
O
FAD
N
H
O
F121
N
N
O
Q122
N
H
O
CH
3
2
N
S
H
H
N
N
H
O
O
H
N
H89
N
E59
S36
0.42 nm
F479
O
O
O
O
E483
R264
N
2
3
1
P
Figure 14-2 (A) Stereoscopic view of the active site of pyruvate oxidase from
the bacterium Lactobacillus plantarium showing the thiamin diphosphate as
well as the flavin part of the bound FAD. The planar structure of the part of
the intermediate enamine that arises from pyruvate is shown by dotted lines.
Only some residues that may be important for catalysis are displayed: G35',
S36', E59', H89', F121', Q122', R264, F479, and E483. Courtesy of Georg E.
Schulz.
119
(B) Simplified view with some atoms labeled and some side chains
omitted. The atoms of the hypothetical enamine that are formed from pyru-
vate, by decarboxylation, are shown in green.
A
B
4. The Variety of Enzymatic Reactions
Involving Thiamin
Most known thiamin diphosphate-dependent
reactions (Table 14-2) can be derived from the five half-
reactions, a through e, shown in Fig. 14-3. Each half-
reaction is an cleavage which
leads to a thiamin- bound enamine
(center, Fig. 14-3) The decarboxyla-
tion of an -oxo acid to an aldehyde
is represented by step b followed
by a in reverse. The most studied
enzyme catalyzing a reaction of this
type is yeast pyruvate decarboxy-
lase, an enzyme essential to alcoholic
fermentation (Fig. 10-3). There are
two ~ 250-kDa isoenzyme forms,
one an
4
tetramer and one with an
()
2
quaternary structure. The iso-
lation of -hydroxyethylthiamin
diphosphate from reaction mixtures
of this enzyme with pyruvate
52
provided important verification of
the mechanisms of Eqs. 14-14, 14-15.
Other decarboxylases produce alde-
hydes in specialized metabolic path-
ways: indolepyruvate decarboxylase
126
in the biosynthesis of the plant hor-
mone indole-3-acetate and ben-
zoylformate decarboxylase in the
mandelate pathway of bacterial
metabolism (Chapter 25).
124a,127
Formation of -ketols from -oxo
acids also starts with step b of Fig.
14-3 but is followed by condensation
with another carbonyl compound
in step c, in reverse. An example is
decarboxylation of pyruvate and
condensation of the resulting active
acetaldehyde with a second pyruvate
molecule to give R--acetolactate, a
reaction catalyzed by acetohydroxy
acid synthase (acetolactate syn-
thase).
128
Acetolactate is the precur-
sor to valine and leucine. A similar
ketol condensation, which is cata-
lyzed by the same synthase, is
required in the biosynthesis of
isoleucine (Fig. 24-17). Since this
chain. This is shown in Fig. 14-2. Substitution of the
corresponding glutamate 51 of yeast pyruvate decar-
boxylase by glutamine or alanine greatly reduced or
eliminated catalytic activity.
125
O
C
H
3
C C
COO

HO CH
3
R--Acetolactate
735 D. Thiamine Diphosphate
Figure 14-3 Half-reactions making up the thiamin-dependent cleavage and condensation reactions.
C HO OH
C O
R
C
HS
S
C O
R
O
C
C
R
O
OH
C
C
R
O
O

O
C O + H
+
C
HO O
S
S
H
+
+
b
c
d
e
H
2
O
CO
2
Acyl-
dihydro-
lipoyl
group
a
H
+
H
C
R
O
S
N
Pyrimidine
O P P
C
HO
R
C
C
O
O
R
synthase is not present in mammals it is a popular
target for herbicides.
129,130
It is inhibited by many of the
most widely used herbicides including sulfometuron
methyl, whose structure is shown here.
Acetolactate is a -oxo acid and is readily decar-
boxylated to acetoin, a reaction of importance in bacterial
fermentations (Eq. 17-26). Acetoin, of both R and S
1. Nonoxidative
Pyruvate decarboxylase*
Indolepyruvate decarboxylase
Benzoylformate decarboxylase*
Glyoxylate carboligase
Acetohydroxy acid synthase (acetolactate synthase)
1-Deoxy-d-xylulose 5-phosphate synthase
Transketolase*
Phosphoketolase
2. Oxidative decarboxylase
Pyruvate oxidase (FAD)*
Pyruvate dehydrogenase (Lipoyl, FAD, NAD
+
)
multienzyme complex
Pyruvate:ferredoxin oxidoreductase
Indolepyruvate:ferredoxin oxidoreductase
TABLE 14-2
Enzymes Dependent upon Thiamin Diphosphate
as a Coenzyme
* Three-dimensional structures for these enzymes had been
determined by 1998.
C
H
3
CO
O S
O O
Sulfometuron methyl
NH
C
O NH
N N
H
3
C CH
3
736 Chapter 14. Coenzymes: Natures Special Reagents
configurations, is also formed by pyruvate decarboxy-
lases acting on acetaldehyde.
103,131
The ketol conden-
sation of two molecules of glyoxylate with decarboxy-
lation to form tartronic semialdehyde (see Fig. 17-6) is
an important reaction in bacterial metabolism. It is
catalyzed by glyoxylate carboligase,
132
another
thiamin diphosphate-dependent enzyme. Formation
of 1-deoxy-D-xylulose 5-phosphate, an intermediate in
the nonmevalonate pathway of isoprenoid synthesis,
is formed in a thiamin diphosphate-catalyzed conden-
sation of pyruvate with glyceraldehyde 3-phosophate
(Fig. 22-2)
132a
However, there is an unresolved prob-
lem. As previously mentioned, the same intermediate
is thought to be a precursor to thiamin diphosphate
(Eq. 25-21). This suggests the presence of an alternative
pathway.
Ketols can also be formed enzymatically by cleavage
of an aldehyde (step a, Fig. 14-3) followed by conden-
sation with a second aldehyde (step c, in reverse). An
enzyme utilizing these steps is transketolase (Eq.
17-15),
132b
which is essential in the pentose phosphate
pathways of metabolism and in photosynthesis.
-Diketones can be cleaved (step d) to a carboxylic
acid plus active aldehyde, which can react either via
a or c in reverse. These and other combinations of
steps are often observed as side reactions of such
enzymes as pyruvate decarboxylase. A related thia-
min-dependent reaction is that of pyruvate and acetyl-
CoA to give the -diketone, diacetyl, CH
3
COCOCH
3
.
133
The reaction can be viewed as a displacement of the
CoA anion from acetyl-CoA by attack of thiamin-
bound active acetaldehyde derived from pyruvate
(reverse of step d, Fig. 14-3 with release of CoA).
5. Oxidative Decarboxylation and
2-Acetylthiamin Diphosphate
The oxidative decarboxylation of pyruvate to
form acetyl-CoA or acetyl phosphate plays a central
role in the metabolism of our bodies and of most other
S
N
C
HO
H
3
C
Pyrimidine
S
N
C
O
H
3
C
Pyrimidine
+
2-Acetylthiamin diphosphate
H
+
+ 2e

(14-22)
organisms. This reaction is usually formulated as the
reverse of step e of Fig. 14-3, which shows the cleavage
of an acyl-dihydrolipoyl derivative. However, there
is a possibility that the lipoyl group functions not as
shown in Fig. 14-3 but as an oxidant that converts the
TDP enamine to 2-acetylthiamin diphosphate (Eq. 14-
22)
134,135
and only after that as an acyl group carrier.
A related reaction that is known to proceed through
acetyl-TDP is the previously mentioned bacterial
pyruvate oxidase. As seen in Fig. 14-2, this enzyme
has its own oxidant, FAD, which is ready to accept the
two electrons of Eq. 14-22 to produce bound acetyl-
TDP. The electrons may be able to jump directly to the
FAD, with thiamin and flavin radicals being formed
at an intermediate stage.
135a
The electron transfers as
well as other aspects of oxidative decarboxylation are
discussed in Chapter 15, Section C.
A reaction that is related to that of transketolase but
is likely to function via acetyl-TDP is phosphoketolase,
whose action is required in the energy metabolism of
some bacteria (Eq. 14-23). A product of phosphoketolase is
acetyl phosphate, whose cleavage can be coupled to
synthesis of ATP. Phosphoketolase presumably cata-
lyzes an cleavage to the thiamin-containing enamine
shown in Fig. 14-3. A possible mechanism of formation
of acetyl phosphate is elimination of H
2
O from this
enamine, tautomerization to 2-acetylthiamin, and
reaction of the latter with inorganic phosphate.
6. Thiamin Coenzymes in Nerve Action
The striking paralysis caused by thiamin deficiency
together with studies of thiamin analogs as metabolites
suggested a special action for this vitamin in nerves.
136
The thiamin analog pyrithiamin (p. 731) both induces
paralytic symptoms and displaces thiamin from nerve
preparations. The nerve poison tetrodotoxin (Chap-
ter 30) blocks nerve conduction by inhibiting inward
diffusion of sodium, but it also promotes release of
thiamin from nerve membranes. Evidence for a meta-
bolic significance of thiamin triphosphate comes from
identification of soluble and membrane-associated
thiamin triphosphatases
137
as well as a kinase that
CH
2
OH
C
C
O
R
H HO
CH
3
C
OPO
3
2
O
C
O H
R
CH
3
COO

Acetyl
phosphate
P
i
ADP
ATP
(14-23)
737
forms protein-bound thiamin triphosphate in the
brain.
138
Mono-, tri-, and tetraphosphates also occur
naturally in smaller amounts. One might speculate
about a possible role for the rapid interconversion of
cationic and yellow anionic forms of thiamin via the
tricyclic form (Eq. 7-19) in some aspect of nerve con-
duction.
E. Pyridoxal Phosphate
The phosphate ester of the aldehyde form of vitamin
B
6
, pyridoxal phosphate (pyridoxal-P or PLP), is
required by many enzymes catalyzing reactions of
amino acids and amines. The reactions are numerous,
and pyridoxal phosphate is surely one of natures most
versatile catalysts. The story begins with biochemical
transamination, a process of central importance in
nitrogen metabolism. In 1937, Alexander Braunstein
and Maria Kritzmann, in Moscow, described the trans-
amination reaction by which amino groups can be
transferred from one carbon skeleton to another.
139,140
For example, the amino group of glutamate can be
transferred to the carbon skeleton of oxaloacetate to
form aspartate and 2-oxoglutarate (Eq. 14-24).
This transamination reaction is a widespread process
of importance in many aspects of the nitrogen metabo-
lism of organisms. A large series of transaminases
(aminotransferases), for which glutamate is most
often one of the reactants, have been shown to catalyze
the reactions of other oxoacids and amino acids.
141 143
In 1944, Esmond Snell reported the nonenzymatic
conversion of pyridoxal into pyridoxamine (Box 14-C)
by heating with glutamate. He recognized that this
was also transamination and proposed that pyridoxal
might be a part of a coenzyme needed for aminotrans-
ferases and that these enzymes might act via two half-
reactions that interconverted pyridoxal and pyridox-
amine (Eq. 14-25). The hypothesis was soon verified
and the coenzyme was identified as pyridoxal 5'-phos-
phate or pyridoxamine 5'-phosphate (Fig. 14-5).
144,145
At about the same time, Gunsalus and coworkers
noticed that the activity of tyrosine decarboxylase
produced by lactic acid bacteria was unusually low
when the medium was deficient in pyridoxine. Addi-
tion of pyridoxal plus ATP increased the decarboxylase
activity of cell extracts.
146
PLP was synthesized and
was found to be the essential coenzyme for this and a
variety of other enzymes.
147
1. Nonenzymatic Models
Pyridoxal or PLP, in the complete absence of en-
zymes, not only undergoes slow transamination with
amino acids but also catalyzes many other reactions
of amino acids that are identical to those catalyzed by
PLP-dependent enzymes. Thus, the coenzyme itself can
be regarded as the active site of the enzymes and can be
studied in nonenzymatic reactions. The latter can be
thought of as models for corresponding enzymatic
reactions. From such studies Snell and associates
drew the following conclusions.
148
a. The aldehyde group of PLP reacts readily and
reversibly with amino acids to form Schiff bases
(Fig. 14-4) which react further to give products.
b. For an aldehyde to be a catalyst, a strong electron-
attracting group, e.g., the ring nitrogen of pyridine
(as in PLP), must be ortho or para to the CHO
group. A nitro group, also strongly electron
attracting, can replace the pyridine nitrogen in
model reactions.
E. Pyridoxal Phosphate
Amino acid 1
Oxoacid 1
Oxoacid 2
Amino acid 2
First half-reaction
Second half-reaction
PMP-Enzyme PLP-Enzyme
Sum: Amino acid 1 + oxoacid 2
oxoacid 1 + amino acid 2
(14-25)

OOC
C
COO

NH
3
+
H

OOC
C
COO

OOC
C
COO

OOC C
COO

NH
3
+
H
Oxaloacetate
l-Glutamate
2-Oxoglutarate
l-Aspartate
Transamination
(14-24)
CHO
NO
2
N
CHO
vs
738 Chapter 14. Coenzymes: Natures Special Reagents
BOX 14-C THE VITAMIN B
6
FAMILY: PYRIDOXINE, PYRIDOXAL, AND PYRIDOXAMINE
Pyridoxine, the usual commercial form of
vitamin B
6
, was isolated and synthesized in 1938.
However, studies of bacterial nutrition soon indicated
the existence of other naturally occurring forms of
the new vitamin which were more active than pyri-
doxine in promoting growth of certain lactic acid
bacteria. The amine pyridoxamine and the aldehyde
pyridoxal were identified by Snell, who found that
pyridoxal could be formed from pyridoxine by mild
oxidation and that pyridoxamine could be formed
from pyridoxal by heating in a solution with gluta-
mic acid via a transamination reaction. These simple
experiments also suggested the correct structures of
the new forms of vitamin B
6
. Animal tissues contain
largely pyridoxal, pyridoxamine, and their phosphate
esters. The lability of the aldehyde explains the ease
of destruction of the vitamin by excessive heat or by
light. On the other hand, plant tissues contain mostly
pyridoxine, which is more stable. Kinases use ATP
to form the phosphate esters, which are intercon-
vertible within cells.
ae
Pyridoxine 5'-phosphate can
be oxidized to PLP
d,e,f
and the latter may undergo
transamination to PMP. The acidbase chemistry
and tautomerism of pyridoxine were discussed in
Chapter 6, Section E,2.
Many poisonous substances as well as useful
drugs react with PLP-requiring enzymes. Thus,
much of the toxic effect of the carbonyl reagents
hydroxylamine, hydrazine, and semicarbazide
stems from their formation of stable derivatives
analogous to Schiff bases with PLP.
Isonicotinyl hydrazide (INH), one of the most
effective drugs against tuberculosis, is inhibitory to
pyridoxal kinase, the enzyme that converts pyridoxal
to PLP.
c
Apparently, the drug reacts with pyridoxal
to form a hydrazone which blocks the enzyme.
Pyridoxal kinase is not the primary target of INH
in mycobacteria. However, patients on long-term
isonicotinyl hydrazide therapy sometimes suffer
symptoms of vitamin B
6
deficiency.
g
PLP-dependent enzymes
are inhibited by a great variety
of enzyme-activated inhibitors
that react by several distinctly
different chemical mechanisms.
h
Here are a few. The naturally
occurring gabaculline mimics
-aminobutyrate (Gaba) and
inhibits -aminobutyrate amino-
transferase as well as other PLP-
dependent enzymes. The inhi-
bitor follows the normal catalytic pathway as
far as the ketimine. There, a proton is lost from the
inhibitor permitting formation of a stable benzene
ring and leaving the inhibitor stuck in the active site:
in pyridoxal
phosphate
in pyridoxal
in pyridoxamine
O
O

P
H
O H
2
C
H
2
C
O

OH
H
2
C
NH
3
CH
3
H
H
N
+ 1
6
5
4
2
3
4
C
O
H
+
Pyridoxine (pyridoxol)
Vitamin B
6
alcohol
-Aminobutyrate
(Gaba)
COO

+
H
3
N
COO

+
NH
3
H
H
H
Gabaculline
H
H
H
+
N
H
2
C
CH
3
N
H
+
O

COO

+
NH
2
H
2
C
CH
3
N
H
+
O

Stably bound
inhibitor
As in Eq. 14-28,
reverse
PLP
COO

N N
H
H
H
H
Hydrazine
N N
H
H
Semicarbazide
C NH
2
O
H
H O NH
2
, R O NH
2
Hydroxylamines
N
Isonicotinyl hydrazide
(isoniazid)
C O
NH
NH
2
739
BOX 14-C (continued)
Beta-chloroalanine and serine O-sulfate can
undergo elimination (as in Eq. 14-29) in active sites of
glutamate decarboxylase or aspartate aminotrans-
ferase. The enzymes then form free aminoacrylate,
a reactive molecule that can undergo an aldol-type
condensation with the external aldimine to give the
following product.
i
Nucleophilic groups from enzymes can add to
double bonds, e.g., in an aminoacrylate Schiff base,
or to multiple bonds present in the inhibitor. An
example is -vinyl -aminobutyrate (4-amino-5-
hexenoic acid), another inhibitor of brain -amino-
butyrate aminotransferase which is a useful anti-
convulsant drug.
Another enzyme-activated inhibitor is the
streptomyces antibiotic D-cycloserine (oxamycin),
an antitubercular drug that resembles D-alanine in
structure. A potent inhibitor of alanine racemase, it
also inhibits the non-PLP, ATP-dependent, D-alanyl-
D-alanine synthetase which is needed in the bio-
synthesis of the peptidoglycan of bacterial cell walls.
L-Cycloserine inhibits many PLP enzymes and
is toxic to humans. This observation led Khomutov
et al. to synthesize the following more specific cyclo-
glutamates, structural analogs of glutamic acid
with fixed conformations.
j,k
Nature apparently
anticipated the synthetic chemist, because it has
been reported that the mushroom Tricholoma muscar-
ium contains one of the same compounds. It is said
to impart two interesting properties to the mush-
room: a delicious flavor and a lethal action on flies
that alight on the mushrooms surface!
l
The substituted cysteine derivative L-penicil-
lamine causes convulsions and low glutamate decar-
boxylase levels in the brain, presumably because
the Schiff base formed with PLP can then undergo
cyclization, the SH group adding to the C = N to
form a stable thiazolidine ring.
Toxopyrimidine, the alcohol derived from the
pyrimidine portion of the thiamin molecule, is a
structural analog of pyridoxal. When fed to rats or
mice it induces running fits which can be stopped
by administration of vitamin B
6
. Phophorylation of
toxopyrimidine by pyridoxal kinase may produce
an antagonistic analog of PLP. In a similar fashion,
4-deoxypyridoxine, which was tested as a possible
anticancer drug, caused convulsions and other
symptons of vitamin B
6
deficiency in humans. A
host of synthetic PLP derivatives have been made,
some of which are effective in blocking PLP enzymes.
h
a
Lepkovsky, S. (1979) Fed. Proc. 38, 26992700
b
McCormick, D. B., Gregory, M. E., and Snell, E. E. (1961) J. Biol.
Chem. 236, 20762084
c
Snell, E. E., and Haskell, B. E. (1970) Comprehensive Biochemistry
21, 4771
d
McCormick, D. B. and Chen, H. (1999) J. Nutr. 129, 325327
e
Hanna, M. L., Turner, A. J., and Kirkness, E. F. (1997) J. Biol.
Chem. 272, 1075610760
f
Ngo, E. O., LePage, G. R., Thanassi, J. W., Meisler, N., and
Nutter, L. M. (1998) Biochemistry 37, 77417748
g
Lui, A., and Lumeng, L. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical Aspects, Vol. 1B
(Dolphin, D., Poulson, R., and Avramovc, O., eds), pp.
601674, Wiley, New York
h
Walsh, C. T. (1986) in Vitamin B
6
, Pyridoxal Phosphate: Chemical,
Biochemical and Medical Aspects, Vol. 1B (Dolphin, D., Poulson,
R., and Avramovc, O., eds), pp. 4370, Wiley, New York
i
Likos, J. J., Ueno, H., Fedhaus, R. W., and Metzler, D. E. (1982)
Biochemistry 21, 43774386
j
Khomutov, R. M., Koveleva, G. K., Severin, E. S., and Vdovina,
L. V. (1967) Biokhim. 32, 900907
k
Sastchenko, L. P., Severin, E. S., Metzler, D. E., and Khomutov,
R. M. (1971) Biochemistry 10, 48884894
l
Iwasaki, H., Kamiya, T., Oka, O., and Veyanagi, J. (1969) Chem.
Pharm. Bull. 17, 866872
E. Pyridoxal Phosphate
H
3
C C
COO
H
H
3
C SH
NH
3
+
H
3
C
C
COO

H
H
3
C
S N
l-Penicillamine
C
PLP
H
PLP
O N

O
H
3
+
N H
C
O
O

A cycloglutamate that inhibits


aspartate aminotransferase
O N
O

H H
3
+
N
C
O

O
An isomeric cycloglutamate tricholomic
acid found in certain mushrooms
H
O N
O
H NH
2
H
O N

O
H NH
3
+
d-Cycloserine
HN
C
H
2
C
COO

C
NH
Lysine (enzyme)
H
PLP
740 Chapter 14. Coenzymes: Natures Special Reagents
c. The presence of an OH group adjacent to the
CHO group greatly enhances the catalytic activity.
Since certain metal ions, such as Cu
2+
and Al
3+
,
increase the rates in model systems and are known
to chelate with Schiff bases of the type formed
with PLP, it was concluded that either a metal ion
or a proton formed a chelate ring and helped to
hold the Schiff base in a planar conformation
(Fig. 14-6). However, such a function for metal ions
has not been found in PLP-dependent enzymes.
d. In model systems the 5-hydroxymethyl and
2-methyl groups are not needed for catalysis.
However, in enzymes the 5-CH
2
OH group is
essential for attachment of the phosphate handle.
The 2-CH
3
group is usually not necessary for
coenzymatic activity.
Many investigations of nonenzymatic reactions of
PLP and related compounds have been and are still
being conducted
149,150
2. A General Mechanism of Action of PLP
Based upon consideration of the various known
PLP-dependent enzymes of amino acid metabolism,
Braunstein and Shemyakin in 1952 proposed a general
mechanism of PLP action
151,152
which, in most details,
was the same as the one proposed independently by
Snell and associates on the basis of the nonenzymatic
reactions.
148
The general mechanism, which has been
verified by studies of many enzymes, can be stated as
follows: Pyridoxal phosphate reacts to convert the amino
group of a substrate into a Schiff base that is electronically
the equivalent of an adjacent carbonyl (Fig. 14-4). However,
a Schiff base of an amino acid with a simple aldehyde
(for example, acetaldehyde) has the opposite polarity
from that of C = O (see the following structures). Such
an imine could not substitute for a carbonyl group in
activating an -hydrogen nor in facilitating CC bond
cleavage in the amino acid. It is necessary to have the
strongly electron-attracting pyridine group conjugated
with the C = N group in such a way that electrons can
flow from the substrate into the coenzyme.
Pyridoxal phosphate (PLP) exists
mainly as the dipolar ion
Carbonyl group reacts
with amino group to
form a Schiff base Bridging H
+
holds conjugated
-system planar
Phosphate
handle
Strong electron-attracting
group assists in stabilizing
transition state
This proton can be
replaced by M
2+
in model reactions
Schiff base
(two resonance forms)
N CH
3
C
N
H
O

CH
2
C
R COO

H
+
+
H
O
2
O
3
P
N CH
3
C
N
H
O
C
R COO

H
CH
2
+
O
2
O
3
P
H
H
H
C
NH
3
+
R COO
H

N
O

CH
3
C
O
C
O
P
O
O

O
H H
H
+
1
2
3
4
5
6
5
H
Figure 14-4 Pyridoxal 5'-phosphate (PLP), a special coenzyme for reactions of amino acids.
741 E. Pyridoxal Phosphate
Lysine
side
chain
External substrate-coenzyme
Schiff base
N CH
3
O

C O
2
O
3
P N
+
C
H
COO

R
H
NH
2
H
+
H
N CH
3
O

C O
2
O
3
P
HN
N H
C
R
COO

H
H
+
H
H
+
Adduct, a geminal diamine
S configuration
Internal enzyme-coenzyme
Schiff base
N CH
3
O

HC O
2
O
3
P N
H
+
Lysine
side
chain
l-Amino acid substrate
C
NH
3
+
H
R COO

(14-26)
Before discussing the reactions of Schiff bases of
PLP we should consider one fact that was not known
in 1952. PLP is bound into an enzymes active site as
a Schiff base with a specific lysine side chain before
a substate binds . This is often called the internal
aldimine. When the substrate binds it reacts with
the internal Schiff base by a two-step process called
transimination (Eq. 14-26) to form the substrate Schiff
base, which is also called the external aldimine.
C
C C
H
CH
3
N
C
O
H
H
R COO

R COO

Schiff base with


acetaldehyde
Carbonyl
group
3. The Variety of PLP-Dependent Reactions
In Fig. 14-5 the reactions of PLP-amino acid Schiff
bases are compared with those of -oxo-acids. Beta-
hydroxy--oxo acids and Schiff bases of PLP with
-hydroxy--amino acids can react in similar ways.
The reactions fall naturally into three groups (a,b,c)
depending upon whether the bond cleaved is from the
-carbon of the substrate to the hydrogen atom, to the
carboxyl group, or to the side chain. A fourth group of
reactions of PLP-dependent enzymes (d) also involve
removal of the -hydrogen but are mechanistically
more complex. Some of the many reactions catalyzed
by these enzymes are listed in Table 14-3.
Loss of the -hydrogen (Group a). Dissociation
of the -hydrogen from the Schiff base leads to a
quinonoidcarbanionic intermediate whose struc-
ture in depicted in Fig. 14-5. The name reflects the
characteristics of the two resonance forms drawn.
Like an enolate anion, this intermediate can react in
several ways (1-4).
(1) Racemization. A proton can be added back
to the original alpha position but without stereospeci-
ficity. A racemase which does this is important to
bacteria. They must synthesize D-alanine and D-glu-
tamic acid from the corresponding L-isomers for use
in formation of their peptidoglycan envelopes.
153154a
The combined actions of alanine racemase plus D-alanine
aminotransferase, which produces D-glutamate as a
product, provide bacteria with both D amino acids.
A fungal alanine racemase is necessary for synthesis
of the immunosuppresant cyclosporin (Box 9-F).
155,155a
High concentrations of free D-alanine are found in
certain regions of the brain and also in various glands.
156
The carboxyl group of an amino acid can also
activate the -hydrogen. This may be the basis for an
aspartate racemase and other racemases that are not
dependent upon PLP.
156 158
See also Chapter 13,
Section B,4.
(2) Cyclization. A second kind of reaction is
represented by the conversion of S-adenosylmethionine
to aminocyclopropanecarboxylic acid, a precursor
to the plant hormone ethylene (see Chapter 24).
159
The quinonoid intermediate cyclizes with elimination
of methylthioadenosine to give a Schiff base of the
product (Eq. 14-27).
160161a
The cyclization step appears
to be a simple S
N
2-like reaction.
162
(3) Transamination. A proton can add to the
carbon attached to the 4 position of the PLP ring (Fig.
14-5) to form a second Schiff base, often referred to as
a ketimine (Eq. 14-28). The latter can readily undergo
hydrolysis to pyridoxamine phosphate (PMP) and
an -oxo acid. This sequence represents one of the
742 Chapter 14. Coenzymes: Natures Special Reagents
two half-reactions (Eqs. 14-24 and 14-25) required for
enzymatic transamination.
Transaminases participate in metabolism of most
of the amino acids, over 60 different enzymes have
been identified.
142,163
Best studied are the aspartate
aminotransferases, a pair of cytosolic and mitochon-
drial isoenzymes which can be isolated readily from
animal hearts. Their presence in heart muscle and
brain in high concentration is thought to be a result
of their functioning in the malateaspartate shuttle
(Chapter 18). The sequences of the
two proteins differ greatly, with
only 50% of the residues being the
same in both isoenzymes. However,
these differences are largely on the
outside surface, the folding pattern
(Fig. 2-6) and internal structure (Fig.
14-6) are almost identical.
142,163 167a
Three-dimensional structures of
aspartate aminotransferases of E.
coli, yeast, chickens, and mammals
are extremely similar, even though
sequence identity may be as low as
20%.
167a
Most other transaminases
also use the L-gutamateoxoglut-
arate pair as one of the product
reactant pairs but a few prefer
smaller substrates with uncharged
side chains. An example is serine:
pyruvate (or alanine:glyoxylate)
aminotransferase, an important mito-
chondrial and peroxisomal enzyme
in both animals and plants.
168
Other
specialized aminotransferases act
on aromatic amino acids,
168a
the
branched chain amino acids valine,
leucine, and isoleucine,
168b,c
and
D-amino acids.
168d
Many of them are
highly specific for individual amino
acids such as phosphoserine,
168e
ornithine,
168f,g
N-acetylornithine,
168h,i
and 8-amino-7-oxononanoate (see
banner, p. 719).
168j
An apparently
internal transamination, which
requires PMP and PLP, converts
glutamate-1-semialdehyde into
-aminolevulinate (see Eq. 24-44)
in the pathway of porphyrin bio-
synthesis used by bacteria and
plants.
168k
(4) Elimination and replace-
ment. When a good leaving group
is present in the position of the
amino acid it can be eliminated
(Fig. 14-5, Eq. 14-29).
170
A large
number of enzymes catalyze such
reactions. Among them are serine and threonine
dehydratases, which eliminate OH

as H
2
O;
171 173a
tryptophan indole-lyase (tryptophanase) of bacteria,
which eliminates indole;
174176c
tyrosine phenol-lyase
(elimination of phenol);
177178a
and alliinase of garlic
(elimination of 1-propenylsulfenic acid).
179,180
Cysta-
thionine, a precursor to methionine, eliminates L-homo-
cysteine through the action of cystathionine lyase
(cystathionase).
181,182
Ammonia is eliminated from the
position of 2,3-diaminopropionate by a bacterial lyase.
183
N
O

H
N
C
2
O
3
PO
CH
3
H
C
H
+
+
H
COO

C
Y
H
R
C
C
O
H
COO

C
HO
H
R
Compare the reactions of the
PLP Schiff bases (below) with
those of this ketone
(a) Removal of H to form enolate anion
and loss of OH (, elimination)
(b) Decarboxylation (of a -oxoacid)
(c) Aldol cleavage
Bonds around the -carbon atom of the
Schiff base can also be cleaved in 3 ways
by withdrawal of electrons into
the coenzyme
(a) Removal of H as H
+
to yield
quinonoid intermediate shown below
(b) Decarboxylation
(c) Aldol cleavage (if Y is OH)
(1) H
+
can be added back
(nonstereospecifically)
to cause racemization
Schiff base (aldimine)
(3) H
+
can add here
to form ketimine.
Subsequent reactions
lead to transamination
a
b c
a
b c
(4) Y

can be eliminated
Quinonoidcarbanionic structure
(analogous to an enolate anion)
N
O

H
N
C
2
O
3
PO
CH
3
H
C
+
H
C
Y
H
R
H
C
O
O

+
H
+
N
O

H
N
C
2
O
3
PO
CH
3
H
C
+
C
Y
H
R
H
C
O
O

The quinonoid intermediate formed by


cleavage at a can react in various ways (14)
(2) Cyclization
(Eq. 14-27)
Figure 14-5 Some reactions of Schiff bases of pyridoxal phosphate. (a)
Formation of the quinonoid intermediate, (b) elimination of a substituent,
and (c) transamination. The quinonoidcarbanionic intermediate can react
in four ways (14) if enzyme specificity and substrate structure allow.
743
S
+
CH
2
CH
2
CH
2
C
H
3
C
NH
3
+
H
COO

R
S
+
CH
2
Adenosyl
CH
2
C
H
3
C
N
COO

HC
CH
2
H
2
C C
NH
+
COO

HC
5'
CH
2
H
2
C C
NH
3
+
PLP
Adenosyl
Quinonoidcarbanionic
intermediate
Schiff base
COO

Adenosyl-S-CH
3
Aminocyclopropane
carboxylate (ACC)
PLP
S-Adenosylmethionine
(14-27)
H
O

N
C
R COO

N CH
3
+
H
O

H
2
C
N
C
R COO

N CH
3
Ketimine
(second Schiff base)
H
H
COO

C
O
R
N CH
3
H
2
C
O

NH
3
+
+
H
2
O
-Oxo acid
H
Pyridoxamine phosphate
(PMP)
H
+
+
+
Quinonoidcarbanionic
form
(14-28)
E. Pyridoxal Phosphate
(a) Removing alpha hydrogen as H
+
(1) Racemization
Alanine racemase*
(2) Cyclization
Aminocyclopropane carboxylate synthase
(3) Amino group transfer
Aspartate aminotransferase*
Alanine aminotransferase
D-Amino acid aminotransferase*
Branched chain aminotransferase
Gamma-aminobutyrate aminotransferase
-Amino acid:pyruvate aminotransferase*
Tyrosine aminotransferase
Serine:pyruvate aminotransferase
(4) Beta elimination or replacement
D- and L- Serine dehydratases (deaminases)
Tryptophan indole-lyase (tryptophanase)*
Tyrosine phenol-lyase*
Alliinase
Cystathionine -lyase (cystathionase)*
O-Acetylserine sulfhydrylase (cysteine
synthase)
Cystathionine -synthase
Tryptophan synthase*
(b) Removal of alpha carboxylate as CO
2
Diaminopimelate decarboxylase
Glycine decarboxylase (requires lipoyl group)
Glutamate decarboxylase
Histidine decarboxylase
Dopa decarboxylase
Ornithine decarboxylase*
Tyrosine decarboxylase
Dialkylglycine decarboxylase (a decarboxylating
transaminase)*
(c) Removal or replacement of side chain (or H) by aldol
cleavage
Serine hydroxymethyltransferase
Threonine aldolase
-Aminolevulinate synthase
Serine palmitoyltransferase
2-Amino-3-oxobutyrate-CoA ligase
(d) Reactions of ketimine intermediates
Aspartate -decarboxylase
Selenocysteine lyase
Nif S protein of nitrogenase
Gamma elimination and replacement
Cystathionine -synthase
Cystathionine -lyase
Threonine synthase
(e) Other enzymes
Lysine 2,3-aminomutase
Glycogen phosphorylase*
Pyridoxamine phosphate (PMP) in synthesis of
3,6-dideoxy hexoses
* The three-dimensional structures of these and other PLP-
dependent enzymes were determined by 2000.
TABLE 14-3
Some Enzymes That Require Pyridoxal Phosphate
as a Coenzyme
744 Chapter 14. Coenzymes: Natures Special Reagents
proton at the -C atom of 2-oxobutyrate
189
suggests
that these steps may occur with the participation of
groups from the enzyme. Before indole can be elim-
inated by tryptophan indolelyase the indole ring must
presumably be tautomerized to the following form of
the quinonoid intermediate. The same species may
be created by tryptophan synthase upon addition of
indole to the enzyme-bound aminoacrylate. The green
arrows on the structure indicate the tautomerization
that would occur to convert the indole ring to the
structure found in tryptophan. The three-dimensional
structure of tryptophan synthase is shown in Fig. 25-3.
It is a complex of two enzymes with a remarkable
tunnel through which the intermediate indole can
pass.
184,190
An unusual PLP-dependent replacement
is used to synthesize a transfer RNA ester of seleno-
cysteine prior to its insertion into special locations in
a few proteins (Chapter 15, Section G, and Chapter 29).
Decarboxylation (Group b). The bond to the car-
boxyl group of an amino acid substrate is broken in reac-
tions catalyzed by amino acid decarboxylases.
191,192
These also presumably lead to a transient quinonoid-
carbanionic intermediate. Addition of a proton at the
original site of decarboxylation followed by breakup
of the Schiff base completes the sequence. Decarboxy-
lation of amino acids is nearly irreversible and frequently
appears as a final step in synthesis of amino compounds.
For example, in the brain glutamic acid is decarboxy-
lated to -aminobutyric acid (Gaba),
193 196b
while
3,4-dihydroxyphenylalanine (dopa) and 5-hydroxy-
H
O

N
C
COO

N CH
3
Y
H
R
H
H
O

N
C
C COO

R
H
N CH
3
C
NH
2
COO

RCH
C
NH
COO

RCH
2
+
+
H H
+
C
O
COO

RCH
2
+
YH
Aminoacrylate or
aminocrotonate
Transimination
Imino acid
Pyruvate or
2-oxobutyrate
H
2
O
Schiff base of
aminoacrylate
(R=H) or
aminocrotonate
(R=CH
3
)
NH
4
+
(14-29)
Beta replacement is catalyzed by such enzymes
of amino acid biosynthesis as tryptophan synthase
(Chapter 25),
184
O-acetylserine sulfhydrylase
(cysteine synthase),
185186a
and cystathionine -synthase
(Chapter 24).
187188c
In both elimination and replace-
ment an unsaturated Schiff base, usually of aminoacryl-
ate or aminocrotonate, is a probable intermediate (Eq.
14-29). Conversion to the final products is usually
assumed to be via hydrolysis to free aminoacrylate,
tautomerization to an imino acid, and hydrolysis of the
latter, e.g., to pyruvate and ammonium ion (Eq. 14-29).
However, the observed stereospecific addition of a
Figure 14-6 Drawing showing pyridoxal phosphate (shaded)
and some surrounding protein structure in the active site of
cytosolic aspartate aminotransferase. This is the low pH
form of the enzyme with an N-protonated Schiff base linkage
of lysine 258 to the PLP. The tryptophan 140 ring lies in front
of the coenzyme. Several protons, labeled H
a
, H
b
, and H
d,
are
represented in
1
H NMR spectra by distinct resonances whose
chemical shifts are sensitive to changes in the active site.
169
Y70
K258
O
H
Y225
N
H
S
O
O
O H
O
H
N
O
H
H
N O
O
H
N
N194
H
N
O
H
N
N
H
H
N
N
N
H
N
N
N
N
O
O
T109
G108
D222
H189 140
W140
T139
H143
N
H
O
H
3
C
H
b
H
d
H193
P
S223
N
H
a
G38

H
CH
3
+
+

N
O

H
N
HC
H
C
COO

N
BH
H
+
+
Quinonoidcarbanionic
intermediate with
tautomerized indole ring
745 E. Pyridoxal Phosphate
tryptophan are acted upon by an aromatic amino
acid decarboxylase to form, respectively, the neuro-
transmitters dopamine and serotonin.
197 199b
Histi-
dine is decarboxylated to histamine.
200 202
However,
not all histidine decarboxylases use PLP as a coen-
zyme (Section F).
Arginine is converted by a PLP-dependent decar-
boxylase to agmatine (Fig. 24-12) which is hydrolyzed
to 1,4-diaminopropane.
191
This important cell con-
stituent is also formed by hydrolysis of arginine to
ornithine (Fig 24-10) and decarboxylation of the lat-
ter.
203 206c
Lysine is formed in bacteria by decarboxyla-
tion of meso-diamino-pimelic acid (Fig. 24-14). Glycine
is decarboxylated oxidatively in mitochondria in a
sequence requiring lipoic acid and tetrahydrofolate as
well as PLP (Fig. 15-20).
207 209b
A methionine decar-
boxylase has been isolated in pure form from a fern.
210
The bacterial dialkylglycine decarboxylase is both a
decarboxylase and an aminotransferase which uses
pyruvate as its second substrate forming a ketone and
L-alanine as products (See Eq. 14-37)
210a, 210b
Side chain cleavage (Group c). In a third type
of reaction the side chain of the Schiff base of Fig. 14-5
undergoes aldol cleavage. Conversely, a side chain can
be added by condensation. The best known enzyme
of this group is serine hydroxymethyltransferase,
which converts serine to glycine and formaldehyde.
211213b
The latter is not released in a free form but is transferred
by the same enzyme specifically to tetrahydrofolic
acid (Eq. 14-30), with which it forms a cyclic adduct.
Threonine is cleaved to acetaldehyde by the same
enzyme. A related reaction is indicated in Fig. 24-27
(top). In a more important pathway of degradation
of threonine the hydroxyl group of its side chain is
dehydrogenated to form 2-amino-3-oxobutyrate which
is cleaved by a PLP-dependent enzyme to glycine and
acetyl-CoA (Eq. 14-31).
214,215
Conversely, ester condensation reactions join acyl
groups from CoA derivatives to Schiff bases derived
from glycine or serine. Succinyl-CoA is the acyl donor
l-Serine
Glycine
THF (Tetrahydrofolate)
See Fig 15-18
N
5
, N
10
-Methylene-THF
(14-30)
H
3
C
C
C
O
H
COO

NH
3
+
Glycine
CoA-SH Acetyl-S-CoA
(14-31)
in Eq. 14-32 for the second known pathway for biosyn-
thesis of -aminolevulinic acid, an intermediate in
porphyrin synthesis (Chapter 24).
216 218b
The enzyme
does not catalyze decarboxylation of glycine in the
absence of succinyl-CoA, and the decarboxylation
probably follows the condensation as indicated in Eq.
14-32.
219
In a similar reaction in the biosynthesis of
sphingosine serine is condensed with palmitoyl-CoA
and decarboxylated to form an aminoketone interme-
diate (Fig. 21-6).
219a
8-Amino-7-oxonanonoate synthase
forms a precursor of biotin (see banner, p. 719).
220
Ketimine intermediate as electron acceptor
(Group d). The fourth group of PLP-dependent reac-
tions are thought to depend upon formation of the
ketimine intermediate of Eq. 14-28. In this form the
original -hydrogen of the amino acid has been
removed and the C = NH
+
bond of the ketimine is
polarized in a direction that favors electron with-
drawal from the amino acid into the imine group.
This permits another series of enzymatic reactions
analogous to those of the -oxo acid shown at the top
of Fig. 14-5. Both elimination and C C bond cleavage
, to the C = N group of the ketimine can occur.
Enzymes of this group catalyze elimination of
substituents from amino acids as illustrated in Fig.
14-7. Eliminated groups may be replaced by other
substituents, either in the or the positions. The
ketimine formed initially by such an enzyme (step a)
undergoes elimination of the substituent ( with
respect to the C = N group) along with a proton from
the position of the original amino acid to form an

OOC
C
S
CoA
O
HCH
N
HC
Quinonoid-carbanionic
intermediate
Glycine
PLP CO
2
CoA-SH
Succinyl-CoA

OOC
C
O
CH
2
NH
2
-Aminolevulinate
PLP

OOC
C
O
C
N
HC
H
H
Schiff base
(14-32)
746 Chapter 14. Coenzymes: Natures Special Reagents
unsaturated intermediate which can
react in one of three ways, depending
upon the enzyme. Addition of HY
leads to replacement (step c), while
addition of a proton at the position
leads, via reaction step d, to an ,-
unsaturated Schiff base. The latter can
react by addition of HY ( replace-
ment, step e) or it can break down to an
-oxo acid and ammonium ion (step f),
just as in the elimination reactions of
Eq. 14-29. An important replacement
reaction is conversion of O-acetyl-,
O-succinyl-, or O-phosphohomoserine
to cystathionine (Eq. 14-33). This
cystathionine -synthase reaction
220a
lies on the pathway of biosynthesis of
methionine by bacteria, fungi, and
Cysteine
Methionine

OOC
COO

Succinate S
C
NH
3
+
COO

H
CH
2
C
H
3
+
N
COO

H
Cystathionine

OOC
C
O
C
NH
3
+
O
COO

H
O-Succinylhomoserine
HS
C
NH
3
+
COO

H
Homocysteine
Pyruvate + NH
4
+
Replacement
Elimination
(14-33)
Figure 14-7 Some PLP-dependent reactions involving
elimination of a substituent. Replacement by another
substituent or by a substituent in the position is possible,
as is deamination to an -oxo acid.
H
3
C
COO

N
H
+
H
b
a
EPLP
Ketimine
Elimination
HY
HY
c
f
Schiff base
H
2
O
EPLP NH
4
+
2-Oxobutyrate
HY
Replacement
d
e
Replacement
Y
NH
3
COO

H
+
H
2
C
COO

N
H
2
C H
+
Y COO

H H
N
H
2
C H
+
Y'
NH
3
COO

H
+
H
3
C
COO

NH
3
H
H
Y
+
higher plants. Subsequent reactions include elimina-
tion from cystathionine of homocysteine
220b
which is
then converted to methionine (Eq. 14-33). Threonine is
formed from O-phosphohomoserine via elimina-
tion followed by replacement with HO

, a reaction
catalyzed by threonine synthase (Fig. 24-13).
220c
The loss of a -carboxyl group as CO
2
can also occur
through a ketimine or quinonoid intermediate. For
example, the bacterial aspartate -decarboxylase
221
converts aspartate to alanine and CO
2
. Selenocysteine
is utilized to create the active sites of several enzymes
(Chapter 15). Excess selenocysteine is degraded by the
PLP-dependent selenocysteine lyase,
222223b
which
evidently eliminates elemental selenium from a ket-
imine or quinonoid state of an intermediate Schiff base
(Eq. 14-34). A similar reaction may occur in the bio-
synthesis of ironsulfur clusters. The Nif S protein is
essential for formation of Fe
4
S
4
clusters in the nitrogen-
fixing enzyme nitrogenase. This enzyme is in some
way involved in transferring the sulfur atom of cysteine
into an ironsulfur cluster.
224
Alanine is the other
product suggesting transfer of S
0
into the cluster using
the sequence of Eq. 14-34.
Another related reaction that goes through a ket-
imine is the conversion of the amino acid kynurenine
to alanine and anthranilic acid.
225
It presumably depends
upon hydration of the carbonyl group prior to cleav-
age (Eq. 14-35). An analogous thiolytic cleavage utilizes
CoA to convert 2-amino-4-ketopentanoate to acetyl-
CoA and alanine.
226
Glycogen phosphorylase. While PLP is ideally
designed to catalyze reactions of amino compounds
it was surprising to find it as an essential cofactor for
glycogen phosphorylase (Fig. 11-5). The PLP is linked
as a Schiff base in the same way as in other PLP-depen-
747
NH
2
C
O
CH
2
C
COO

NH
3
+
H
C CH
2
C
COO

N
OH O
H
Kynurenine
H
2
O, PLP
COO

NH
3
+
Ketimine
+
Alanine
H
Anthranilic acid
(14-35)
NH
3
+
COO

CH
2
H

Se
H
2
C COO

N
H
2
C H
H
+
H
3
C COO

N
H
2
C
+
H
Selenocysteine
Ketimine
Se
0
Elemental selenium
CH
2
COO

N
H
2
C

Se
H
+
EPLP
EPLP
l-Alanine
(14-34)
dent enzymes, but there is no obvious function for the
coenzyme ring. As suggested in Chapter 12, the phos-
phate group probably acts as an acidbase catalyst. It
has been estimated that 50% of the vitamin B
6
in our
body is present as PLP in muscle phosphorylase.
227
Studies of vitamin B
6
-deficient rats suggest that PLP
in phosphorylase serves as a reserve supply, much of
which can be taken for other purposes during times
of deficiency.
4. Pyridoxamine Phosphate as a Coenzyme
If PLP is a cofactor designed to react with amino
groups of substrates, might not pyridoxamine phosphate
(PMP) act as a coenzyme for reactions of carbonyl
compounds? An example of this kind of function has
been found
228 230
in the formation of 3,6-dideoxyhexoses
needed for bacterial cell surface antigens (Fig. 4-15).
Glucose (as cytidine diphosphate glucose; CDP-glu-
cose) is first converted to 4-oxo-6-deoxy-CDP-glucose.
The conversion of the latter to 3,6-dideoxy-CDP-glucose
(Eq. 14-36) requires PMP as well as NADH or NADPH.
The student may find it of interest to propose a
mechanism for this reaction, taking into account the
expected direct transfer of a hydrogen from NADH as
described in Chapter 15, before consulting published
papers. Part of the reaction cycle appears to involve a
free radical derived from the PMP. This is discussed
further in Chapter 20 together with free radical-form-
ing PLP enzymes
5. Stereochemistry of PLP-Requiring Enzymes
According to stereoelectronic principles, the bond
in the substrate amino acid that is to be broken by a
PLP-dependent enzyme should lie in a plane perpen-
dicular to the plane of the cofactorimine system
(Fig. 14-8). This would minimize the energy of the
transition state by allowing maximum overlap
between the breaking bond and the ringimine system.
It also would provide the geometry closest to that of
the planar quinonoid intermediate to be formed, thus
minimizing molecular motion in the approach to the
transition state. Figure 14-8 shows three orientations
of an amino acid in which the -hydrogen, the carboxyl
group, and the side chain, respectively, are positioned
for cleavage. For each orientation shown, another
geometry suitable for cleavage of the same bond is
E. Pyridoxal Phosphate
O
HO
CH
3
OH
O
O
P P Cytidine
O
H
CH
3
O
O
P P Cytidine
NADH + H
+
NADH
+
H
PMP
(14-36)
748 Chapter 14. Coenzymes: Natures Special Reagents
obtained by rotating the amino acid through 180.
Dunathan suggested that this stereoelectronic
requirement explains certain side reactions observed
with PLP-requiring enzymes. The idea also received
support from experiments with a bacterial -dialkyl-
glycinedecarboxylase.
231,232
The enzyme ordinarily
catalyzes, as one half-reaction, the combination decarb-
oxylationtransamination reaction shown in Eq. 14-37.
It also acts on both D- and L-alanine, decarboxylating
the former but catalyzing only removal of the -H
from L-alanine. The results can be rationalized by
assuming that the enzyme possesses a definite site for
one alkyl group but that the position of the second
alkyl group can be occupied by H or COO

and that
the group labilized lies perpendicular to the system:
Glycine is unreactive, suggesting that occupation of
the alkyl binding site is required for catalysis.
According to Dunathans postulate, there are only
two possible orientations of the amino acid substrate
in an aminotransferase. One is shown in Fig. 14-8.
In the other, the amino acid is rotated 180 so that the
-hydrogen protrudes behind the plane of the paper.
Dunathan studied pyridoxamine:pyruvate amino-
transferase, an enzyme closely related to PLP-requir-
ing aminotransferases and which catalyzes the trans-
amination of pyridoxal with L-alanine to form pyridox-
amine and pyruvate. The same reaction is catalyzed
by the apoenzyme of aspartate aminotransferase. In
both cases, when the alanine contained
2
H in the
position the
2
H was transferred stereospecifically into
C
NH
3
+
CH
3
H
3
C
COO

C
O
CH
3
H
3
C
CO
2
PLP
PMP
(14-37)
C
N
CH
3
H
3
C
COO

C
N
CH
3

OOC
H
C
N
CH
3
H
COO

Alkyl
binding
site
l-Alanine d-Alanine
Figure 14-8 Some stereochemical aspects of catalysis of PLP-requiring enzymes.
N
C
N
H

O
H*
R

OOC
H
+
COO

H R
C
COO

H
H O
H
H
H*
COO

H
NH
2
In each case the bond to be broken lies perpendicular to system of Schiff base
a
-Hydrogen is
positioned for
removal as H
+
,
e.g., in an
aminotransferase
Addition of
proton from
si-face
(toward
reader)
b
COO

is positioned
for decarboxylation
D
2
O
CO
2
Deuterium
enters
pro-R
position
c
Side chain is positioned
for aldol cleavage
CH
2
O
(transferred to
tetrahydrofolic
acid)
Tritium-
containing
position
is pro-S
3
H
2
O
tritiated
H
2
O
D
H R
NH
2
N
C
N
H

O
R

OOC
+
H*
H
pro-S
H
+
H
+
749
the pro-S position at C-4' of the pyridoxam-
ine (indicated by asterisks in Fig. 14-8).
233
The results suggested that a group from
the protein abstracts a proton from the
position and transfers it on the same side
of the system (suprafacial transfer),
adding it to the si face of the C = N group
as shown in Fig. 14-8. Later, the same
stereospecific proton transfer was demon-
strated for the PLP present in the holoen-
zyme.
234
Not surprisingly, the D-amino
acid aminotransferase adds the proton to
the re face of the C = N group.
235
When a decarboxylase acts on an
amino acid in
2
H
2
O, an atom of
2
H is incorporated in
the pro-R position, the position originally occupied by
the carboxyl group (Fig. 14-8). Cleavage of serine by
serine hydroxymethyltransferase in
3
H-containing
water leads to incorporation of
3
H in the pro-S position.
Stereospecific introduction of
2
H or
3
H has been ob-
served in the position of 2-oxobutyrate formed in
or elimination reactions. Conversion of serine to
tryptophan by tryptophan synthetase occurs without
inversion at C-3.
236
These and many other observations
on PLP-dependent enzymes
189,237,238
can be generalized
by saying that enzymatic reactions of PLP Schiff bases
usually take place on only one face of the relatively
planar structure. This is the si face at C-4' of the coen-
zyme (see Fig. 14-8). This result is expected if a single
acidbase group serves as proton acceptor in one step
and as proton donor in a later step. This leads naturally
to the observed retention of configuration in steps
involving replacement and the suprafacial transfer of
protons from one position on that face to another.
6. Seeing Changes in the Optical Properties
of the Coenzyme
The absorption of light in the ultraviolet and visible
regions is a striking characteristic of many coenzymes.
It can be measured accurately and displayed as an
absorption spectrum and may also give rise to circular
dichroism and to fluorescence (Chapter 23). The optical
properties of the vitamin B
6
coenzymes are sensitive to
changes both in environment and in the state of proto-
nation of groups in the molecule. For example, PMP
in the neutral dipolar ionic form, which exists at pH 7,
has three strong light absorption bands centered at
327, 253, and 217 nm.
239
The other ionic forms of PMP
and other derivatives of vitamin B
6
also each have
three absorption bands spaced at roughly similar
intervals, but with varying positions and intensities.
The minor tautomer of PMP containing an uncharged
ring (Eq. 14-38) has its low-energy (long-wavelength)
band at 283 nm. When both the ring nitrogen and
phenolic oxygen are protonated, the band shifts again
to 294 nm and if both groups are deprotonated the
resulting anion absorbs at 312 nm. Thus, observation
of the absorption spectrum of the coenzyme bound to
an enzyme surface can tell us whether particular groups
are protonated or unprotonated. The peak of bound
PMP at 330 nm in aspartate aminotransferase (Fig. 14-9)
is indicative of the dipolar ionic form (Eq. 14-38). How-
ever, the 5-nm shift from the position of free PMP sug-
gests a distinct change in environment.
E. Pyridoxal Phosphate
(14-38)
N
2
O
3
PO
CH
2
NH
3
+
O

CH
3
H
+
Dipolar ionic form
I
II
III
327 nm
253 nm
217 nm
N
2
O
3
PO
CH
2
NH
3
+
OH
CH
3
Minor non-dipolar ionic tautomer
I 283 nm
Figure 14-9 Absorption spectra of various forms of aspar-
tate aminotransferase compared with that of free pyridoxal
phosphate. The low pH form of the enzyme observed at pH
< 5 is converted to the high pH form with pK
a
~ 6.3. Addi-
tion of erythro-3-hydroxyaspartate produces a quinonoid
form whose spectrum here is shown only 1/3 its true height.
The spectrum of free PLP at pH 8.3 is also shown . The
spectrum of the apoenzyme (- - -) contains a small amount of
residual absorption of uncertain origin in the 300- to 400-nm
region.
Wave number, cm
1
10
-3
Wavelength, nm
18 22 26 30 34
500 400 300
10
8
6
4
2
0
M
o
l
a
r

e
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t
,


1
0
-
3
Strong
absorption
bands of
aromatic
amino acids
+
e
r
y
t
h
r
o
-

-
O
H
-
A
s
p
5
0
0

n
m
4
3
0



L
o
w

p
H
3
9
0



F
r
e
e

P
L
P



p
H

8
.
3
3
6
3



H
i
g
h

p
H
3
3
0



P
M
P

f
o
r
m 12
750 Chapter 14. Coenzymes: Natures Special Reagents
Pyridoxal phosphate exists in an equilibrium
between the aldehyde and its covalent hydrate (as in
Eq. 13-1). The aldehyde has a yellow color and absorbs
at 390 nm (Fig. 14-9), while the hydrate absorbs at nearly
the same position as does PMP. The absorption bands
of Schiff bases of PLP are shifted even further to longer
wavelengths, with N-protonated forms absorbing at
415 430 nm. Forms with an unprotonated C = N
group absorb at shorter wavelengths.
149,240
Imine groups in free enzymes. When, in 1957,
W. T. Jenkins examined one of the first highly purified
aspartate aminotransferase preparations he noted a
surprising fact: The bound coenzyme, at pH 5, absorbed
not at 390 nm, as does PLP, but at 430 nm, like a Schiff
base (Fig. 14-9). When the pH was raised, the absorp-
tion band shifted to 363 nm. The result suggests disso-
ciation of a proton (with a pK
a
of ~ 6.3) from the
hydrogen-bonded position in a Schiff base of the type
shown in Fig. 14-6. It was quickly demonstrated for
this enzyme and for many other PLP-dependent en-
zymes that reduction with sodium borohydride caused
the spectrum to revert to one similar to that of PMP
and fixed the coenzyme to the protein. After complete
HCl digestion of such borohydride-reduced proteins a
fluorescent amino acid containing the reduced pyridoxyl
group was obtained and in every case was identified as
-pyridoxyl-lysine. Thus, PLP-containing enzymes in
the absence of substrates usually exist as Schiff bases
with lysine side chains of the proteins. Even the PLP
in glycogen phosphorylase is joined in this way.
However, its absorption maximum at 330 nm shows
that in phosphorylase it is present as the nondipolar
ionic tautomer with a 3-OH group on the ring as in
Eq. 14-38.
Absorption bands at 500 nm. With many PLP
enzymes certain substrates and inhibitors cause the
appearance of intense and unusually narrow bands
at ~ 500 nm. Such a band is observed with aspartate
aminotransferases acting on erythro-3-hydroxyaspartate
(Fig. 14-9). This substrate undergoes transamination
very slowly, and the 500-nm absorbing form which
accumulates is probably an intermediate in the normal
reaction sequence. A similar spectrum is produced
by tryptophan indole-lyase acting on the competitive
inhibitor L-alanine. Under the same conditions the
enzyme promotes a rapid exchange of the -hydrogen
of the alanine with
2
H of
2
H
2
O. Serine hydroxymethyl-
transferase gives a 495 - to 500-nm band with both
D-alanine and the normal product glycine.
241
Similar
spectra have been produced in nonenzymatic model
reactions
242
and probably represent the postulated
quinonoidcarbanionic intermediates.
7. Atomic Structures
The three-dimensional structures of aspartate
aminotransferases from E. coli to humans are very
similar.
163167a
The folding pattern (Fig. 2-6) and active
site structure (Figs. 14-6 and 14-10) are completely
conserved. The major domain of the protein contains
a central sheet surrounded by helices with coenzyme
attached to a lysine at the C terminus of one of the
strands. The protein is a dimer with the two major
domains held together by both polar and nonpolar
interactions. The two active sites are located at the
interface between the subunits and residues from both
subunits participate in forming the active site (Fig. 14-10).
The internal Schiff base is formed with Lys 258. The
protonated ring nitrogen of the dipolar ionic PLP
forms an ion pair with the carboxylate of Asp 222
which protrudes from a central seven-stranded sheet.
The phenolic O

forms a hydrogen bond with the


OH of Tyr 225. The interactions of Asp 222 and Tyr
225 fix the ring as the dipolar ionic tautomer.
The phosphate group of the coenzyme, which
31
P
NMR shows to be predominantly dianionic,
243
forms
an ion pair with the side chain of Arg 266 and hydrogen
bonds to a backbone N H at the N terminus of a long
helix where it can interact with the positive end of the
helix dipole. In addition, the phosphate forms hydro-
gen bonds to four OH groups of Ser, Thr, and Tyr side
chains. In front of the coenzyme ring are two guani-
dinium groups from the side chains of Arg 386 and of
Arg 292* (from the second subunit). These have been
shown by X-ray crystallography to bind the two car-
boxylate groups of a substrate such as glutamate,
2-oxoglutarate, aspartate, oxaloacetate, or cysteine
sulfinate; of quasi-substrates such as 2-methylaspartate
and erythro--hydroxyaspartate; or of dicarboxylic
inhibitors (Fig. 14-10).
Comparison of amino acid sequences suggests
that many other PLP-dependent enzymes have folding
patterns similar to those of aspartate aminotransferase
but that there are four or more additional different
folding patterns.
205,243a,b
Among the enzymes resem-
bling aspartate aminotransferase are -amino acid:
pyruvate aminotransferase,
244
2,3-dialkylglycine decar-
boxylase,
232
tyrosine phenol-lyase,
177
a bacterial orni-
thine decarboxylase,
182,204
and cystathionine -lyase.
182
The tryptophan synthase subunit has a second folding
pattern,
184
while alanine racemase
153
and eukaryotic
H
+
N
CH
2
OH
H
2
C
O

H
3
C
N
C
COO

NH
3
+
H
H
-Pyridoxyllysine
751
ornithine decarboxylases
205
have ()
8
-barrel structures
resembling that in Fig. 2-28. A fourth structural pattern
is that of D-amino acid aminotransferase.
235
It is anti-
cipated that the branched-chain aminotransferase
245
will have a similar stucture. Glycogen phosphorylase
(Fig 11-5) has a fifth folding pattern.
8. Constructing a Detailed Picture of the
Action of a PLP Enzyme
Consider the number of different steps that must
occur in about one-thousanths of a second during the
action of an aminotransferase. First, the substrate
binds to form the Michaelis complex. Then the
transimination (Eq. 14-26) takes place in two steps and
is followed by the removal of the -hydrogen to form
the quinonoid intermediate. An additional four steps
are needed to form the ketimine, to hydrolyze it, and
to release the oxoacid product to give the PMP form
of the enzyme. The reaction sequences in some of the
other enzymes are even more complex. How can one
enzyme do all this?
The first step in the sequence is the binding of the
substrate to form the Michaelis complex. The posi-
tive charges on Arg 386 and Arg 292 doubtless attract
the carboxylate groups of the substrate and aid in
guiding it toward a correct fit. In a similar manner the
O

of the coenzyme, which is distributed by resonance


into the C = N of the Schiff base linkage, attracts the
NH
3
+
of the substrate. When a substrate or inhibitor
binds to the two guanidinium groups a small structural
domain of the enzyme moves and closes around the
substrate which now has very little contact with the
external solvent. In the initial Michaelis complex
the NH
3
+
group of the substrate lies directly in front
of the C-4' carbon of the coenzyme (Fig. 14-10), where
it can initiate the transimination reaction of Eq. 14-26.
However, before this can happen a proton must be
removed to convert the NH
3
+
to NH
2
.
Long before the three-dimensional structures were
known, Ivanov and Karpeisky
246
suggested that in the
free enzyme the positively charged group (Arg 386)
that binds the -carboxylate of the substrate interacts
electrostatically with the O

of the coenzyme. This is


one of the factors that keeps the pK
a
of the CH=N
+
H
that is conjugated with this O

at a low value of
~ 6.3 (at 0.1 M anion concentration). However, in the
Michaelis complex this interaction of the + charge of
Arg 386 with the imine group must be weakened
because of the pairing of the COO

of the substrate
with the + charge. This will increase the basicity of the
imine nitrogen and will also cause a decrease in the
pK
a
of the substrate NH
3
+
, making is easier for a
proton to jump from the NH
3
+
to the imine group.
This proton transfer is shown in Eq. 14-39, step a.
Thus, the nucleophilic NH
2
group is generated by a
process that at the same time increases the electrophilic
properties of the carbon atom of the imine group. This
favors the immediate addition of NH
2
to C = N
+
H to
give the adduct a geminal diamine (Eq. 14-39, step b),
which is shown in three dimensions in Fig. 14-10B.
Notice that each step in the overall sequence
changes the electronic or steric characteristics of the
complex in a way that facilitates the next step.
246
This
is an important principle that is applicable throughout
enzymology: For an enzyme to be an efficient catalyst
each step must lead to a change that sets the stage for the
next. These consecutive steps often require proton
transfers, and each such transfer will influence the
subsequent step in the sequence. Some steps also
require alterations in the conformation of substrate,
coenzyme, and enzyme. One of these is the transimi-
nation sequence (Eqs. 14-26, 14-39). On the basis of
the observed loss of circular dichroism in the external
aldimine, Ivanov and Karpeisky suggested that a
E. Pyridoxal Phosphate
(14-39)
C
b
O
N
H
+
O

N
H
2
C
CH
3
2
O
3
P
Michaelis complex
K258
O
N
H
+
O

N
CH
3
2
O
3
P
O
O C
C
C
H
After proton transfer
K258
a
+
+
:
HN
Geminal diamine
K258
H
H
2
C
O
O

H
N
H
H
N
H
N
H
R386
+
O
O C
C
C
O
H
O

H
N
H
H
N
H
N
H
R386
NH
3
+
NH
2
C
C
H
O
O

H
N
H
H
N
H
N
H
R386
+ O
O
+
O
N
H
O

CH
3
2
O
3
P
H
C
N
+
H
H
752 Chapter 14. Coenzymes: Natures Special Reagents
A
B
C
rotation of the coenzyme occurs as the NH
2
of the
substrate adds to the C = N bond during transimination
(Eq. 14-39, step b). This accomplishes the essential
shortening by ~ 0.15 nm of the distance between C-4'
and the N atom from a van der Waals contact distance
to a covalent bond distance while the carboxylate
groups of the substrate remain
bound in their initial positions.
That the coenzyme really
does change its orientation was
suggested by a dramatic change
in the absorption spectrum of
a crystal recorded with plane
polarized light (linear dichroism)
when 2-methylaspartate was
soaked into a crystal (Fig. 23-9).
247
X-ray crystallography confirmed
rotation of the ring by ~30.
163,242,247
To complete the transimination
sequence, which is shown only
partially by Eq. 14-39, another
proton transfer is needed to move
the positive charge on the sub-
strate N
+
H
2
to that of lysine
258, whose amino group is then
eliminated. This requires addi-
tional tilting of the ring. The
crystal structure of the external
aldimine with -methylaspartate
has been determined
248
as have
those of ketimines with glutamate
and aspartate,
249,250
a carbinol-
amine,
251
and quinonoid com-
plexes of related enzymes.
212a
The -amino group, which is
eliminated in Eq. 14-26, is basic
and functions in the next several
steps of catalysis, including the
abstraction of the -hydrogen
and in its transfer to the 4'-carbon
(Eq. 14-28). This amino group
can be seen in Fig. 14-10C, where
it is positioned beside a water
molecule that is needed to hydro-
lyze the ketimine. From this
figure it can be seen that the
group is able to move from site
to site on one side of the planar
external aldimine, quinonoid
carbanionic, and nearly planar
ketimine forms. If the Lys 258
amino group is the catalytic base
for these reactions it must be
present at pH 7. What is its pK
a
?
And why doesnt this pK
a
show
up in the plot of V
max
vs pH as in
Fig 9-9? In fact, the maximum
velocity of aspartate aminotransferase and many other
PLP-dependent enzymes is independent of pH over a
broad range. The answer is probably that the pK
a
s of
both the NH
2
groups of Lys 258 and the amino acid in
the Michalis complex are high when there is no posi-
tive charge on the adjacent Schiff base C = N but
Figure 14-10 Models of catalytic intermediates for aspartate aminotransferase in a
half-transamination reaction from aspartate to oxalocetate. For clarity, only a selec-
tion of the active site groups are shown. (A) Michaelis complex of PLP enzyme with
aspartate. (B) Geminal diamine. (C) Ketimine intermediate. The circle indicates a
bound water molecule. See Jansonius and Vincent in Jurnak and McPherson.
163
Courtesy of J.N. Jansonius.
753
act as built-in sensors able to detect small changes in
the electronic environment. For example, when the
Schiff base proton dissociates around the pK
a
of ~ 6.2
the NMR resonance of H
a
shifts upfield from 17.2 to
15.2 ppm as a result of donation of electons into the
ring from the O

of the coenzyme.
169
This shift illus-
trates the reality of the strong electrostatic forces that
operate across heterocyclic aromatic rings within
active sites of proteins. In alanine racemase a different
histidine cluster is present beneath the active site and
constitutes part of the solvent in which the catalyzed
reaction takes place. At least in the case of aspartate
aminotransferase, none of the histidines are absolutely
essential for activity but the hydrogen-bonded network,
which can be altered in mutant forms, may be important.
The detailed description of a reaction sequence
given here has to be altered for each specific enzyme.
A vast amount of work, only a little of which is cited
here, has been done on PLP enzymes.
253 254a
These
studies involve calorimetry,
255
kinetics,
210,256 259
crystal-
lography, optical spectroscopy,
188,260,261
NMR,
243,262 264
and genetic engineering and chemical modifica-
tion.
178,185,186,265
F. Pyruvoyl Groups and Other Unusual
Electrophilic Centers
A few enzymes that might be expected to have
PLP at their active sites have instead a prosthetic
group consisting of pyruvic acid bound by an amide
linkage, a pyruvoyl group (Table 14-4). These and
several apparently related enzymes are the subject of
this section.
very low when the Schiff base is protonated ( C =
NH
+
). This is a result of the large electrostatic effect
of a closely adjacent charge in a medium of low dielec-
tric constant (see Chapter 7, Section A). When the
Schiff base is unprotonated, as in the first structure of
Eq. 14-39, the adjacent amino group has a high pK
a
and
is mostly protonated. However, the proton can jump
as in step a of that equation to give a protonated Schiff
base. Now the unprotonated NH
2
group has a very
low pK
a
, but it is still strongly nucleophilic and can
readily add to the Schiff base double bond in step b of
Eq. 14-39. In a similar fashion the amino group of Lys
258 will alternately be unprotonated and then proto-
nated, its microscopic pK
a
alternating between low
and high, as it catalyzes the steps of abstracting the -
hydrogen and forming and hydrolyzing the ketimine.
Only two microscopic pK
a
values, one very high and
one very low appear in the V
max
vs pH profile. This
alternation of microscopic pK
a
values may be a com-
mon characteristic of enzymes that bind ionized sub-
strates and can make good use of the strong
electrostatic effects that arise in the active sites to
facilitate essential proton transfers.
Aspartate aminotransferases are distinguished
from most other PLP-dependent enzymes including
transaminases by the relatively low pK
a
of ~ 6.3 for the
free enzyme. The unprotonated Schiff base in the free
enzyme can then react with the protonated amino
group of the substrate. How can other PLP enzymes
react with amino acids when they have protonated
Schiff bases even at relatively high pH? A logical answer
is that some basic group with a low pK
a
is close to the
Schiff base and acts to deprotonate the substrate NH
3
+
so that transimination can occur. Clausen et al. sug-
gested that in cystathionine -lyase this is probably
tyrosine (Y111), which is adjacent to the Schiff base
C = NH
+
of the external aldimine and is thought to
be ionized in the active enzyme.
182
NMR evidence
suggests that protonation of an adjacent catalytic base
occurs upon substrate binding in D-serine dehydratase
as well.
252
Many other variations in active site environ-
ments are seen among PLP-dependent enzymes. As
with aspartate aminotransferases, there is often an
essential carboxylate group that holds a proton onto
the pyridine ring of the coenzyme. However, in alanine
racemase a guanidinium group from arginine is hydro-
gen bonded to the coenzyme ring.
153
Below the active site of aspartate aminotransferase,
as shown in Fig. 14-6, is a cluster of three buried his-
tidine side chains in close contact with each other. The
imidazole of H143 is hydrogen bonded to the D222
carboxylate, the same carboxylate that forms an ion
pair with the coenzyme. This system looks somewhat
like the catalytic triad of the serine proteases in reverse.
As with the serine proteases, the proton-labeled H
b
in
Fig. 14-6 can be seen by NMR spectroscopy (Fig. 3-30).
So can the proton H
a
on the PLP ring. These protons
Decarboxylases Product
Histidine (bacterial)
S-Adenosylmethionine
Aspartate - decarboxylase -Alanine
Phosphatidylserine Phosphatidyl-
ethanolamine
4' - Phosphopantothenylcysteine 4' - Phosphopan-
tetheine
Reductases (clostridial)
Proline
Glycine
TABLE 14-4
Some Pyruvoyl Enzymes
Adapted from van Peolje and Snell.
267
F. Pyruvoyl Groups and Other Unusual Electrophilic Centers
754 Chapter 14. Coenzymes: Natures Special Reagents
1. Decarboxylases
Mammalian histidine decarboxylase contains
PLP but the enzyme from many bacteria contains a
pyruvoyl group, as do a few other decarboxylases
both of bacterial and eukaryotic origin.
266,267
These
enzymes are inhibited by carbonyl reagents and by
borohydride. When
3
H-containing borohydride was
used to reduce the histidine decarboxylase of Lactoba-
cillus,
3
H was incorporated and was recovered in lactic
acid following hydrolysis. This suggested the presence
of a pyruvoyl group attached by an amide linkage and
undergoing the chemical reactions that are shown in
Eq. 14-40. Reduction in the presence of the substrate
histidine resulted in covalent binding of the histidine
to the bound pyruvate. Thus, as with the PLP-contain-
ing decarboxylases, a Schiff base is formed with the
substrate. Decarboxylation is presumably accomplished
by using the electron-attracting properties of the car-
bonyl group of the amide:
carry the N-terminal pyruvoyl group, which was formed
from Ser 82. The activation occurs spontaneously by
incubations of the proenzyme for 24 48 h at pH ~ 7 in
the presence of divalent metal ions.
Substituted serine residues under mildly alkaline
conditions readily undergo , elimination to form
dehydroalanine residues. When prohistidine decarb-
oxylase containing
18
O in its serine side chains was
activated
18
O was found in the carboxylate group of
Ser 81 of the chains. It was shown that it had been
transferred from the side chain of Ser 82. This suggested
the formation of an intermediate oxygen ester of Ser 81
during formation of the pyruvoyl group (Eq. 14-41).
272
S-Adenosylmethionine decarboxylase is the
first enzyme in the biosynthetic pathway to spermi-
dine (Chapter 24). Whether isolated from bacteria,
yeast, animals, or other eukaryotes, this enzyme always
contains a bound pyruvoyl group.
273274b
Both the
(14-40)
O C
C
N
CH
3
NH
Enzyme
C
C
H
CH
3
O

O
N
N
H
Bound pyruvate
Histidine
combined as
Schiff base
O
C
C
NH
CH
3
O
Enzyme
C
C
NH
CH
3
O
Enzyme
3
H
HO
C
CH
3
Pyruvoyl
group
3
H HO
NaBH
4

OOC
Acid
hydrolysis
Lactate
H
3
+
N-Enzyme
When
14
C-labeled serine was fed to organisms
producing histidine decarboxylase,
14
C was incorporated
into the bound pyruvoyl group (Fig. 14-11). Thus,
serine is a precursor of the bound pyruvate. The
enzyme is manufactured in the cell as a longer 307-
residue proenzyme which associates as hexamers
(designated
6
). The active enzyme was found to be
formed by cleavage of the chains between Ser 81 and
Ser 82 to form 226-residue chains and 81-residue
chains which associate as ()
6
.
270,271
The chains
N
C
C
N
C
C
N
H O
O H
H
2
C H
H
CH
2
O
H
OH
N
C
C
N
C
C
N
H
O
H
2
C H
H
OH
O CH
2
O
N
C
C
O
CH
2
C
O
H
2
C H
OH
C
O
N
H
H
2
N
H
N
C
COO

H
2
C H
OH
C C
O
N
H
H
H
2
N
H
2
C
82
C
H
2
C
O
N
H
Dehydroalanine
H
+
H
O
H
3
C
81
H
Pyruvoyl group
Tautomerization
and hydrolysis
NH
4
+
CH
2
CH
2
CH
2
H
2
C
H
3
C
(14-41)
755
Figure 14-11 Schematic diagram of the active site of the
pyruvoyl enzyme histidine decarboxylase showing key polar
interactions between the pyruvoyl group and groups of the
inhibitor O-methylhistidine and surrounding enzyme groups.
Aspartate 63 appears to form an ion pair with the imidazolium
group of the substrate.
268
Hydrogen bonds are indicated by
dotted lines. See Gallagher et al.
269
E197
K155
NH
3
+
O(H)
O
O
O
O
O
O
O
O
O
O
O
O
D63
Y62
E66
O
F195
N H
CH
3
S81
N76
H
2
N
Prv
O oxygen
nitrogen
carbon
mammalian enzyme and that from E. coli are ()
4
tetramers formed in a manner similar to that of bacte-
rial histidine decarboxylase. Other pyruvoyl decarbo-
xylases are phosphatidylserine decarboxylase,
275,276
an intrinsic membrane protein used to form phospha-
tidylethanolamine, aspartate -decarboxylase,
277
which forms alanine needed for biosynthesis of
coenzyme A, and 4'-phosphopantethenoylcysteine
decarboxylase,
278
the second of two decarboxylases
required in the synthesis of coenzyme A. Because of
the lack of a primary amino group, its mechanism
must be somewhat different from that of other en-
zymes in this group.
267
2. Proline and Glycine Reductases
An enzyme required in the anaerobic breakdown
of proline by clostridia utilizes a dithiol-containing
protein to reductively open the ring (Eq. 14-42).
279,280
(14-43)
N
COO

H
H
3
+
N
COO

2
H H
H
2
+
S S
protein
SH SH
protein
2
H
2
O
(14-42)
This enzyme also contains an N-terminal pyruvoyl
residue as does one subunit of a selenium-containing
glycine reductase which utilizes a dithiol to convert
glycine into acetate with coupled formation of ATP:
Glycine + ADP + P
i
+ 2 e

acetate
-
+ NH
4
+
+ ATP
In both cases it has been proposed that the pyruvoyl
group forms a positively charged Schiff base with the
substrate. The bond-breaking mechanisms are not
obvious but some ideas have been proposed.
281 283a
The ATP may arise from reaction with an intermediate
acetyl phosphate.
282
See Eq. 15-61.
3. Dehydroalanine and Histidine and
Phenylalanine Ammonia-Lyases
Catabolism of histidine in most organisms proceeds
via an initial elimination of NH
3
to form urocanic acid
(Eq. 14-44). The absence of the enzyme L-histidine
ammonia-lyase (histidase) causes the genetic disease
histidinemia.
284,285
A similar reaction is catalyzed by the
important plant enzyme L-phenylalanine ammonia-
lyase. It eliminates NH
3
+
along with the pro-S
hydrogen in the position of phenylalanine to form
trans-cinnamate (Eq. 14-45). Tyrosine is converted to
p-coumarate by the same enzyme. Cinnamate and
coumarate are formed in higher plants and are con-
verted into a vast array of derivatives (Box 21-E,
Fig. 25-8).
The reactions of Eqs. 14-44 and 14-45 are unusual
because the nucleophilic substituent eliminated is on
the -carbon atom rather than the . There is nothing
in the structures of the substrates that would permit an
N
NH

OOC
H
3
N H
H
S
H
R
+
N
NH

OOC
5'
H
+
NH
4
+
Urocanate

OOC
H
3
N H
H
R
H
S
+

OOC
trans-cinnamate
H
+
NH
4
+
(14-44)
(14-45)
F. Pyruvoyl Groups and Other Unusual Electrophilic Centers
756 Chapter 14. Coenzymes: Natures Special Reagents
easy elimination of the -amino group. Thus, it
should not be surprising that both enzymes contain a
special active center. When
2
H is introduced into the
position of phenylalanine, no isotope effect on the rate
is observed. Rather, the rate-limiting step appears to
be the release of ammonia from the coenzyme group.
It appears that the enzyme must in some way make
the amino group a much better leaving group than it
would be otherwise.
F. Pyruvoyl Groups and Other Unusual Electrophillic Centers
BOX 14-D DIETARY REQUIREMENTS FOR B VITAMINS
It is difficult to determine the amounts of vita-
mins needed for good health and they may differ
considerably from one individual to another. The
quantities listed below are probably adequate for
most young adults but must be increased during
pregnancy and lactation and after very strenuous
exercise.
Pantothenic acid: 10 15 mg/day. Deficiency
causes apathy, depression, impaired adrenal function,
and muscular weakness. -Methylpantothenic acid
is a specific antagonist. The calcium salt, calcium
pantothenate, is the usual commercial form.
Biotin: 0.15 0.3 mg/day. The discovery that
biotin deficiency in young chickens can lead to
sudden death resulted in a recommendation to
supplement infant formulations with biotin.
a
Desthiobiotin, in which the sulfur has been re-
moved and replaced by two hydrogen atoms, can
replace biotin in some organisms and appears to lie
on one pathway of biosynthesis.
b,c
Oxybiotin, in
which the sulfur has been replaced by oxygen, is
active for many organisms and partially active for
others. No evidence for conversion to biotin itself
has been reported, and oxybiotin may function
satisfactorily in at least some enzymes.
Thiamin: 0.23 mg or more per 1000 kcal of food
consumed and a minimum total of 0.8 mg/day.
Replacement of the methyl group on the pyrimidine
ring by ethyl, propyl, or isopropyl gives compounds
with some vitamin activity, but replacement by hydro-
gen cuts activity to 5% of the original. The butyl
analog is a competitive inhibitor.
Vitamin B
6
: 1.5 2 mg/day; 0.4 mg/day for
infants. Vitamin B
6
is widely distributed in foods,
and symptoms of severe deficiency are seldom
observed. However, a number of cases of convul-
sions have been attributed to partial destruction of
vitamin B
6
in infant liquid milk formulas. Convul-
sions occurred when the vitamin B
6
content was
reduced to about one-half that normally present in
human milk.
Several cases of children with an abnormally
high vitamin B
6
requirement (2 10 mg/day) have
been reported, and rare metabolic diseases are
known
df
in which specific enzymes, such as cysta-
thionine synthase, have a reduced affinity for PLP.
Patients with these diseases also benefit from a higher
than normal intake of the vitamin. Excessive excre-
tion of the vitamin may also occur, an example
being provided by a strain of laboratory mice that
require twice the normal amount of vitamin B
6
and
which die in convulsions after a brief period of
vitamin B
6
depletion.
d
Dietary supplementation
with large amounts of vitamin B
6
for treatment of
medical conditions such as carpal tunnel syndrome
has been controversial.
g
Amounts of pyridoxine
over 50 mg per day may damage peripheral nerves,
probably because of the chemical reactivity of pyri-
doxal and PLP.
Nicotinamide: About 7.5 mg/day. Tryptophan
can substitute to some extent. See also Box 15-A.
Riboflavin: About 1.5 mg/day. See also Box 15-B.
Folic acid: About 0.2 0.4 mg/day. See also
Box 15-D.
Vitamin B
12
: About 2 g/day. See also Box
16-B.
Vitamin C (ascorbic acid): 50 200 mg/day.
See also Box 18-D.
a
Parry, R. J., and Kunitni, M. G. (1976) J. Am. Chem. Soc. 98,
40244025
b
Gibson, K. J., Lorimer, G. H., Rendina, A. R., Taylor, W. S.,
Cohen, G., Gatenby, A. A., Payne, W. G., Roe, D. C., Lockett, B.
A., Nudelman, A., Marcovici, D., Nachum, A., Wexler, B. A.,
Marsilii, E. L., Turner, S., IM, Howe, L. D., Kalbach, C. E., and
Chi, H. (1995) Biochemistry 34, 1097610984
c
Huang, W., Jia, J., Gibson, K. J., Taylor, W. S., Rendina, A. R.,
Schneider, G., and Lindqvist, Y. (1995) Biochemistry 34,
1098510995
d
Mudd, S. H., Levy, H. L., and Skovby, F. (1995) in The Metabolic
and Molecular Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver,
C. R., Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp.
12791327, McGraw-Hill, New York
e
Bell, R. R., and Haskell, B. E. (1971) Arch. Biochem. Biophys. 147,
588601
f
Pascal, T. A., Gaull, G. E., Beratis, N. G., Gillam, B. M., Tallan,
H. H., and Hirschhorn, K. (1975) Science 190, 12091211
g
Bender, D. A. (1999) Br. J. Nutr. 81, 720
Both enzymes are inhibited by sodium borohydride
and also by nitromethane. After reduction with NaB
3
H
4
and hydrolysis,
3
H-containing alanine was isolated.
This suggested that they contain dehydroalanine,
which could arise by dehydration of a specific serine
residue.
286,287
For phenylalanine ammonia-lyase from
Pseudomonas putida this active site residue has been
identified as S143. Replacement by cysteine in the
S143C mutant also gave active enzyme while S143A
757
C
H
H
COO

H
2
N
H
N
N
H
2
C
O
H
C
H
H
S
COO

N
H
H
2
C
N
O
H
H
+
H
Dehydroalanine
H
2
C
N
O

B
NH
2
H
H
+
H
H
H
+
b
a
Cinnamate
(14-46)
was inactive.
288,289
These results support the formation
of dehydroalanine. One proposed mechanism of action
of the enzymes involved addition of the substrate
amino group to the C = C bond of the dehydroalanine
followed by elimination (Eq. 14-46). Dissociation of
the C H bond in this step would be assisted by the
electron-accepting properties of the adjacent aromatic
ring. However, the proposed chemistry was not
convincing. Noting that a 5-nitro substituent on the
histidine greatly enhances the rate of reaction, Langer
N
N
O
142
CH
2
144
N
N
O
142
144
OH
N
N
O
142
H
OH
4-Methylidene-imidazole-5-one (MIO)
H
Ser
143
H
O
144
O
H
H
3
C
N
Ala
Gly
H
H
2
O
H
2
O
et al. proposed that the histidine reacts with the
electrophilic carbon in the pyruvoyl center as in
Eq. 14-47.
290,291
However, this chemistry, too, was
unprecedented in enzymology. Determination of the
three-dimensional structure of histidine ammonia-
lyase
292
led to the discovery of a new prosthetic group
and a solution to the problem. Two dehydration steps,
(Eq. 14-47) convert an Ala-Gly-Ser sequence within the
protein into 4-methylidine-imidazole-5-one (MIO),
a modified dehydroalanine with enhanced electron-
accepting properties. The proposed mechanism of
action is portrayed in Eq. 14-48.
(14-47)
N
N

OOC
H
3
N H
H H
N
N

OOC
H
3
N H
H
H
N
N
144
O
142
CH
2
+
H
H
N
N
H H
N
N
OH
CH
2
+
H
H
+
+
N
N
O
CH
2
H
H
(14-48)
F. Pyruvoyl Groups and Other Unusual Electrophilic Centers
758 Chapter 14. Coenzymes: Natures Special Reagents
48. Tanaka, K. (1981) N. Engl. J. Med. 304, 839840
49. Craft, D. V., Goss, N. H., Chandramouli, N.,
and Wood, H. G. (1985) Biochemistry 24, 2471
2476
50. Chauhan, J., and Dakshinamurti, K. (1986) J.
Biol. Chem. 261, 42684275
51. Alberts, A. W., and Vagelos, P. R. (1972) in The
Enzymes, 3rd ed., Vol. 6 (Boyer, P. D., ed), pp.
3782, Academic Press, New York
52. Kumar, G. K., Beegen, H., and Wood, H. G.
(1988) Biochemistry 27, 59725978
53. Lynen, F., Knappe, J., Lorch, E., Jutting, G.,
and Ringelmann, E. (1959) Angew. Chem. Int.
Ed. Engl. 71, 481486
54. Wood, H. G. (1979) Trends Biochem. Sci. 4,
N300N302
55. Polakis, S. E., Guchhait, R. B., and Lane, M. D.
(1972) J. Biol. Chem. 247, 12351337
55a. Chapman-Smith, A., Forbes, B. E., Wallace, J.
C., and Cronan, J. E., Jr. (1997) J. Biol. Chem.
272, 2601726022
55b. Reddy, D. V., Shenoy, B. C., Carey, P. R., and
Snnichsen, F. D. (2000) Biochemistry 39, 2509
2516
56. Tipton, P. A., and Cleland, W. W. (1988) J. Am.
Chem. Soc. 110, 58665869
57. Tipton, P. A., and Cleland, W. W. (1988)
Biochemistry 27, 4317 4325
58. Ogita, T., and Knowles, J. R. (1988) Biochemistry
27, 80288033
58a. Branson, J. P., and Attwood, P. V. (2000)
Biochemistry 39, 74807491
59. Perrin, C. L., and Dwyer, T. J. (1987) J. Am.
Chem. Soc. 109, 51635167
60. Kluger, R., Davis, P. P., and Adawadkar, P. D.
(1979) J. Am. Chem. Soc. 101, 59956000
61. King, S. W., Natarajan, R., Bembi, R., and Fife,
T. H. (1992) J. Am. Chem. Soc. 114, 1071510721
62. Kluger, R., and Taylor, S. D. (1991) J. Am.
Chem. Soc. 113, 996 1001
63. Taylor, S. D., and Kluger, R. (1993) J. Am.
Chem. Soc. 115, 867 871
64. Hoving, H., Crysell, B., and Leadbay, P. F.
(1985) Biochemistry 24, 61636169
65. OKeefe, S. J., and Knowles, J. R. (1986)
Biochemistry 25, 6077 6084
66. Kuo, D. J., and Rose, I. A. (1993) J. Am. Chem.
Soc. 115, 387390
67. Stubbe, J., Fish, S., and Abeles, R. H. (1980) J.
Biol. Chem. 255, 236242
68. Reed, G. H., and Scrutton, M. G. (1974) J. Biol.
Chem. 249, 61566162
68a. Werneburg, B. G., and Ash, D. E. (1997)
Biochemistry 36, 1439214402
69. DeTitta, G. T., Blessing, R. H., Moss, G. R.,
King, H. F., Sukumaran, D. K., and Roskwital-
ski, R. L. (1994) J. Am. Chem. Soc. 116, 6485
6493
70. Legge, G. B., Branson, J. P., and Attwood, P. V.
(1996) Biochemistry 35, 38493856
71. Mabrouk, G. M., Helmy, I. M., Thampy, K. G.,
and Wakil, S. J. (1990) J. Biol. Chem. 265, 6330
6338
72. Kim, K.-H., Lpez-Casillas, F., Bai, D. H., Luo,
X., and Pape, M. E. (1989) FASEB J. 3, 2250
2256
73. Harwood, J. L. (1988) Trends Biochem. Sci. 13,
330 331
74. LauBermair, E., Schwarz, E., Oesterhelt, D.,
Reinke, H., Beyreuther, K., and Dimroth, P.
(1989) J. Biol. Chem. 264, 1471014715
74a. Jockel, P., Schmid, M., Choinowski, T., and
Dimroth, P. (2000) Biochemistry 39, 43204326
75. Laussermair, E., Schwarz, E., Oesterhelt, D.,
Reinke, H., Beyreuther, K., and Dimroth, P.
(1989) J. Biol. Chem. 264, 1471014715
76. Huder, J. B., and Dimroth, P. (1993) J. Biol.
Chem. 268, 2456424571
1. Moodie, S. L., Mitchell, J. B. O., and Thornton,
J. M. (1996) J. Mol. Biol. 263, 486500
2. Lipmann, F. (1945) J. Biol. Chem. 160, 173190
3. Roskoski, R. N., Jr. (1987) Trends Biochem. Sci.
12, 136138
3a. Bentley, R. (2000) Trends Biochem. Sci. 25, 302
305
4. Martin, D. P., and Drueckhammer, D. G.
(1992) J. Am. Chem. Soc. 114, 72877288
5. Baraniak, J., and Frey, P. A. (1988) J. Am. Chem.
Soc. 110, 40594060
6. Richter, D., and Hilz, H. (1979) Trends Biochem.
Sci. 1, N123N124
7. Ohlrogge, J. B., Jaworski, J. G., and Post-
Beittenmiller, D. (1993) in Lipid Metabolism in
Plants (Moore, T. S., Jr., ed), pp. 332, CRC
Press, Boca Raton, Florida
8. Pavela-Vrancic, M., Pfeifer, E., Schrder, W.,
von Dhren, H., and Kleinkauf, H. (1994) J.
Biol. Chem. 269, 1496214966
9. Haese, A., Pieper, R., von Ostrowski, T., and
Zocher, R. (1994) J. Mol. Biol. 243, 116122
10. Pieper, R., Ebert-Khosla, S., Cane, D., and
Khosla, C. (1996) Biochemistry 35, 2054 2060
11. Cortes, J., Wiesmann, K. E. H., Roberts, G. A.,
Brown, M. J. B., Staunton, J., and Leadlay, P. F.
(1995) Science 268, 14871489
12. Brambl, R., and Plesofsky-Vig, N. (1986) Proc.
Natl. Acad. Sci. U.S.A. 83, 36443648
13. Berg, M., Hilbi, H., and Dimroth, P. (1996)
Biochemistry 35, 46894696
14. Dimroth, P. (1987) Microbiol. Rev. 51, 320340
14a. Hoenke, S., Wild, M. R., and Dimroth, P.
(2000) Biochemistry 39, 1322313232
15. Jackowski, S., and Rock, C. O. (1984) J. Biol.
Chem. 259, 18911895
15a. McAllister, K. A., Peery, R. B., Meier, T. I.,
Fischl, A. S., and Zhao, G. (2000) J. Biol. Chem.
275, 3086430872
15b. Chirgadze, N. Y., Briggs, S. L., McAllister, K.
A., Fischl, A. S., and Zhao, G. (2000) EMBO J.
19, 52815287
16. Whitty, A., Fierke, C. A., and Jencks, W. P.
(1995) Biochemistry 34, 1167811689
17. Fierke, C. A., and Jencks, W. P. (1986) J. Biol.
Chem. 261, 76037606
18. Langer, B. W., Jr., and Gyorgy, P. (1968) in The
Vitamins, 2nd ed., Vol. 2 (Sebrell, W. H., Jr., and
Harris, R. S., eds), pp. 294322, Academic
Press, New York
19. Bentley, R. (1985) Trends Biochem. Sci. 10, 5156
20. DeTitta, G. T., Edmonds, J. W., Stallings, W.,
and Donohue, J. (1976) J. Am. Chem. Soc. 98,
19201926
20a. Garton, S. D., Temple, C. A., Dhawan, I. K.,
Barber, M. J., and Rajagopalan, K. V. (2000) J.
Biol. Chem. 275, 67986805
20b. McIver, L., Baxter, R. L., and Campopiano, D.
J. (2000) J. Biol. Chem. 275, 1388813894
21. Wright, L. D., Cresson, E. L., Skeggs, H. R.,
Peck, R. L., Wolf, D. E., Wood, T. R., Valent, J.,
and Folkers, K. (1951) Science 114, 635636
22. Knowles, J. R. (1989) Ann. Rev. Biochem. 58,
195221
23. Lpez-Casillas, F., Bai, D.-H., Luo, X., Kong, I.
S., Hermodson, M. A., and Kim, K.-H. (1988)
Proc. Natl. Acad. Sci. U.S.A. 85, 57845788
24. Browner, M. F., Taroni, F., Sztul, E., and
Rosenberg, L. E. (1989) J. Biol. Chem. 264,
1268012685
25. Samols, D., Thornton, C. G., Murtif, V. L.,
Kumar, G. K., Haase, F. C., and Wood, H. G.
(1988) J. Biol. Chem. 263, 64616464
26. Len-Del-Rio, A., Leclerc, D., Akerman, B.,
Wakamatsu, N., and Gravel, R. A. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 46264630
27. Ahmad, P. M., and Ahmad, F. (1991) FASEB J.
5, 24822485
28. Wang, X., Wurtele, E. S., Keller, G., McKean,
A. L., and Nikolau, B. J. (1994) J. Biol. Chem.
269, 1176011769
28a. McKean, A. L., Ke, J., Song, J., Che, P.,
Achenbach, S., Nikolau, B. J., and Wurtele, E.
S. (2000) J. Biol. Chem. 275, 55825590
29. Weaver, L. M., Lebrun, L., Wurtele, E. S., and
Nikolau, B. J. (1995) Plant Physiol. 107, 1013
1014
30. Al-Feel, W., Chirala, S. S., and Wakil, S. J.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89, 4534
4538
31. Lamzin, V. S., Dauter, Z., Popov, V. O.,
Harutyunyan, E. H., and Wilson, K. S. (1994)
J. Mol. Biol. 236, 759785
32. Roessler, P. G., and Ohlrogge, J. B. (1993) J.
Biol. Chem. 268, 1925419259
32a. Abu-Elheiga, L., Almarza-Ortega, D. B.,
Baldini, A., and Wakil, S. J. (1997) J. Biol.
Chem. 272, 1066910677
32b. Abu-Elheiga, L., Brinkley, W. R., Zhong, L.,
Chirala, S. S., Woldegiorgis, G., and Wakil, S.
J. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 1444
1449
32c. Nikolskaya, T., Zagnitko, O., Tevzadze, G.,
Haselkorn, R., and Gornicki, P. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1464714651
32d. Schulte, W., Tpfer, R., Stracke, R., Schell, J.,
and Martini, N. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 34653470
32e. Jitrapakdee, S., Booker, G. W., Cassady, A. I.,
and Wallace, J. C. (1997) J. Biol. Chem. 272,
2052220530
33. Lim, F., Morris, C. P., Occhiodoro, F., and
Wallace, J. C. (1988) J. Biol. Chem. 263, 11493
11497
34. Kalousek, F., Darigo, M. D., and Rosenberg, L.
E. (1980) J. Biol. Chem. 255, 6065
35. Leon Del-Rio, A., and Gravel, R. A. (1994) J.
Biol. Chem. 269, 2296422968
36. Sasaki, Y., Hakamada, K., Suama, Y., Nagano,
Y., Furusawa, I., and Matsuno, R. (1993) J.
Biol. Chem. 268, 2511825123
37. Gornicki, P., Podkowinski, J., Scappino, L. A.,
DiMaio, J., Ward, E., and Haselkorn, R. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91,
68606864
38. Nenortas, E., and Beckett, D. (1996) J. Biol.
Chem. 271, 75597567
39. Waldrop, G. L., Rayment, I., and Holden, H.
M. (1994) Biochemistry 33, 1024910256
39a. Blanchard, C. Z., Lee, Y. M., Frantom, P. A.,
and Waldrop, G. L. (1999) Biochemistry 38,
33933400
40. Li, S.-J., and Cronan, J. E., Jr. (1992) J. Biol.
Chem. 267, 855863
41. Li, S.-J., and Cronan, J. E. J. (1992) J. Biol.
Chem. 267, 1684116847
42. Ha, J., Daniel, S., Broyles, S. S., and Kim, K.-
H. (1994) J. Biol. Chem. 269, 2216222168
43. Wood, H. G., Harmon, F. R., Wuhr, B.,
Hubner, K., and Lynen, F. (1980) J. Biol. Chem.
255, 73977409
44. Shenoy, B. C., and Wood, H. G. (1988) FASEB
J. 2, 23962401
45. Wilson, K. P., Shewchuk, L. M., Brennan, R.
G., Otsuka, A. J., and Matthews, B. W. (1992)
Proc. Natl. Acad. Sci. U.S.A. 89,
92579261
46. Wolf, B. (1995) in The Metabolic and Molecular
Bases of Inherited Disease, 7th ed., Vol. 2
(Scriver, C. R., Beaudet, A. L., Sly, W. S., and
Valle, D., eds), pp. 31513177, McGraw-Hill,
New York
47. Taroni, F., and Rosenberg, L. E. (1991) J. Biol.
Chem. 266, 13267 13271
47a. Reche, P., and Perham, R. N. (1999) EMBO J.
18, 26732682
References
759
77. Bendrat, K., and Buckel, W. (1993) Eur. J.
Biochem. 211, 697702
78. Ahmad, F., Lygre, D. G., Jacobson, B. E., and
Wood, H. G. (1972) J. Biol. Chem. 247, 6299
6305
79. Metzler, D. E. (1960) in The Enzymes, 2nd ed.,
Vol. 2 (Boyer, P. D., Lardy, H., and Myrback,
K., eds), pp. 295337, Academic Press, New
York
80. Schellenberger, A., and Schowen, R. L., eds.
(1988) Thiamin Pyrophosphate Biochemistry,
CRC Press, Boca Raton, Florida (2 Vols.)
81. Krampitz, L. O. (1970) Thiamin Diphosphate
and Its Catalytic Functions, Dekker, New York
82. Gubler, C. J., ed. (1976) Thiamine, Wiley, New
York
83. Gallo, A., Mieyal, J. J., and Sable, H. Z. (1978)
Bioorg. Chem. 4, 147177
83a. Jordan, F., Li, H., and Brown, A. (1999)
Biochemistry 38, 63696373
84. Kluger, R. (1987) Chem. Rev. 87, 863876
85. Sable, H. Z., ed., and Gubler, C. J., ed. (1982)
Ann. N.Y. Acad. Sci. 378
86. Karlson, P. (1984) Trends Biochem. Sci. 9, 536
537
87. Panijpan, B. (1979) J. Chem. Educ. 56, 805806
88. Suchy, J., Mieyal, J. J., Bantle, G., and Sable, H.
Z. (1972) J. Biol. Chem. 247, 59055912
89. Williams, R. R., and Spies, T. D. (1938) Vitamin
B
1
and Its Use in Medicine, Macmillan, New
York
90. Kluger, R., Lam, J. F., Pezacki, J. P., and Yang,
C.-M. (1995) J. Am. Chem. Soc. 117, 1138311389
91. Windheuser, J. J., and Higuchi, T. (1962) J.
Pharm. Sci. 51, 354 364
92. Costello, C. A., Kelleher, N. L., Abe, M.,
McLafferty, F. W., and Begley, T. P. (1996) J.
Biol. Chem. 271, 34453452
92a. Campobasso, N., Costello, C. A., Kinsland, C.,
Begley, T. P., and Ealick, S. E. (1998)
Biochemistry 37, 1598115989
92b. Webb, E., and Downs, D. (1997) J. Biol. Chem.
272, 1570215707
92c. Nosaka, K., Onozuka, M., Nishino, H.,
Nishimura, H., Kawasaki, Y., and Ueyama, H.
(1999) J. Biol. Chem. 274, 3412934133
93. Mizuhara, S., and Handler, P. (1954) J. Am.
Chem. Soc. 76, 571573
94. Breslow, R. (1958) J. Am. Chem. Soc. 80, 3719
3726
95. Hopmann, R. F. W., Brugnoni, G. P., and Fol,
B. (1982) J. Am. Chem. Soc. 104, 13411344
96. Washabaugh, M. W., and Jencks, W. P. (1989)
J. Am. Chem. Soc. 111, 683692
97. Breslow, R., and McNelis, E. (1959) J. Am.
Chem. Soc. 81, 30803082
98. Kluger, R., Chin, J., and Smyth, T. (1981) J.
Am. Chem. Soc. 103, 884888
99. Kluger, R., and Smyth, T. (1981) J. Am. Chem.
Soc. 103, 12161218
100. Kluger, R., Karimian, K., Gish, G., Pangborn,
W. A., and DeTitta, G. T. (1987) J. Am. Chem.
Soc. 109, 618620
101. Crosby, J., Stone, R., and Lienhard, G. E.
(1970) J. Am. Chem. Soc. 92, 28912900
102. Shin, W., Oh, D.-G., Chae, C.-H., and Yoon,
T.-S. (1993) J. Am. Chem. Soc. 115, 1223812250
103. Lobell, M., and Crout, D. H. G. (1996) J. Am.
Chem. Soc. 118, 18671873
104. Gallo, A. A., and Sable, H. Z. (1974) J. Biol.
Chem. 249, 13821389
105. Schellenberger, A. (1967) Angew. Chem. Int. Ed.
Engl. 6, 10241035
106. Golbik, R., Neef, H., Hbner, G., Knig, S.,
Seliger, B., Meshalkina, L., Kochetov, G. A.,
and Schellenberger, A. (1991) Bioorg. Chem. 19,
1017
References
107. Knig, S., Schellenberger, A., Neef, H., and
Schneider, G. (1994) J. Biol. Chem. 269, 10879
10882
108. Jordan, F., and Mariam, Y. H. (1978) J. Am.
Chem. Soc. 100, 2534 2541
109. Metzler, D. E. (1977) Biochemistry; The
Chemical Reactions of Living Cells, Academic
Press, New York (440 441)
109a. Kern, D., Kern, G., Neef, H., Tittmann, K.,
Killenberg-Jabs, M., Wikner, C., Schneider, G.,
and Hbner, G. (1997) Science 275, 6770
110. Singleton, C. K., Wang, J. J.-L., Shan, L., and
Martin, P. R. (1996) Biochemistry 35, 15865
15869
111. Guo, F., Zhang, D., Kahyaoglu, A., Farid, R.
S., and Jordan, F. (1998) Biochemistry 37,
1337913391
111a. Li, H., Furey, W., and Jordan, F. (1999)
Biochemistry 38, 999210003
112. Gish, G., Smyth, T., and Kluger, R. (1988) J.
Am. Chem. Soc. 110, 62306234
113. Annan, N., Paris, R., and Jordan, F. (1989) J.
Am. Chem. Soc. 111, 88958901
114. Alvarez, F. J., Ermer, J., Hbner, G., Schellen-
berger, A., and Schowen, R. L. (1991) J. Am.
Chem. Soc. 113, 84028409
115. Alvarez, F. J., Ermer, J., Hbner, G., Schellen-
berger, A., and Schowen, R. L. (1995) J. Am.
Chem. Soc. 117, 16781683
116. Sun, S., Duggleby, R. G., and Schowen, R. L.
(1995) J. Am. Chem. Soc. 117, 73177322
117. Harris, T. K., and Washabaugh, M. W. (1995)
Biochemistry 34, 1399414000
118. Harris, T. K., and Washabaugh, M. W. (1995)
Biochemistry 34, 1400114011
119. Muller, Y. A., and Schulz, G. E. (1993) Science
259, 965967
120. Muller, Y. A., Schumacher, G., Rudolph, R., and
Schulz, G. E. (1994) J. Mol. Biol. 237, 315335
121. Dyda, F., Furey, W., Swaminathan, S., Sax, M.,
Farrenkopf, B., and Jordan, F. (1993) Biochem-
istry 32, 61656170
122. Arjunan, P., Umland, T., Dyda, R., Swami-
nathan, S., Furey, W., Sax, M., Farrenkopf, B.,
Gao, Y., Zhang, D., and Jordan, F. (1996) J.
Mol. Biol. 256, 590600
122a. Dobritzsch, D., Knig, S., Schneider, G., and
Lu, G. (1998) J. Biol. Chem. 273, 2019620204
122b. Chang, A. K., Nixon, P. F., and Duggleby, R.
G. (2000) Biochemistry 39, 94309437
122c. Li, H., and Jordan, F. (1999) Biochemistry 38,
1000410012
123. Wikner, C., Meshalkina, L., Nilsson, U.,
Nikkola, M., Lindqvist, Y., Sundstrm, M.,
and Schneider, G. (1994) J. Biol. Chem. 269,
3214432150
124. Nikkola, M., Lindqvist, Y., and Schneider, G.
(1994) J. Mol. Biol. 238, 387404
124a. Hasson, M. S., Muscate, A., McLeish, M. J.,
Polovnikova, L. S., Gerlt, J. A., Kenyon, G. L.,
Petsko, G. A., and Ringe, D. (1998) Biochemis-
try 37, 99189930
125. Killenberg-Jabs, M., Knig, S., Eberhardt, I.,
Hohmann, S., and Hbner, G. (1997) Biochem-
istry 36, 19001905
126. Koga, J., Adachi, T., and Hidaka, H. (1992) J.
Biol. Chem. 267, 1582315828
127. Hasson, M. S., Muscate, A., Henehan, G. T.
M., Guidinger, P. F., Petsko, G. A., Ringe, D.,
and Kenyon, G. L. (1995) Protein Sci. 4, 955959
128. Ibdah, M., Bar-Ilan, A., Livnah, O., Schloss, J.
V., Barak, Z., and Chipman, D. M. (1996)
Biochemistry 35, 1628216291
129. Bernasconi, P., Woodworth, A. R., Rosen, B.
A., Subramanian, M. V., and Siehl, D. L. (1995)
J. Biol. Chem. 270, 1738117385
130. Ott, K.-H., Kwagh, J.-G., Stockton, G. W.,
Sidorov, V., and Kakefuda, G. (1996) J. Mol.
Biol. 263, 359368
131. Stivers, J. T., and Washabaugh, M. W. (1993)
Biochemistry 32, 13472 13482
132. Chang, Y.-Y., Wang, A.-Y., and Cronan, J. E., Jr.
(1993) J. Biol. Chem. 268, 39113919
132a. Kuzuyama, T., Takahashi, S., Takagi, M., and
Seto, H. (2000) J. Biol. Chem. 275, 1992819932
132b. Wikner, C., Nilsson, U., Meshalkina, L.,
Udekwu, C., Lindqvist, Y., and Schneider, G.
(1997) Biochemistry 36, 1564315649
133. Chuang, L. F., and Collins, E. B. (1968) J.
Bacteriol. 95, 20832089
134. Gruys, K. J., Datta, A., and Frey, P. A. (1989)
Biochemistry 28, 90719080
135. Flournoy, D. S., and Frey, P. A. (1989) Biochem-
istry 28, 95949602
135a. Tittmann, K., Proske, D., Spinka, M., Ghisla,
S., Rudolph, R., Hbner, G., and Kern, G.
(1998) J. Biol. Chem. 273, 1292912934
136. Matsuda, T., and Cooper, J. R. (1983) Biochem-
istry 22, 22092213
137. Barchi, R. L., and Viale, R. O. (1976) J. Biol.
Chem. 251, 193197
138. Nishino, K., Itokawa, Y., Nishino, N., Piros,
K., and Cooper, J. R. (1983) J. Biol. Chem. 258,
187111878
139. Braunstein, A. E., and Kritzmann, M. G. (1937)
Enzymologia 2, 751752
140. Braunstein, A. E., and Kritzmann, M. G.
(1937) Biokhim. 2, 242 262,859874
141. Braunstein, A. E. (1973) in The Enzymes, 3rd
ed., Vol. 9 (Boyer, P. D., ed), pp. 379481,
Academic Press, New York
142. Christen, P., and Metzler, D. E., eds. (1984)
Transaminases, Wiley, New York
143. Torchinsky, Y. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical
Aspects, Vol. 1A (Dolphin, R., Poulson, R., and
Avramovic, O., eds), pp. 169221, Wiley,
New York
144. Snell, E. E. (1944) J. Biol. Chem. 154, 313314
145. Snell, E. E. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical
Aspects, Vol. 1A (Dolphin, D., Poulson, R., and
Avramovc, O., eds), pp. 112, Wiley, New York
146. Gunsalus, I. C., Bellamy, W. D., and Umbreit,
W. W. (1944) J. Biol. Chem. 155, 685686
147. Snell, E. E. (1981) in Methods in Vitamin B
6
Nutrition (Leklem, J. E., and Reynolds, R. D.,
eds), pp. 119, Plenum, New York
148. Metzler, D. E., Ikawa, M., and Snell, E. E.
(1954) J. Am. Chem. Soc. 76, 648 652
149. Kallen, R. G., Korpela, T., Martell, A. E.,
Matsushima, Y., Metzler, C. M., Metzler, D. E.,
Morozov, Y. V., Ralston, I. M., Savin, F. A.,
Torchinsky, Y. M., and Ueno, H. (1984) in
Transaminases (Christen, P., and Metzler, D. E.,
eds), pp. 37106, Wiley, New York
150. Leussing, D. L. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical
Aspects, Vol. 1A (Dolphin, D., Poulson, R., and
Avramovc, O., eds), pp. 69115, Wiley, New
York
151. Braunstein, A. E., and Kritzmann, M. G.
(1952) Dokl. Akad. Nauk. SSSR 85, 11151118
152. Braunstein, A. E., and Kritzmann, M. G.
(1952) Biokhim. 18, 393411
153. Shaw, J. P., Petsko, G. A., and Ringe, D. (1997)
Biochemistry 36, 13291342
153a. Stamper, C. G. F., Morollo, A. A., and Ringe,
D. (1998) Biochemistry 37, 1043810445
153b. Morollo, A. A., Petsko, G. A., and Ringe, D.
(1999) Biochemistry 38, 32933301
153c. Sun, S., and Toney, M. D. (1999) Biochemistry
38, 40584065
154. Aswad, D. W., and Johnson, B. A. (1987)
Trends Biochem. Sci. 12, 155158
154a. Watababe, A., Kurokawa, Y., Yoshimura, T.,
Kurihara, T., Soda, K., and Esaki, N. (1999) J.
Biol. Chem. 274, 41894194
760 Chapter 14. Coenzymes: Natures Special Reagents
185a. Burkhard, P., Tai, C.-H., Jansonius, J. N., and
Cook, P. F. (2000) J. Mol. Biol. 303, 279286
186. Cook, P. F., Tai, C.-H., Hwang, C.-C., Woehl,
E. U., Dunn, M. F., and Schnackerz, K. D.
(1996) J. Biol. Chem. 271, 2584225849
186a. Benci, S., Vaccari, S., Mozzarelli, A., and
Cook, P. F. (1997) Biochemistry 36, 15419
15427
187. Kery, V., Bukovska, G., and Kraus, J. P. (1994)
J. Biol. Chem. 269, 2528325288
188. Peracchi, A., Bettati, S., Mozzarelli, A., Rossi,
G. L., Miles, E. W., and Dunn, M. F. (1996)
Biochemistry 35, 18721880
188a. Jhee, K.-H., McPhie, P., and Miles, E. W. (2000)
J. Biol. Chem. 275, 1154111544
188b. Taoka, S., Widjaja, L., and Banerjee, R. (1999)
Biochemistry 38, 1315513161
188c. Ojha, S., Hwang, J., Kabil, O., Penner-Hahn, J.
E., and Banerjee, R. (2000) Biochemistry 39,
1054210547
189. Palcic, M. M., and Floss, H. G. (1986) in
Vitamin B
6
, Pyridoxal Phosphate: Chemical,
Biochemical and Medical Aspects, Vol. 1A
(Dolphin, D., Poulson, R., and Avramovc, O.,
eds), pp. 2568, Wiley, New York
190. Pan, P., Woehl, E., and Dunn, M. F. (1997)
Trends Biochem. Sci. 22, 2227
191. Sukhareva, B. S. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical
Aspects, Vol. 1B (Dolphin, D., Poulson, R., and
Avramovc, O., eds), pp. 325353, Wiley, New
York
192. Sandmeier, E., Hale, T. I., and Christen, P.
(1994) Eur. J. Biochem. 221, 9971002
193. Nathan, B., Hsu, C.-C., Bao, J., Wu, R., and
Wu, J.-Y. (1994) J. Biol. Chem. 269, 72497254
194. Porter, T. G., and Martin, D. L. (1988) Biochim.
Biophys. Acta. 874, 235244
195. Bu, D.-F., Erlander, M. G., Hitz, B. C.,
Tillakaratne, N. J. K., Kaufman, D. L.,
Wagner-McPherson, C. B., Evans, G. A., and
Tobin, A. J. (1992) Proc. Natl. Acad. Sci. U.S.A.
89, 21152119
196. Dirkx, R., Jr., Thomas, A., Li, L., Lernmark, .,
Sherwin, R. S., DeCamilli, P., and Solimena,
M. (1995) J. Biol. Chem. 270, 22412246
196a. Qu, K., Martin, D. L., and Lawrence, C. E.
(1998) Protein Sci. 7, 10921105
196b. Kanaani, J., Lissin, D., Kash, S. F., and
Baekkeskov, S. (1999) J. Biol. Chem. 274,
3720037209
197. Malashkevich, V. N., Filipponi, P., Sauder, U.,
Dominici, P., Jansonius, J. N., and Voltattorni,
C. B. (1992) J. Mol. Biol. 224, 11671170
198. Dominici, P., Filipponi, P., Schinnin, M. E.,
Barra, D., and Voltattorni, C. B. (1990) Ann.
N.Y. Acad. Sci. 585, 162171
199. Reith, J., Benkelfat, C., Sherwin, A., Yasuhara,
Y., Kuwabara, H., Andermann, F., Bachneff,
S., Cumming, P., Diksic, M., Dyve, S. E.,
Etinne, P., Evans, A. C., Lal, S., Shevell, M.,
Savard, G., Wong, D. F., Chouinard, G., and
Gjedde, A. (1994) Proc. Natl. Acad. Sci. U.S.A.
91, 1165111654
199a. Hayashi, H., Tsukiyama, F., Ishii, S.,
Mizuguchi, H., and Kagamiyama, H. (1999)
Biochemistry 38, 1561515622
199b. Nishino, J., Hayashi, H., Ishii, S., and
Kagamiyama, H. (1997) J. Biochem. 121, 604
611
200. Bhattacharjee, M. K., and Snell, E. E. (1990) J.
Biol. Chem. 265, 66646668
201. Chudomelka, P. J., Ramaley, R. F., and Murrin,
L. C. (1990) Neurochemical Research 15, 1724
202. Yatsunami, K., Tsuchikawa, M., Kamada, M.,
Hori, K., and Higuchi, T. (1995) J. Biol. Chem.
270, 3081330817
155. Hoffmann, K., Schneider-Scherzer, E.,
Kleinkauf, H., and Zocher, R. (1994) J. Biol.
Chem. 269, 1271012714
155a. Cheng, Y.-Q., and Walton, J. D. (2000) J. Biol.
Chem. 275, 49064911
156. Schnell, M. J., Cooper, O. B., and Snyder, S. H.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 2013
2018
157. Yamauchi, T., Choi, S.-Y., Okada, H., Yohda,
M., Kumagai, H., Esaki, N., and Soda, K.
(1992) J. Biol. Chem. 267, 1836118364
158. Yohda, M., Endo, I., Abe, Y., Ohta, T., Iida, T.,
Maruyama, T., and Kagawa, Y. (1996) J. Biol.
Chem. 271, 2201722021
159. Li, N., and Mattoo, A. K. (1994) J. Biol. Chem.
269, 6908 6917
160. Li, N., Jiang, X. N., Cai, G. P., and Yang, S. F.
(1996) J. Biol. Chem. 271, 2573825741
161. Hohenester, E., White, M. F., Kirsch, J. F., and
Jansonius, J. N. (1994) J. Mol. Biol. 243, 947949
161a. Capitani, G., Hohenester, E., Feng, L., Storici,
P., Kirsch, J. F., and Jansonius, J. N. (1999) J.
Mol. Biol. 294, 745756
162. Ramalingam, K., Lee, K.-M., Woodard, R. W.,
Bleecker, A. B., and Kende, H. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 78207824
163. Rhee, S., Silva, M. M., Hyde, C. C., Rogers, P.
H., Metzler, C. M., Metzler, D. E., and Arnone,
A. (1997) J. Biol. Chem. 272, 1729317302
164. Malashkevich, V. N., Strokopytov, B. V.,
Borisov, V. V., Dauter, Z., Wilson, K. S., and
Torchinsky, Y. M. (1995) J. Mol. Biol. 247, 111
124
165. McPhalen, C. A., Vincent, M. G., and Jansonius,
J. N. (1992) J. Mol. Biol. 225, 495517
166. Kamitori, S., Okamoto, A., Hirotsu, K.,
Higuchi, T., Kuramitsu, S., Kagamiyama, H.,
Matsuura, Y., and Katsube, Y. (1990)
J. Biochem. 108, 175184
167. Okamoto, A., Higuchi, T., Hirotsu, K.,
Kuramitsu, S., and Kagamiyama, H. (1994) J.
Biochem. 116, 95107
167a. Jeffery, C. J., Barry, T., Doonan, S., Petsko, G.
A., and Ringe, D. (1998) Protein Sci. 7, 1380
1387
168. Uchida, C., Funai, T., Oda, T., Ohbayashi, K.,
and Ichiyama, A. (1994) J. Biol. Chem. 269,
88468856
168a. Matsui, I., Matsui, E., Sakai, Y., Kikuchi, H.,
Kawarabayasi, Y., Ura, H., Kawaguchi, S.-i,
Kuramitsu, S., and Harata, K. (2000) J. Biol.
Chem. 275, 48714879
168b. Okada, K., Hirotsu, K., Sato, M., Hayashi, H.,
and Kagamiyama, H. (1997) J. Biochem. 121,
637641
168c. Davoodi, J., Drown, P. M., Bledsoe, R. K.,
Wallin, R., Reinhart, G. D., and Hutson, S. M.
(1998) J. Biol. Chem. 273, 49824989
168d. van Ophem, P. W., Peisach, D., Erickson, S. D.,
Soda, K., Ringe, D., and Manning, J. M. (1999)
Biochemistry 38, 13231331
168e. Hester, G., Stark, W., Moser, M., Kallen, J.,
Markovic-Housley, Z., and Jansonius, J. N.
(1999) J. Mol. Biol. 286, 829850
168f. Storici, P., Capitani, G., Mller, R., Schirmer,
T., and Jansonius, J. N. (1999) J. Mol. Biol. 285,
297309
168g. Shah, S. A., Shen, B. W., and Brnger, A. T.
(1997) Structure 5, 10671075
168h. Ledwidge, R., and Blanchard, J. S. (1999)
Biochemistry 38, 30193024
168i. Smith, M. A., King, P. J., and Grimm, B. (1998)
Biochemistry 37, 319329
168j. Kck, H., Sandmark, J., Gibson, K., Schneider,
G., and Lindqvist, Y. (1999) J. Mol. Biol. 291,
857876
168k. Contestabile, R., Angelaccio, S., Maytum, R.,
Bossa, F., and John, R. A. (2000) J. Biol. Chem.
275, 38793886
169. Kintanar, A., Metzler, C. M., Metzler, D. E.,
and Scott, R. D. (1991) J. Biol. Chem. 266,
1722217229
170. Miles, E. W. (1986) in Vitamin B
6
, Pyridoxal
Phosphate: Chemical, Biochemical and Medical
Aspects, Vol. 1B (Dolphin, D., Poulson, R., and
Avramovc, O., eds), pp. 253310, Wiley,
New York
171. Marceau, M., Lewis, S. D., Kojiro, C. L., and
Shafer, J. A. (1989) J. Biol. Chem. 264, 27532757
172. Ogawa, H., Gomi, T., Konishi, K., Date, T.,
Nakashima, H., Nose, K., Matsuda, Y.,
Peraino, C., Pilot, H. C., and Fujioka, M.
(1989) J. Biol. Chem. 264, 1581815823
173. Obmolova, G., Tepliakov, A., Harutyunyan,
E., Wahler, G., and Schnackerz, K. D. (1990)
J. Mol. Biol. 214, 641642
173a. Ogawa, H., Takusagawa, F., Wakaki, K., Kishi,
H., Eskandarian, M. R., Kobayashi, M., Date,
T., Huh, N.-H., and Pitot, H. C. (1999) J. Biol.
Chem. 274, 1285512860
174. Snell, E. E. (1975) Adv. Enzymol. 42, 287333
175. Metzler, C. M., Viswanath, R., and Metzler, D.
E. (1991) J. Biol. Chem. 266, 93749381
176. Sloan, M. J., and Phillips, R. S. (1996) Biochem-
istry 35, 1616516173
176a. Isupov, M. N., Antson, A. A., Dodson, E. J.,
Dodson, G. G., Dementieva, I. S.,
Zakomirdina, L. N., Wilson, K. S., Dauter, Z.,
Lebedev, A. A., and Harutyunyan, E. H.
(1998) J. Mol. Biol. 276, 603623
176b. Ikushiro, H., Hayashi, H., Kawata, Y., and
Kagamiyama, H. (1998) Biochemistry 37, 3043
3052
176c. Phillips, R. S., Sundararaju, B., and Faleev, N.
G. (2000) J. Am. Chem. Soc. 122, 10081014
177. Antson, A. A., Demidkina, T. V., Gollnick, P.,
Dauter, Z., Von Tersch, R. L., Long, J.,
Berezhnoy, S. N., Phillips, R. S.,
Harutyunyan, E. H., and Wilson, K. S. (1993)
Biochemistry 32, 4195 4206
177a. Sundararaju, B., Antson, A. A., Phillips, R. S.,
Demidkina, T. V., Barbolina, M. V., Gollnick,
P., Dodson, G. G., and Wilson, K. S. (1997)
Biochemistry 36, 65026510
178. Chen, H. Y., Demidkina, T. V., and Phillips, R.
S. (1995) Biochemistry 34, 1227612283
178a. Sundararaju, B., Chen, H., Shilcutt, S., and
Phillips, R. S. (2000) Biochemistry 39, 8546
8555
179. Block, E. (1985) Sci. Am. 252(Mar), 114119
180. Block, E., Gillies, J. Z., Gillies, C. W., Bazzi, A.
A., Putman, D., Revelle, L. K., Wang, D., and
Zhang, X. (1996) J. Am. Chem. Soc. 118, 7492
7501
181. Gentry-Weeks, C. R., Spokes, J., and Thomp-
son, J. (1995) J. Biol. Chem. 270, 76957702
182. Clausen, T., Huber, R., Laber, B., Pohlenz, H.-
D., and Messerschmidt, A. (1996) J. Mol. Biol.
262, 202224
183. Nagasawa, T., Tanizawa, K., Satoda, T., and
Yamada, H. (1988) J. Biol. Chem. 263, 958964
184. Hyde, C. C., Ahmed, S. A., Padlan, E. A.,
Miles, E. W., and Davies, D. R. (1988) J. Biol.
Chem. 263, 1785717871
184a. Rhee, S., Miles, E. W., and Davies, D. R. (1998)
J. Biol. Chem. 273, 85538555
184b. Weyand, M., and Schlichting, I. (1999)
Biochemistry 38, 1646916480
184c. Sachpatzidis, A., Dealwis, C., Lubetsky, J. B.,
Liang, P.-H., Anderson, K. S., and Lolis, E.
(1999) Biochemistry 38, 1266512674
184d. Rhee, S., Miles, E. W., Mozzarelli, A., and
Davies, D. R. (1998) Biochemistry 37, 10653
10659
185. Rege, V. D., Kredich, N. M., Tai, C.-H.,
Karsten, W. E., Schnackerz, K. D., and Cook,
P. F. (1996) Biochemistry 35, 13485 13493
References
761
References
203. Osterman, A. L., Kinch, L. N., Grishin, N. V.,
and Phillips, M. A. (1995) J. Biol. Chem. 270,
1179711802
204. Momany, C., Ernst, S., Ghosh, R., Chang, N.-
L., and Hackert, M. L. (1995) J. Mol. Biol. 252,
643655
205. Osterman, A. L., Brooks, H. B., Jackson, L.,
Abbott, J. J., and Phillips, M. A. (1999)
Biochemistry 38, 1181411826
206. Viguera, E., Trelles, O., Urdiales, J. L., Mats,
J. M., and Snchez-Jimnez, F. (1994) Trends
Biochem. Sci. 19, 318319
206a. Almrud, J. J., Oliveira, M. A., Kern, A. D.,
Grishin, N. V., Phillips, M. A., and Hackert,
M. L. (2000) J. Mol. Biol. 295, 716
206b. Jackson, L. K., Brooks, H. B., Osterman, A. L.,
Goldsmith, E. J., and Phillips, M. A. (2000)
Biochemistry 39, 1124711257
206c. Grishin, N. V., Osterman, A. L., Brooks, H. B.,
Phillips, M. A., and Goldsmith, E. J. (1999)
Biochemistry 38, 1517415184
207. Kume, A., Koyata, H., Sakakibara, T.,
Ishiguro, Y., Kure, S., and Hiraga, K. (1991) J.
Biol. Chem. 266, 33233329
208. Pasternack, L. B., Laude, D. A., Jr., and Appling,
D. R. (1992) Biochemistry 31, 87138719
209. Kennard, O., and Salisbury, S. A. (1993) J. Biol.
Chem. 268, 1070110704
209a. Okamura-Ikeda, K., Fujiwara, K., and
Motokawa, Y. (1999) J. Biol. Chem. 274, 17471
17477
209b. Guilhaudis, L., Simorre, J.-P., Blackledge, M.,
Marion, D., Gans, P., Neuburger, M., and
Douce, R. (2000) Biochemistry 39, 42594266
210. Akhtar, M., Stevenson, D. E., and Gani, D.
(1990) Biochemistry 29, 76487660
210a. Zhou, X., and Toney, M. D. (1999) Biochemistry
38, 311320
210b. Malashkevich, V. N., Strop, P., Keller, J. W.,
Jansonius, J. N., and Toney, M. D. (1999) J.
Mol. Biol. 294, 193200
211. Scarsdale, J. N., Kazanina, G., Radaev, S.,
Schirch, V., and Wright, H. T. (1999)
Biochemistry 38, 83478358
212. Stover, P. J., Chen, L. H., Suh, J. R., Stover, D.
M., Keyomarsi, K., and Shane, B. (1997) J. Biol.
Chem. 272, 18421848
212a. Szebenyi, D. M. E., Liu, X., Kriksunov, I. A.,
Stover, P. J., and Thiel, D. J. (2000) Biochemistry
39, 1331313323
213. Webb, H. K., and Matthews, R. G. (1995) J.
Biol. Chem. 270, 1720417209
213a. Jagath, J. R., Sharma, B., Appaji Rao, N., and
Savithri, H. S. (1997) J. Biol. Chem. 272, 24355
24362
213b. Scarsdale, J. N., Radaev, S., Kazanina, G.,
Schirch, V., and Wright, H. T. (2000) J. Mol.
Biol. 296, 155168
214. Tong, H., and Davis, L. (1994) J. Biol. Chem.
269, 40574064
215. Tong, H., and Davis, L. (1995) Biochemistry 34,
33623367
216. Cotter, P. D., Baumann, M., and Bishop, D. F.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89, 4028
4032
217. Ferreira, G. C., Neame, P. J., and Dailey, H. A.
(1993) Protein Sci. 2, 19591965
218. Cox, T. C., Bottomley, S. S., Wiley, J. S.,
Bawden, M. J., Matthews, C. S., and May, B.
K. (1994) N. Engl. J. Med. 330, 675679
218a. Hunter, G. A., and Ferreira, G. C. (1999) J. Biol.
Chem. 274, 1222212228
218b. Hunter, G. A., and Ferreira, G. C. (1999)
Biochemistry 38, 37113718
219. Williams, R. D., Nixon, D. W., and Merrill, A.
H., Jr. (1984) Cancer Research 44, 19181923
220. Webster, S. P., Alexeev, D., Campopiano, D. J.,
Watt, R. M., Alexeeva, M., Sawyer, L., and
Baxter, R. L. (2000) Biochemistry 39, 516528
220a. Steegborn, C., Messerschmidt, A., Laber, B.,
Streber, W., Huber, R., and Clausen, T. (1999)
J. Mol. Biol. 290, 983996
220b. Steegborn, C., Clausen, T., Sondermann, P.,
Jacob, U., Worbs, M., Marinkovic, S., Huber,
R., and Wahl, M. C. (1999) J. Biol. Chem. 274,
1267512684
220c. Laber, B., Gerbling, K.-P., Harde, C., Neff, K.-
H., Nordhoff, E., and Pohlenz, H.-D. (1994)
Biochemistry 33, 34133423
221. Tate, S. E., and Meister, A. (1971) Adv.
Enzymol. 35, 503543
222. Mihara, H., Kurihara, T., Watanabe, T.,
Yoshimura, T., and Esaki, N. (2000) J. Biol.
Chem. 275, 61956200
223. Esaki, N., Seraneeprakarn, V., Tanaka, H., and
Soda, K. (1988) J. Bacteriol. 170, 751756
223a. Mihara, H., Maeda, M., Fujii, T., Kurihara, T.,
Hata, Y., and Esaki, N. (1999) J. Biol. Chem.
274, 1476814772
223b. Fujii, T., Maeda, M., Mihara, H., Kurihara, T.,
Esaki, N., and Hata, Y. (2000) Biochemistry 39,
12631273
224. Zheng, L., and Dean, D. R. (1994) J. Biol. Chem.
269, 1872318726
225. Phillips, R. S., and Dua, R. K. (1991) J. Am.
Chem. Soc. 113, 73857388
226. Dua, R. K., Taylor, E. W., and Phillips, R. S.
(1993) J. Am. Chem. Soc. 115, 12641270
227. Krebs, E. G., and Fischer, E. H. (1964) Vitam.
Horm.(N. Y.) 22, 399410
228. Rubenstein, P. A., and Strominger, J. L. (1974)
J. Biol. Chem. 249, 37763781
229. Johnson, D. A., Gassner, G. T., Bandarian, V.,
Ruzicka, F. J., Ballou, D. P., Reed, G. H., and
Liu, H.-w. (1996) Biochemistry 35, 1584615856
230. Gonzalez-Porqu, P. (1986) in Vitamin B
6
,
Pyridoxal Phosphate: Chemical, Biochemical and
Medical Aspects, Vol. 1B (Dolphin, D., Poulson,
R., and Avramovc, O., eds), pp. 391419, Wiley,
New York
231. Bailey, G. B., Chotamangsa, O., and Vuttivej,
K. (1970) Biochemistry 9, 32433248
232. Toney, M. D., Hohenester, E., Cowan, S. W.,
and Jansonius, J. N. (1993) Science 261, 756759
233. Dunathan, H. C. (1971) Adv. Enzymol. 35, 79
134
234. Tobler, H. P., Gehring, H., and Christen, P.
(1987) J. Biol. Chem. 262, 89858989
235. Sugio, S., Petsko, G. A., Manning, J. M., Soda,
K., and Ringe, D. (1995) Biochemistry 34,
96619669
236. Syke, G. E., Potts, R., and Floss, H. G. (1974) J.
Am. Chem. Soc. 96, 15931595
237. Floss, H. G., and Vederas, J. C. (1982) in New
Comprehensive Biochemistry Stereochemistry,
Vol. 3 (Tamm, C., ed), pp. 161199, Elsevier,
Amsterdam
238. Palcic, M. M., and Floss, H. G. (1986) in
Vitamin B
6
, Pyridoxal Phosphate: Chemical,
Biochemical and Medical Aspects, Vol. 1A
(Dolphin, D., Poulson, R., and Avramovc, O.,
eds), pp. 2568, Wiley, New York
239. Metzler, D. E., Harris, C. M., Johnson, R. J.,
Siano, D. B., and Thomson, J. A. (1973)
Biochemistry 12, 53775392
240. Metzler, C. M., Cahill, A. E., and Metzler, D.
E. (1980) J. Am. Chem. Soc. 102, 60756082
241. Schirch, L., and Jenkins, W. T. (1964) J. Biol.
Chem. 239, 3801 3807
242. Metzler, C. M., Harris, A. G., and Metzler, D.
E. (1988) Biochemistry 27, 49234933
243. Schnackerz, K. D. (1986) in Vitamin B
6
,
Pyridoxal Phosphate: Chemical, Biochemical and
Medical Aspects, Vol. 1A (Dolphin, D., Poulson,
R., and Avramovc, O., eds), pp. 245264,
Wiley, New York
243a. Mehta, P. K., and Christen, P. (1998) in
Advances in Enzymology and Related Areas of
Molecular Biology, Vol. 74 (Purich, D. L., ed),
Wiley, New York (pp. 129184)
243b. Jansonius, J. N. (1998) Current Opinion in
Structural Biology 8, 759769
244. Watanabe, N., Yonaha, K., Sakabe, K., Sakabe,
N., Aibara, S., and Morita, Y. (1991) in Enzymes
Dependent on Pyridoxal Phosphate and Other
Carbonyl Compounds as Cofactors (Fukui, T.,
Kagamiyama, H., Soda, K., and Wada, H.,
eds), pp. 121124, Pergamon Press, Oxford
245. Huston, S. M., Bledsoe, R. K., Hall, T. R., and
Dawson, P. A. (1995) J. Biol. Chem. 270, 30344
30352
246. Ivanov, V. I., and Karpeisky, M. Y. (1969) Adv.
Enzymol. 32, 2153
247. Arnone, A., Christen, P., Jansonius, J. N., and
Metzler, D. E. (1985) in Transaminases (Christen,
P., and Metzler, D. E., eds), pp. 326370, Wiley,
New York
248. Rhee, S., Silva, M. M., Hyde, C. C., Rogers, P.
H., Metzler, C. M., Metzler, D. E., and Arnone,
A. (1997) J. Biol. Chem. 272, 1729317302
249. Metzler, C. M., Mitra, J., Metzler, D. E.,
Makinen, M. W., Hyde, C. C., Rogers, P., and
Arnone, A. (1988) J. Mol. Biol. 203, 197220
250. Malashkevich, V. N., Toney, M. D., and
Jansonius, J. N. (1993) Biochemistry 32, 13451
13462
251. von Stosch, A. G. (1996) Biochemistry 35,
1526015268
252. Metzler, C. M., Metzler, D. E., Kintanar, A.,
Scott, R. D., and Marceau, M. (1991) Biochem.
Biophys. Res. Commun. 178, 385392
253. Fukui, T., Kagamiyama, H., Soda, K., and
Wada, H., eds. (1991) Enzymes Dependent on
Pyridoxal Phosphate and Other Carbonyl
Compounds as Cofactors, Pergamon Press,
Oxford
254. Marino, G., Sannia, G., and Bossa, F., eds.
(1994) Biochemistry of Vitamin B
6
and PQQ,
Birkhuser, Basel
254a. Iriarte, A., Kagan, H. M., and Martinez-
Carrion, M., eds. (2000) Biochemistry and
Molecular Biology of Vitamin B6 and PQQ-
dependent Proteins, Birkhuser Verlag, Basel
255. Relimpio, A., Iriarte, A., Chlebowski, J. F., and
Martinez-Carrion, M. (1981) J. Biol. Chem. 256,
44784488
256. Gloss, L. M., and Kirsch, J. F. (1995) Biochemis-
try 34, 1232312332
257. Hwang, C.-C., Woehl, E. U., Minter, D. E.,
Dunn, M. F., and Cook, P. F. (1996) Biochemis-
try 35, 63586365
258. Pan, P., and Dunn, M. F. (1996) Biochemistry
35, 50025013
259. Anderson, K. S., Kim, A. Y., Quillen, J. M.,
Sayers, E., Yang, X.-J., and Miles, E. W. (1995)
J. Biol. Chem. 270, 2993629944
260. Schnackerz, K. D., Tai, C.-H., Simmons, J. W.,
III, Jacobson, T. M., Rao, G. S. J., and Cook, P.
F. (1995) Biochemistry 34, 12152 12160
261. Hayashi, H., and Kagamiyama, H. (1995)
Biochemistry 34, 94139423
262. Mattingly, M. E., Mattingly, J. R., Jr., and
Martinez-Carrion, M. (1982) J. Biol. Chem. 257,
88728878
263. Metzler, D. E. (1997) Methods Enzymol. 280,
3040
264. Higaki, T., Tanase, S., Nagashima, F., Morino,
Y., Scott, A. I., Williams, H. J., and Stolowich,
N. J. (1991) Biochemistry 30, 25192526
265. Goldberg, J. M., and Kirsch, J. F. (1996)
Biochemistry 35, 5280 5291
266. Recsei, P. A., and Snell, E. E. (1984) Ann. Rev.
Biochem. 53, 357387
267. van Peolje, P. D., and Snell, E. E. (1990) Ann.
Rev. Biochem. 59, 2959
762 Chapter 14. Coenzymes: Natures Special Reagents
268. Pishko, E. J., and Robertus, J. D. (1993)
Biochemistry 32, 4943 4948
269. Gallagher, T., Snell, E. E., and Hackert, M. L.
(1989) J. Biol. Chem. 264, 1273712743
270. Recsei, P. A., Huynh, Q. K., and Snell, E. E.
(1983) Proc. Natl. Acad. Sci. U.S.A. 80, 973977
271. Van Poelje, P. D., and Snell, E. E. (1988) Proc.
Natl. Acad. Sci. U.S.A. 85, 84498453
272. Huynh, Q. H., and Snell, E. E. (1986) J. Biol.
Chem. 261, 15211524
273. Shirahata, A., and Pegg, A. E. (1986) J. Biol.
Chem. 261, 1383313837
274. Stanley, B. A., Shantz, L. M., and Pegg, A. E.
(1994) J. Biol. Chem. 269, 79017907
274a. Xiong, H., and Pegg, A. E. (1999) J. Biol. Chem.
274, 3505935066
275. Clancey, C. J., Chang, S.-C., and Dowhan, W.
(1993) J. Biol. Chem. 268, 2458024590
276. Satre, M., and Kennedy, E. P. (1978) J. Biol.
Chem. 253, 479483
277. Williamson, J. M., and Brown, G. M. (1979) J.
Biol. Chem. 254, 80748082
278. Yang, H., and Abeles, R. H. (1987) Biochemis-
try 26, 4076 4081
279. Seto, B., and Stadtman, T. C. (1976) J. Biol. Chem.
251, 24352439
280. Seto, B. (1978) J. Biol. Chem. 253, 45254529
281. Hodgins, D. S., and Abeles, R. H. (1969) Arch.
Biochem. Biophys. 130, 274285
282. Arkowitz, R. A., and Abeles, R. H. (1989)
Biochemistry 28, 46394644
283. Stadtman, T. C., and Davis, J. N. (1991) J. Biol.
Chem. 266, 2214722153
283a. Kabisch, U. C., Grntzdrffer, A., Schierhorn,
A., Rcknagel, K. P., Andreesen, J. R., and
Pich, A. (1999) J. Biol. Chem. 274, 84458454
284. Furuta, T., Takahashi, H., Shibasaki, H., and
Kasuya, Y. (1992) J. Biol. Chem. 267, 12600
12605
285. Levy, H. L., Taylor, R. G., and McInnes, R. R.
(1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds),
pp. 11071123, McGraw-Hill, New York
286. Havir, E. A., and Hanson, K. R. (1985)
Biochemistry 24, 29592967
287. Consevage, M. W., and Phillips, A. T. (1985)
Biochemistry 24, 301308
288. Langer, M., Reck, G., Reed, J., and Rtey, J.
(1994) Biochemistry 33, 64626467
289. Langer, M., Lieber, A., and Rtey, J. (1994)
Biochemistry 33, 1403414038
290. Langer, M., Pauling, A., and Rtey, J. (1995)
Angew. Chem. Int. Ed. Engl. 34, 14641465
291. Schuster, B., and Rtey, J. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 84338437
292. Schwede, T. F., Rtey, J., and Schulz, G. E.
(1999) Biochemistry 38, 53555361
References
763
Study Questions
1. Discuss the role of biotin in metabolism and the
chemical mechanism of its action. Illustrate with
examples.
2. Biotin has been shown to be an essential compo-
nent of a bacterial oxaloacetate decarboxylase that
pumps two sodium ions out of a cell for each
oxaloacetate molecule decarboxylated. Propose a
chemical mechanism for the functioning of biotin
and also any ideas that you may have for the
operation of the sodium pump.
3. Write out the equation by which acetyl-CoA and
pyruvate can be converted into L-glutamate,
which can be observed in living animals by
13
C
NMR. If [2-
13
C] sodium acetate were injected into
the animal what labeling pattern could be antici-
pated in the L-glutamate?
4. Compare mechanisms and coenzyme requirements
for biological decarboxylation of the following
three groups of compounds:
-oxoacids
-oxoacids
-amino acids
5. Illustrate, using structural equations, the chemical
mechanisms of the following biochemical reactions.
a) Pyruvate acetaldehyde + CO
2
b) 2-Pyruvate -acetolactate + CO
2
c) Pyruvate + NAD
+
+ CoA-SH acetyl-CoA +
NADH + H
+
+ CO
2
d) Fructose 6-P + glyceraldehyde-3-P
xylulose-5-P + erythrose-4-P
6. 3-Fluoropyruvate is converted quantitatively by
pyruvate decarboxylase from wheat germ into
acetate, fluoride (F

), and carbon dioxide. Propose


a reaction mechanism. See Gish, G., Smyth, T.,
and Kluger, R. (1988) J. Am. Chem. Soc. 110, 6230-
6234.
7. In E. coli L-cysteine is formed from L-serine and
the sulfide ion S
2
in a reaction that also requires
acetyl-CoA and is catalyzed by the consecutive
action of an acyl transferase and cysteine synthase.
Outline the mechanism of this conversion indicat-
ing participation of any essential coenzymes.
8. Illustrate, using structural equations, the chemical
mechanisms of the following biochemical reactions.
a) L-Glutamate + oxaloacetate 2-oxoglutarate +
L-aspartate
b) L-Serine pyruvate + NH
4
+
c) L-Serine + indole (from cleavage of indole-3-
glycerol phosphate) L-tryptophan
d) L-Serine + tetrahydrofolate glycine + N
5
,
N
10
-methylene-tetrahydrofolate
e) L-Selenocysteine L-alanine + Se
0
9. Threonine is formed by E. coli from homoserine
via the intermediate -phosphohomoserine.
Write out an abbreviated reaction sequence for
its conversion to L-threonine by the action of
threonine synthase.
10. Write out a plausible step-by-step mechanism by
which 1-aminocyclopropane-1-carboxylate syn-
thase (ACC synthase) of plant tissues can form
ACC from S-adenosylmethionine. This reaction
requires a specific cofactor
11. Tissues of the mammalian central nervous system
contain a pyridoxal phosphate-dependent gluta-
mate decarboxylase that catalyzes conversion of
Glu to -aminobutyrate (GABA), an inhibitory
synaptic transmitter. GABA is degraded by trans-
imination with -oxoglutarate as the acceptor to
yield succinic semialdehyde, which then is oxidized
to succinate by an NAD-linked dehydrogenase.
a) Show how these reactions can operate as a
shunt pathway that allows the citric acid cycle
to function without the enzymes -oxoglutarate
dehydrogenase and succinate thiokinase.
b) Is the shunt more or less efficient than the
normal cycle from the standpoint of energy
recovery? Explain.
COO
NH
3

+
764 Chapter 15. Coenzymes of OxidationReduction Reactions
Contents
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
1. Structure of Pterins
2. Folate Coenzymes
3. Dihydrofolate Reductase
Inhibitors
Structure and mechanism
4. Single-Carbon Compounds and Groups in
Metabolism
Serine as a C1 donor
The glycine decarboxylase synthetase system
Starting with formate or carbon dioxide
N
5
-Formyl-THF (leucovorin)
Catabolism of histidine and purines
Thymidylate synthase
Synthesis of methyl groups
E. Specialized Coenzymes of Methanogenic Bacteria
F. Quinones, Hydroquinones, and Tocopherols
1. Pyrroloquinoline Quinone (PQQ)
2. Copper Amine Oxidases
Topaquinone (TPQ)
Lysine tyrosylquinone (LTQ)
Tryptophan tryptophanylquinone (TTQ)
Three-dimensional structures
Mechanisms
3. Ubiquinones, Plastoquinones, Tocopherols, and
Vitamin K
4. Quinones as Electron Carriers
5. Vitamin K and -Carboxyglutamate Formation
6. Tocopherols (Vitamin E) as Antioxidants
G. Selenium-Containing Enzymes
References
Study Questions
Boxes
Box 15-A Nicotinic Acid and Nicotinamide
Box 15-B Riboflavin
Box 15-C Thioredoxin and Glutaredoxin
Box 15-D Folic Acid (Pteroylglutamic Acid)
Box 15-E Thymidylate Synthase, a Target Enzyme in
Cancer Chemotherapy
Box 15-F The Vitamin K Family
Box 15-G The Vitamin E Family: The Tocopherols
Box 15-H Glutathione Peroxidase and Abnormalities
of Red Blood Cells
Tables
Table 15-1 Some Biochemical Hydrogen Transfer
Reactions
Table 15-2 Some Dehydrogenation Reactions
Catalyzed by Flavoproteins
Table 15-3 Single-Carbon Compounds in Order of
Oxidation State of Carbon
Table 15-4 Selenium-Containing Proteins
802
802
803
804
805
805
808
808
808
810
810
810
810
813
813
815
815
816
816
817
817
817
817
818
819
820
822
822
828
835
769
783
786
802
812
821
823
826
766
784
808
825
767
768
768
770
771
771
772
774
775
775
776
777
777
778
779
779
780
780
780
783
783
788
788
789
792
794
794
795
795
796
798
799
799
799
799
800
802
A. Pyridine Nucleotide Coenzymes and
Dehydrogenases
1. Three-Dimensional Structures of Dehydrogenases
2. Stereospecificity and Mechanism
One step or two-step transfer?
Coenzyme and substrate analogs
Conformational changes during dehydrogenase action
Zinc-containing alcohol dehydrogenases
Other alcohol dehydrogenases and aldo-keto reductases
3. Dehydrogenation of Amino Acids and Amines
4. Glyceraldehyde-3-Phosphate Dehydrogenase and
the Generation of ATP in Fermentation Reactions
5. Reduction of Carboxyl Groups
6. Reduction of CarbonCarbon Double Bonds
7. Transient Carbonyl Groups in Catalysis
8. ADP Ribosylation and Related Reactions of NAD
+
and NADP
+
9. The Varied Chemistry of the Pyridine Nucleotides
Addition to NAD
+
and NADP
+
Modification of NADH in acid
Other reactions of pyridine nucleotides
B. The Flavin Coenzymes
1. Flavoproteins and Their Reduction Potentials
2. Typical Dehydrogenation Reactions Catalyzed by
Flavoproteins
3. More Flavoproteins
4. Modified Flavin Coenzymes
5. Mechanisms of Flavin Dehydrogenase Action
6. Half-Reduced Flavins
7. Metal Complexes of Flavins and
Metalloflavoproteins
8. Reactions of Reduced Flavins with Oxygen
C. Lipoic Acid and the Oxidative Decarboxylation
of -Oxoacids
1. Chemical Reactions of Lipoic Acid
2. Oxidative Decarboxylation of Alpha-Oxoacids
3. Other Functions of Lipoic Acid
4. Additional Mechanisms of Oxidative
Decarboxylation
Pyruvate oxidase
Pyruvate:ferredoxin oxidoreductase
Oxidative decarboxylation by hydrogen peroxide
Pyruvate formate-lyase reaction
5. Cleavage of -Oxoacids and Substrate-Level
Phosphorylation
Biological oxidation-reduction reactions rely upon many organic coenzymes and
transition metal ions. Left, a molecule of the hydrogen carrier pyrroloquinoline
quinone (PQQ, Fig. 15-23) is seen at the bottom of a 7-bladed propeller of a bacterial
methanol dehydrogenase. In adrenodoxin reductase (right) the reducing power of
NADPH is passed to FAD and then to the small redox protein adrenodoxin. In this
stereoscopic view the pyridine ring of the oxidized coenzyme NADP
+
and the tricyclic
flavin ring of FAD are seen stacked against each other in the center; the adenylate ends
stretching toward the top and bottom of the complex. From Ziegler and Schulz, (2000)
Biochemistry 39, 10986-10995.
765
The dehydrogenation of an alcohol to a ketone or
aldehyde (Eq. 15-1) is one of the most frequent biological
oxidation reactions. Although the hydrogen atoms
removed from the substrate are often indicated simply
as 2[H], it was recognized early in the twentieth century
that they are actually transferred to hydrogen-carrying
coenzymes such as NAD
+
, NADP
+
, FAD, and riboflavin
5'-phosphate (FMN). This chapter deals with these
coenzymes and also with a number of other organic
oxidationreduction coenzymes. They may be con-
sidered either as carriers of hydrogen or as carriers
of electrons (H = H
+
+ e

) in metabolic reactions.
When NAD
+
becomes reduced by dehydrogena-
tion of an alcohol, one of the hydrogen atoms removed
from the alcohol becomes firmly attached to the NAD
+
,
converting it to NADH. The other is released as a
proton (Eq. 15-2). Study of
2
H-labeled alcohols and
their oxidation by NAD
+
has shown that dehydrogenases
catalyze direct transfer to NAD
+
of the hydrogen that is
attached to carbon in the alcohol. There is never any
exchange of this hydrogen atom with protons of the
medium. At the same time, the hydrogen attached to
the oxygen of the alcohol is released into the medium
as H
+
:
Coenzymes of OxidationReduction Reactions
15
H C OH C O + 2[H]
(15-1)
The foregoing observations suggested that these
biological dehydrogenations may be viewed as removal
of a hydride ion (H

) together with a proton (H


+
) rather
than as removal of two hydrogen atoms. NAD
+
and
NADP
+
are regarded as hydride ion-accepting coen-
zymes. However, it has been impossible to establish
conclusively that the hydrogen atom and electron are
transferred simultaneously as H

. Transfer of the
hydrogen atom to or from these coenzymes may con-
ceivably be followed by or preceded by transfer of an
electron. The situation is even less clear for the flavin
coenzymes FAD and riboflavin phosphate for which
intermediate free radical oxidation states are known
to exist. However, regardless of the actual mechanism,
it is convenient to classify most hydrogen transfer
reactions of metabolism as if they occurred by transfer
of a hydride ion. The hydride ion can be regarded as a
nucleophile which can add to double bonds or can be elimi-
nated from substrates in reactions of types that we have
already considered. Some of these reactions are listed
in Table 15-1.
Why are there four major hydrogen transfer coen-
zymes, NAD
+
, NADP
+
, FAD, and riboflavin phosphate
(FMN), instead of just one? Part of the answer is that
the reduced pyridine nucleotides NADPH and
NADH are more powerful reducing agents than are
reduced flavins (Table 6-7). Conversely, flavin coen-
zymes are more powerful oxidizing agents than are
(15-2)
H C O C O + NADH NAD
+
+ + H
+
This hydrogen is released as a proton, H
+
H
This H is transferred directly to NAD
+
766 Chapter 15. Coenzymes of OxidationReduction Reactions
NAD
+
and NADP
+
. This difference reflects the chemi-
cal difference between the vitamins riboflavin and
nicotinamide which form the oxidationreduction
centers of the coenzymes. Another difference is that
NAD
+
and NADP
+
tend to be present in free forms
within cells, diffusing from a site on one enzyme to a
site on another. These coenzymes are sometimes tightly
bound but flavin coenzymes are usually firmly bound
to proteins, fixed, and unable to move. Thus, they
Reaction Example (oxidant)
C
Dehydrogenation of an alcohol
Alcohol dehydrogenase (NAD
+
)
Dehydrogenation of an amine
Amino acid dehydrogenases, amine oxidases
(NAD
+
or flavin)
Dehydrogenation of adduct of thiol and aldehyde
Glyceraldehyde 3-phosphate dehydrogenase
(NAD
+
or NADP
+
)
D Dehydrogenation of acyl-CoA, acyl-ACP, or
carboxylic acid
Reduction of desmosterol to cholesterol by NADPH
Reduction of desmosterol to
cholesterol by NADPH
A
TABLE 15-1
Some Biochemical Hydrogen Transfer Reactions
a
Acyl-CoA dehydrogenases (Flavin)
Succinic dehydrogenase (Flavin)
(Y = SCoA or OH) Opposite: enoyl reductase (NADPH)
tend to accept hydrogen atoms from one substrate and
to pass them to a second substrate while attached to a
single enzyme.
The oxidationreduction potential of a pyridine
nucleotide coenzyme system is determined by the
standard redox potential for the free coenzyme (Table
6-8) together with the ratio of concentrations of oxi-
dized to reduced coenzyme ([NAD
+
] / [NADH], Eq.
6-64). If these concentrations are known, a redox
a
Reaction type 9 A,B of Table on the inside cover at the end of the book: A hypothetical hydride ion H

is transferred from the substrate to a


coenzyme of suitable reduction potential such as NAD
+
, NADP
+
, FAD, or riboflavin 5-phosphate. The reverse of hydrogenation is shown
for E. Many of the reactions are reversible and often go spontaneously in the reverse direction from that shown here.
HC NH
2
C NH H

+
C O
H
+
H
2
O
NH
4
+
HC OH H

+ C O
H
+
R SH C
O
C
OH
SR
C
O
SR
+
H
H

+
H
+
H
R C
H
H
C C Y
O
H
H
R C
H
C C Y
O
H
H +

H
+
C C H R

CH CH R +
H
+
H
H
E
B
767
potential can be defined for the NAD
+
system within
a cell. This potential may vary in different parts of the
cell because of differences in the [NAD
+
] / [NADH]
ratio, but within a given region of the cell it is constant.
On the other hand, the redox potentials of flavoproteins
vary. Since the flavin coenzymes are not dissociable,
two flavoproteins may operate at very different poten-
tials even when they are physically close together.
Why are there two pyridine nucleotides, NAD
+
and NADP
+
, differing only in the presence or absence
of an extra phosphate group? One important answer
is that they are members of two different oxidation
reduction systems, both based on nicotinamide but
functionally independent. The experimentally measured
ratio [NAD
+
] / [NADH] is much higher than the ratio
[NADP
+
] / [NADPH]. Thus, these two coenzyme
systems also can operate within a cell at different
redox potentials. A related generalization that holds
much of the time is that NAD
+
is usually involved in
pathways of catabolism, where it functions as an oxidant,
while NADPH is more often used as a reducing agent in
biosynthetic processes. See Chapter 17, Section I for
further discussion.
A. Pyridine Nucleotide Coenzymes and
Dehydrogenases
In 1897, Buchner discovered
that yeast juice, prepared by
grinding yeast with sand and filter-
ing, catalyzed fermentation of sugar.
This was a major discovery which
excited the interest of many other
biochemists.
1
Among them were
Harden and Young, who, in 1904,
showed that Buchners cell-free yeast
juice lost its ability to ferment glucose
to alcohol and carbon dioxide when
it was dialyzed. Apparently, fer-
mentation depended upon a low-
molecular-weight substance that
passed out through the pores of the
dialysis membrane. Fermentation
could be restored by adding back
to the yeast juice either the concen-
trated dialysate or boiled yeast juice
(in which the enzyme proteins had
been destroyed). The heat-stable
material, which Harden and Young
called cozymase, was eventually
found to be a mixture of inorganic
phosphate ions, thiamin diphosphate,
and NAD
+
. However, characteriza-
tion of NAD
+
was not accomplished
until 1935.
Pure NADP
+
was isolated from red blood cells in
1934 by Otto Warburg and W. Christian, who had been
studying the oxidation of glucose 6-phosphate by
erythrocytes.
1a
They demonstrated a requirement for
a dialyzable coenzyme which they characterized and
named triphosphopyridine nucleotide (TPN
+
, but
now officially NADP
+
; Fig. 15-1). Thus, even before
its recognition as an important vitamin in human nutri-
tion, nicotinamide was identified as a component of
NADP
+
.
Warburg and Christian recognized the relationship
of NADP
+
and NAD
+
(then called DPN
+
) and proposed
that both of these compounds act as hydrogen carriers
through alternate reduction and oxidation of the pyri-
dine ring. They showed that the coenzymes could be
reduced either enzymatically or with sodium dithionite
Na
2
S
2
O
4
.
S
2
O
4
2
+ NAD
+
+ H
+
NADH + 2 SO
2
The reduced coenzymes NADH and NADPH were
characterized by a new light-absorption band at 340
nm. This is not present in the oxidized forms, which
absorb maximally at 260 nm (Fig. 15-2). The reduced
forms are stable in air, but their reoxidation was found
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
(15-3)
Figure 15-1 The hydrogen-carrying coenzymes NAD
+
(nicotinanide adenine
dinucleotide) and NADP
+
(nicotinamide adenine dinucleotide phosphate).
We use the abbreviations NAD
+
and NADP
+
, even though the net charge on
the entire molecule at pH 7 is negative because of the charges on oxygen atoms
of the phospho groups.
N
C
O
NH
2
N
H H
C NH
2
O
O
OH OH
CH
2
H
2
C
O
OH OH
N
N
N
N
NH
2
O
P
O
P
O

O
O
O
+
2'
4
4
H

OPO
3
2
AMP
in NADP
+
NAD
+
A hydride ion,
H

adds here
during reduction
The vitamin,
nicotinamide
Note -glycosidic
linkage
d-Ribose
Structure of reduced
nicotinamide ring
768 Chapter 15. Coenzymes of OxidationReduction Reactions
to be catalyzed by certain yellow enzymes which were
later identified as flavoproteins.
1. Three-Dimensional Structures of
Dehydrogenases
Most NAD
+
- or NADP
+
- dependent dehydrogenases
are dimers or trimers of 20- to 40-kDa subunits. Among
them are some of the first enzymes for which complete
structures were determined by X-ray diffraction
methods. The structure of the 329-residue per subunit
muscle (M
4
) isoenzyme of lactate dehydrogenases
(see Chapter 11) from the dogfish was determined to
0.25 nm resolution by Rossmann and associates in
1971.
2 4
More recently, structures have been determined
for mammaliam muscle and heart type (H
4
) isoen-
zymes,
5
for the testicular (C
4
) isoenzyme from the
mouse,
6
and for bacterial lactate dehydrogenases.
6 8
In all of these the polypeptide is folded nearly identi-
cally. The structures of the homologous cytosolic and
mitochondrial isoenzymes of malate dehydrogenase
are also similar,
911
as are those of the bacterial en-
zyme.
12,13
All of these proteins consist of two structural
domains and the NAD
+
is bound to the nucleotide-
binding domain in a similar manner as is shown in
Fig. 2-13 for glyceraldehyde phosphate dehydrogenase.
The coenzyme-binding domains of the dehydrogenases
of known structures all have this nearly constant
structural feature (often called the Rossmann fold)
consisting of a six-stranded parallel sheet together
with several helical coils
14
(Figs. 2-13 and 2-27).
The coenzyme molecule curls around one end of
the nucleotide-binding domains in a C conformation
with the nicotinamide ring lying in a pocket (Figs. 2-13,
15-3). Even before the crystal structure of lactate dehy-
drogenase was known, the lack of pH dependence of
coenzyme binding from pH 5 to 10 together with
observed inactivation by butanedione suggested that
the pyrophosphate group of NAD
+
binds to a guani-
dinium group of an arginine residue. This was identi-
fied by X-ray diffraction studies as Arg 101. This ion
pairing interaction, as well as the hydrogen bond
between Asp 53 and the 2' oxygen atom of a ribose ring
(Fig. 15-3), is present in all of the lactate and malate
dehydrogenases.
The adenine ring of the coenzyme is bound in a
hydrophobic pocket with its amino group pointed out
into the solvent. A second structural domain holds
additional catalytic groups needed to form the active site.
2. Stereospecificity and Mechanism
When NAD
+
is reduced in
2
H
2
O by dithionite
(Eq. 15-3) an atom of
2
H is introduced into the reduced
pyridine. Chemical degradation showed the
2
H to be
present at the 4 position of the ring para to the nitrogen
atom
15
(see Fig. 15-1). As shown by Westheimer and
associates, during enzymatic reduction of NAD
+
by
deuterium-containing ethanol, CH
3
C
2
H
2
OH, one of
the
2
H atoms is transferred into the NADH formed,
thus establishing the direct transfer of a hydrogen atom.
16,17
When the NAD
2
H formed in this way is reoxidized
enzymatically with acetaldehyde, with regeneration
of NAD
+
and ethanol, the
2
H is completely removed.
This was one of the first recognized examples of
the ability of an enzyme to choose between two iden-
tical atoms at a pro-chiral center (Chapter 9). The two
sides of the nicotinamide ring of NAD were designated
A and B and the two hydrogen atoms at the 4 position
of NADH as H
A
(now known as pro-R) and H
B
(pro-S).
Alcohol dehydrogenase always removes the pro-R
hydrogen. Malate, isocitrate, lactate, and D-glycerate
dehydrogenases select the same hydrogen. However,
Figure 15-2 Absorption spectra of NAD
+
and NADH.
Spectra of NADP
+
and NADPH are nearly the same as these.
The difference in absorbance between oxidized and reduced
forms at 340 nm is the basis for what is probably the single
most often used spectral measurement in biochemistry.
Reduction of NAD
+
or NADP
+
or oxidation of NADH or
NADPH is measured by changes in absorbance at 340 nm in
many methods of enzyme assay. If a pyridine nucleotide is
not a reactant for the enzyme being studied, a coupled
assay is often possible. For example, the rate of enzymatic
formation of ATP in a process can be measured by adding to
the reaction mixture the following enzymes and substrates:
hexokinase + glucose + glucose-6-phosphate dehydrogenase
+ NADP
+
. As ATP is formed, it phosphorylates glucose via
the action of hexokinase. NADP
+
then oxidizes the glucose
6-phosphate that is formed with production of NADPH,
whose rate of appearance is monitored at 340 nm.
M
o
l
a
r

a
b
s
o
r
p
t
i
v
i
t
y
,


1
0
-
3
NADH
NAD
+
Wave number, cm
1
10
-3
Wavelength, nm
20 24 28 32 36 40 44 48
500 400 350 300 250 220
18
15
12
9
6
3
0
769
BOX 15-A NICOTINIC ACID AND NICOTINAMIDE
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
Nicotinic acid was prepared in 1867 by oxida-
tion of nicotine. Although it was later isolated by
Funk and independently by Suzuki in 19111912
from yeast and rice polishings, it was not recog-
nized as a vitamin. Its biological significance was
established in 1935 when nicotinamide was identi-
fied as a component of NAD
+
by von Euler and
associates and of NADP
+
by Warburg and Christian.
a
Both forms of the vitamin are stable, colorless com-
pounds highly soluble in water.
In 1937, Elvehjem and coworkers demonstrated
that nicotinic acid cured canine blacktongue. In
the same year it was found to cure human pellagra,
a terrible disease characterized by weakness, indi-
gestion, and loss of appetite followed by dermatitis,
a
Schlenk, F. (1984) Trends Biochem. Sci. 9, 286 288
b
Wagner, A. F., and Folkers, K. (1964) Vitamins and Coenzymes,
Wiley (Interscience), New York (p. 73)
c
Rosenkrantz, B. G. (1974) Science 183, 949 950
d
Teply, L. J. (1993) FASEB J. 7, 1300
e
Sauberlich, H. E. (1987) in Pyridine Nucleotide Coenzymes:
Chemical, Biochemical and Medical Aspects, Vol. B (Dolphin, D.,
Avramovic, O., and Poulson, R., eds), pp. 599 626, Wiley
(Interscience), New York
Figure 15-3 Diagrammatic structure of NAD
+
and L-lactate bound into the active site of lactate dehydrogenase. See
Eventhoff et al.
5
diarrhea, mental disorders, and eventual death. At
that time pellagra was common in the United States,
especially in the south. The U.S. Public Health
Service estimated that during 19121916 there
were 100,000 victims and 10,000 deaths a year.
b,c
The daily requirement for an adult is about
7.5 mg. The amount is decreased by the presence
in the diet of tryptophan, which can be converted
partially to nicotinic acid (Chapter 25).
d,e
Trypto-
phan is about 1/60 as active as nicotinic acid itself.
The one-time prevalence of pellagra in the southern
United States was a direct consequence of a diet
high in maize whose proteins have an unusually
low tryptophan content.

O
OH C
N
Nicotinic acid
O
C
N
Nicotinamide
NH
2
31
O
N
H
H
O
P
P
O
O O
O
O
H
O
O
O
H
H
O
O

O
C
29
D53
Adenine
OH
N
+
O
N
H
H
R101 N
H
N
H
H
O
O
O
N
H
H

+
H
R171
H195
H
H
CH
3
O
O
H
O
H
N
N
H
H
H
N
H
N
N
H H
+ H
2
N
N
H
N
D168
C
O
O

R109
O
N140
NH
2
LOOP
N
O
99
+
770 Chapter 15. Coenzymes of OxidationReduction Reactions
dehydrogenases acting on glucose 6-phosphate, gluta-
mate, 6-phosphogluconate, and 3-phosphoglyceralde-
hyde remove the pro-S hydrogen. By 1979 this stereo-
specificity had been determined for 127 dehydrogenases,
about half of which were found A-specific and half
B-specific.
18
Isotopically labeled substrates
have usually been used for this purpose but a simple
NMR method has been devised.
19,20
How complete is the stereospecificity? Does the
enzyme sometimes make a mistake? Lactate dehydro-
genase displays nearly absolute specificity, transferring a
proton into the incorrect pro-S position no more than
once in 5 x 10
7
catalytic cycles.
21
This suggests a differ-
ence in transition-state energies of about 40 kJ / mol for
the two isomers. X-ray structural studies suggested
that a major factor determining this stereoselectivity
might be the location of hydrogen-bonding groups
that hold the CONH
2
group of the nicotinamide. If
these are located in such a way that the nicotinamide
ring has a syn orientation with respect to the ribose
ring to which it is attached (Fig. 15-4), the A face of the
coenzyme will be against the substrate undergoing
oxidation or reduction. If the ring has an anti confor-
mation, the B side will be against the substrate.
18,22
However, the explanation cannot be this simple. High
specificity is still retained when NADH is modified,
for example, by replacement of the CONH
2
on the
pyridine ring with COCH
3
or CHO or by use of
-NADH, in which the ribosenicotinamide linkage
is instead of as in normal NADH.
23
An intriguing idea is that the enzyme selectively
stabilizes one of the boat conformations of NADH or
NADPH. According to stereoelectronic principles the
axially oriented hydrogen in such a boat structure
(pro-R in Fig. 15-4) will be the most readily transferred.
On the basis of this principle, together with the idea
that enzymes adjust the Gibbs energies of intermediate
states to achieve optimum catalytic rates, the following
prediction was made by Benner and associates:
24
The
thermodynamically most easily reduced carbonyl compounds
will react by enzymatic transfer of the hydride ion from the
pro-R position of NADH. Conversely, the most difficultly
reduced carbonyl compounds will receive the hydride
ion from the pro-S position of NADH. The proposal
has been controversial and it has been argued that
evolutionary relationships play a dominant role in
determining stereoselectivity.
23
Theoretical computa-
tions suggest that the boatlike puckering of the ring is
N
CONH
2
H
B
H
A
1
2
3
4
5
6
pro-S pro-R
flexible, raising some doubts about the rigidity of the
reactants in the active site.
12,25
Nevertheless, it is likely
that the coenzyme as well as bound substrates must
assume very specific conformations before the enzyme
is able to move to the transition state for the reaction.
A great deal of effort has been expended in trying to
understand other factors that may explain the high
stereospecificity of dehydrogenases.
12,22,26 28
When a hydrogen atom is transferred by an enzyme
from the 4 position of NADH or NADPH to an aldehyde
or ketone to form an alcohol, the placement of the
hydrogen atom on the alcohol is also stereospecific.
Thus, alcohol dehydrogenase acting on NAD
2
H con-
verts acetaldehyde to (R)-mono-[
2
H]ethanol (Eq. 9-73).
Pyruvate is reduced by lactate dehydrogenase to
L-lactate, and so on. Even mutations that disrupt the
binding of the carboxylate of lactate, e.g., substitution
of arginine 171 (see Fig. 15-3) by tryptophan or phenyl-
alanine and introduction of a new arginine on the other
side of the active site, do not alter the L-stereospecificity.
However, the specificity for lactate is lost.
29
One step or two-step transfer? Another major
question about dehydrogenases is whether the hydro-
gen atom that is transferred moves as a hydride ion,
as is generally accepted, or as a hydrogen atom with
separate transfer of an electron and with an intermedi-
ate NAD or NADPH free radical. In one study para-
substituted benzaldehydes were reduced with NADH
and NAD
2
H using yeast alcohol dehydrogenase as a
catalyst.
30
This permitted the application of the Ham-
mett equation (Box 6-C) to the rate data. For a series
of benzaldehydes for which
+
varied widely, a value
H
S
H
R
N
C
O
N
H
H
O
CH
2
OH HO
Figure 15-4 The nicotinamide ring of NADH in a syn boat
conformation suitable for transfer of an axially oriented pro-R
hydrogen atom from its A face as H

. The flow of electrons


is shown by the solid arrows. The dashed arrows indicate
competing resonance which favors planarity of the ring and
opposes the H

transfer. Hydrogen bonds from the protein


to the carboxamide group (dashed lines) affect both this
tendency and the conformation of the nucleotide.
771
of = +2.2 was observed for the rate constant with
both NADH and NAD
2
H. Thus, electron-accepting
substituents in the para position hasten the reaction.
While the significance of this observation is not im-
mediately obvious, the relatively low value of is pro-
bably incompatible with a mechanism that requires
complete transfer of a single electron from NADH to
acetaldehyde in the first step. A primary isotope effect
on the rates was k
H
/k
D
= 3.6, indicating that the CH
bond in NADH is broken in the rate-limiting step.
The fact that the isotope effect is the same for all of the
substituted benzaldehydes also argued in favor of a
hydride ion transfer.
Studies of the kinetics of nonenzymatic model
reactions of NADH with quinones
31,32
have also been
interpreted to favor a single-step hydride ion transfer.
Application of Marcus theory (Chapter 9, Section D,4)
to data from model systems also supports the hydride
transfer mechanism.
33,34
Quantum mechanical tunneling
may be involved in enzymatic transfer of protons,
hydride ions, and electrons.
3536
Tunneling is often
recognized by unusually large primary or secondary
kinetic isotope effects. According to semi-classical theory
(Chapter 12), the maximum effects for deuterium and
tritium are given by the following ratios:
( k
2
H
/k
1
H
) = ( k
3
H
/k
2
H
)
3.3
7.
Higher ratios, which suggest tunneling, are frequently
observed for dehydrogenases.
36a,b
Tunneling is apparently
coupled to fluctuations in motion within the enzyme-
substrate complex. Study of effects of pressure on
reactions provides a new approach that can aid
interpretation.
36c,36d
Coenzyme and substrate analogs. The structures
of enzymeNAD
+
substrate complexes (Fig. 15-3) may
be studied by X-ray crystallography under certain
conditions or can be inferred from those of various
stable enzyme-inhibitor complexes or from enzyme
reconstituted with NAD
+
that has been covalently
linked to the substrate. In the following diagram,
27
NAD
+
(I) is shown with L-lactate lying next to its A
face, ready to transfer a hydride ion to the pro-R posi-
tion in NADPH. Also shown (II) is 5-(S)-lac-NAD
+
,
with the covalently linked lactate portion in nearly the
same position as in diagram I. This NAD derivative
was used to obtain the first 0.27-nm structure with a
bound substrate-like molecule in lactate dehydro-
genase.
37
Since then the structures of many complexes
with a variety of dehydrogenases have been studied.
In coenzyme analog III the ring is bound to ribose
with a C C bond and it lacks the positive charge on
the nicotinamide ring in NAD
+
. It does not react with
substrate. However, it binds to the coenzyme site of
alcohol dehydrogenase and forms with ethanol a
ternary complex whose structure has been solved.
38
A related approach is to study complexes formed
with normal NAD
+
but with an unreactive second
substrate. An example is oxamate, which binds well
to lactate dehydrogenase to form stable ternary com-
plexes for which equilibrium isotope effects have been
studied.
39
In the structure of the lactate dehydrogenase active
site shown in Fig. 15-3, the lactate carboxylate ion is
held and neutralized by the guanidinium group of Arg
171, and the imidazole group of His 195 is in position
to serve as a general base catalyst to abstract a proton
from the hydroxyl group of the substrate (Eq. 15-4).
This imidazole group is also hydrogen bonded to the
carboxylate of Asp 168 as in the charge-relay system
of the serine proteases (Fig. 12-10). The same features
are present in the active site of malate dehydrogenases
and have been shown essential by study of various
mutant forms.
11,40 42
The His:Asp pair of the dehydro-
genases is not part of the nucleotide-binding domain
but is present in the second structural domain, the
catalytic domain. This is another feature reminiscent
of the serine proteases. A bacterial D-glycerate dehydro-
genase also has a similar structure and the same cata-
lytic groups. However, the placement of the catalytic
histidine and the arginine that binds the -carboxylate
group of the substrate is reversed, allowing the enzyme
to act on the D-isomer.
43
Conformational changes during dehydrogenase
action. Dehydrogenases bind coenzyme and substrate
in an ordered sequence. The coenzyme binds first,
then the oxidizable or reducible substrate. The binding
of the coenzyme to lactate dehydrogenase is accom-
panied by a conformational change by which a loop,
involving residues 98 120 and including one helix,
N
R
C
O
NH
2
N
R
C
O
NH
2
C
O
H
C
O O
N
R
C
O
NH
2
C
O
H
C
O O
5
CH
3
H
3
+ +
R = Ribose-P-P-adenosine
I II III
5-(S)-lac-NAD
+
,
(3S)-5-(3-carboxy-3
hydroxypropyl)-NAD
+
Lactate
NAD
+
H

3
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
C C
O
H
2
N
O
O

Oxamate
772 Chapter 15. Coenzymes of OxidationReduction Reactions
(15-5)
HN NH
HN
R171
H H
O
C
O
C
H
2
C
H
O H
N N
H
O
O
D168
H195
C
O
O
H
H
HN
NH
HN
R102
N
C
NH
2
O
HN
H
2
N
NH
2
R109

HN NH
HN
R171
H H
O
C
O
C
H
2
C
H
O H
N N
H
O
O
D168
H195
C
O
O
H
H
HN
NH
HN
R102

NAD
+
+

N
C
NH
2
O
+
+

+
NADH
+
H
+
HN
H
2
N
NH
2
R109
+
folds over the coenzyme like a lid.
41,44
This conforma-
tional change must occur during each catalytic cycle,
just as in the previously discussed cases of citrate
synthase (Chapter 13) and aspartate aminotransferase
(Chapter 14). One effect of folding of the loop is to
bring the side chain of Arg 109 into close proximity
to His 195 and to the OH group of the bound lactate
(Eq. 15-4). The closing of the loop also forces the
positively charged nicotinamide ring more deeply into
a relatively nonpolar pocket. This may induce a move-
ment of the positive charge toward the 4'
carbon of the ring, assisting in the transfer
of the hydride ion (Eq. 15-4).
41
At the end of
this transfer both Arg 109 and His 196 are
positively charged and electrostatic repulsion
between them may help to move the loop
and release the products. If the reaction pro-
ceeds in the opposite direction the presence
of two positive charges will assist in the
hydride ion transfer. The importance of Arg
109 is demonstrated by the fact that a mutant
with glutamine in place of Arg 109 has a value
of k
cat
only 1/ 400 that of native enzyme.
45
The loop closure also causes significant
changes in the Raman spectra of the bound
NAD
+
, especially in vibrational modes that
involve the carboxamide group of the nicoti-
namide ring. These have been interpreted as
indicating an increased ridigity of binding of
the coenzyme in the closed conformation.
46
As with many other enzymes acting on polar
substrates, a characteristic of the pretransition
state complex appears to be formation of a
complex with a network of hydrogen bonds
extending into the protein, exclusion of most
water molecules, and tight packing of protein
groups around the substrate. It is also signi-
ficant that an overall net electrical charge on
the ES complexes must be correct for tight
binding of substrates to occur for lactate or
malate dehydrogenase.
44
Positive and negative
charges are balanced except for one excess posi-
tive charge which may be needed for catalysis.
Zinc-containing alcohol dehydro-
genases. Liver alcohol dehydrogenase is a
relatively nonspecific enzyme that oxidizes
ethanol and many other alcohols. The much
studied horse liver enzyme
4760
is a dimer of
374-residue subunits, each of which contains
a catalytic Zn
2+
ion deeply buried in a crevice
between the nucleotide-binding and catalytic
domains. The enzyme also contains a struc-
tural Zn
2+
ion
48
that is bound by four sulfur
atoms from cysteine side chains but does not
represent a conserved feature of all alcohol
dehydrogenases. The catalytic Zn
2+
is ligated
by sulfur atoms from cysteines 46 and 174 and by a
nitrogen atom of the imidazole group of His 67 (Fig. 15-
5). In the free enzyme a water molecule is thought to
occupy the 4th coordination position and its dissocia-
tion to form the Zn
+
HO

complex (Eq. 15-5) may


account for a pK
a
of 9.2 in the free enzyme and of 7.6
in the NAD
+
complex.
48
The apparent pK
a
drops
further to about 6.4 in the presence of the substrate
(15-4)
Zn
2+
OH
2
Zn
+
OH + H
+
773
ethanol. The assignment of this pK
a
value has been
controversial.
48,49
Histidine 51 (Fig. 15-5), Glu 68 and
Asp 49 have side chains close to the zinc and, as we have
seen (Chapter 7), macroscopic pK
a
values of proteins
can be shared by two or more closely placed groups.
Substrate binding also induces a conformational
change in this enzyme. When both coenzymes and
substrate bind the closed conformation of the enzyme
is formed by a rotation of the catalytic domains of the
two subunits relative to the coenzyme-binding do-
mains.
5051a
Structures of ternary complexes with inhibi-
tors and with substrates have also been established.
For example, liver alcohol dehydrogenase was crystal-
lized as the enzymeNAD
+
p-bromobenzyl alcohol
complex with saturating concentrations of substrates
in an equilibrium mixture
51b
and studied at low reso-
lution. Transient kinetic studies or direct spectroscopic
determinations led to the conclusion that the internal
equilibrium (ENAD
+
alcohol = ENADHaldehyde)
favors the NAD
+
alcohol complex.
52
Subsequently,
the complex was studied at higher resolution, and
the basic structural features were confirmed with a
Figure 15-5 Structure of the complex of horse liver alcohol dehy-
drogenase with NAD
+
and the slow substrate p-bromobenzyl alcohol.
The zinc atom and the nicotinamide ring of the bound NAD
+
are
shaded. Adjacent to them is the bound substrate. Courtesy of Bryce
Plapp. Based on Ramaswamy et al.
53
F198
I224
D223
K228
O
O
H
H
O O
O
O
O
H
3
N
O
O P
P
O
O
O
N
N
N
N
NH
2
H
2
N
H
2
N
N
+
N
N
R271
O
O
I269
H
O
O
O
O
H
H
H
N
H
H
N
H
H51
S
S48
S
C46
C 174
O
N
Zn
NH
O
O
H67
F93
L141
L57
Br
V294
I318
N
O
+
S M306
NH
HN
H
R47
+

R369

H
D49
H
H
H
H
H
O 317
O 292
N
H
N
H
H
O
H
N H
H
+
E68
N
N
O
O
H
H

H
H
N
2
structure of the enzyme complexed with NAD
+
and
2,3,4,5,6-pentafluorobenzyl alcohol.
53
From the crystal
structures of the NAD
+
p-bromobenzyl alcohol com-
plex and the previously mentioned complex with
ethanol and analog III it appears that the oxygen of the
alcohol substrate coordinates with the Zn
2+
, displacing
the bound water. Binding of the chromophoric aldehyde
4-trans-(N, N-dimethylamino) cinnamaldehyde shifts
the wavelength of maximum absorbance by 66 nm,
suggesting that Zn
2+
binds directly to the
oxygen of this ligand (Eq. 15-6).
54
Reso-
nance-enhanced Raman spectroscopy (Chap-
ter 23) of the complex of dimethylamino-
benzaldehyde with alcohol dehydrogenase
also supports an intermediate in which the
Zn
2+
becomes coordinated directly with the
substrate oxygen (Eq. 15-6).
55
Rapid scanning
spectrophotometry of complexes with an-
other chromophoric substrate, 3-hydroxy-4-
nitrobenzyl alcohol, also suggested that the
alcohol first formed a complex with an undis-
sociated alcoholic OH group (Eq. 15-7, step
a) and then lost a proton to form a zinc
alcoholate complex (step b)
56
which could
react by hydride ion transfer (step c).
Eklund et al. suggested that the side
chains of Ser 48 and His 51 act as a proton relay
system to remove the proton from the alcohol,
in step b of Eq. 15-7, leaving the transient
zinc-bound alcoholate ion, which can then
transfer a hydride ion to NAD
+
, in step c.
52
The shaded hydrogen atom leaves as H
+
.
The role of His 51 as a base is supported by
studies of the inactivation of the horse liver
enzyme by diethyl pyrocarbonate
57
and by
directed mutation of yeast and liver enzymes.
When His 51 was substituted by Gln the pK
a
of 7 was abolished and the activity was
decreased ten-fold.
58
The functioning of zinc ions in enzymes
has been controversial and other mechanisms
have been proposed. Makinen et al. suggested
a transient pentacoordinate Zn
2+
complex
on the basis of EPR measurements on en-
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
N
H
3
C
H
3
C
C
H
O
N
H
3
C
H
3
C
C
H
O Zn
+
+
Zn
++
(15-6)
774 Chapter 15. Coenzymes of OxidationReduction Reactions
zyme containing Co
2+
. Such a complex, in which the
side chain of nearby Glu 68 would participate, would
allow the coordinated water molecule to act as the
base in deprotonation of the alcohol.
59
Molecular
dynamics calculations indicate that Glu 68 can coordi-
nate the zinc ion in this fashion, but Ryde suggests that
its function may be to assist the exchange of ligands,
i.e., the release of an alcohol or aldehyde product.
60
A
variety of kinetic and spectroscopic studies have pro-
vided additional information that makes alcohol dehy-
drogenase one of the most investigated of all enzymes.
Liver alcohol dehydrogenase is important to the
metabolism of ethanol by drinkers. Human beings
exhibit small individual differences in their rates of
alcohol metabolism which may reflect the fact that
there are several isoenzymes and a number of genetic
variants whose distribution differs from one person
to the next as well as among tissues.
61 64
Inhibition of
these isoenzymes by uncompetitive inhibitors, discussed
in Chapter 9, is a goal in treatment of poisoning by meth-
anol or ethylene glycol. Inhibition of the dehydrogenases
slows the two-step oxidation of these substrates to toxic
carboxylic acids.
65
Yeast contains two cytosolic alcohol dehydrogen-
ase isoenzymes.
66
Alcohol dehydrogenase I, present
in large amounts in cells undergoing fermentation,
functions to reduce acetaldehyde in the fermentation
process. Alcohol dehydrogenase II is synthesized by
cells growing on such carbon sources as ethanol itself
and needing to oxidize it to obtain energy. A third
isoenzyme is present in mitochondria. An alcohol
dehydrogenase isoenzyme of green plants is induced
by anaerobic stress such as flooding. It permits ethan-
olic fermentation to provide energy temporarily to
submerged roots and other tissues.
67
Some bacteria
contain an NADP
+
-dependent, Zn
2+
-containing alcohol
dehydrogenase.
67a
Other alcohol dehydrogenases and aldo-keto
reductases. The oxidation of an alcohol to a carbonyl
compound and the reverse reaction of reduction of a
carbonyl group are found in so many metabolic path-
ways that numerous specialized dehydrogenases exist.
A large group of short-chain dehydrogenases and
reductases had at least 57 known members by 1995.
6869a
Their structures and functions are variable. Most
appear to be single-domain proteins with a large
sheet, a nucleotide-binding pocket, and a conserved
pair of residues: Tyr 152 and Lys 156. These may
function in a manner similar to that of the His-Asp
pair in lactate dehydrogenase. A possible role of a
cysteine side chain has been suggested for another
member of this group, 3-hydroxyisobutyrate dehydro-
genase, an enzyme of valine catabolism.
70
Dehydrogenases often act primarily to reduce a
carbonyl compound rather than to dehydrogenate an
alcohol. These enzymes may still be called dehydro-
genases. For example, in the lactic acid fermentation
lactate is formed by reduction of pyruvate but we still
call the enzyme lactate dehydrogenase. In our bodies
this enzyme functions in both directions. However, some
enzymes that act mainly in the direction of reduction
are called reductases. An example is aldose reductase,
a member of a family of aldo-keto reductases
71 73
which have ( /)
8
-barrel structures.
74 76
The normal physiological function of aldose reduc-
tase is uncertain but it can cause a problem in diabetic
persons by reducing glucose to sorbitol (glucitol),
ribose to ribitol, etc. The resulting sugar alcohols
accumulate in the lens and are thought to promote
cataract formation and may also be involved in the
severe damage to retinas and kidneys that occurs in
diabetes mellitus. Inhibitors of aldose reductase delay
development of these complications in animals but the
compounds tested are too toxic for human use.
76
The active sites of aldo-keto reductases contain
an essential tyrosine (Tyr 48) with a low pK
a
value.
The nearby His 110, Asp 43, and Lys 77 may also
participate in catalysis.
76,77
The kinetics are unusual.
Both NAD
+
and NADH are bound tightly and the
overall rate of reduction of a substrate is limited by
the rate of dissociation of NAD
+
.
78
Citrate is a natural
uncompetitive inhibitor of aldose reductase.
79
a
O
O
H
H N
N
51
O
H
O
C 269
O
H
CH
2
S48
O
Zn
C
H
H
N
S
S
R
N
NH
2
O
O
O
H
H N
N
51
O
H
O
C 269
O
H
CH
2
S48
O
Zn
C
H
H
N
S
S
R
N
NH
2
O
+
H

+
Enzyme NAD
+
RCH
2
OH
b
H
+
c
R CH=O
Enzyme NADH (15-7)
775
3. Dehydrogenation of Amino Acids and
Amines
The dehydrogenation of an amine or the reverse
reaction, the reduction of a Schiff base (Table 15-1,
reaction type B), is another important pyridine nucle-
otide-dependent process. Glutamate dehydrogenase,
a large oligomeric protein whose subunits contain 450
or more residues, is the best known enzyme catalyzing
this reaction. An intermediate Schiff base of 2-oxo-
glutarate and NH
3
is a presumed intermediate.
80,81
Similar reactions are catalyzed by dehydrogenases for
alanine,
82,83
leucine,
84
phenylalanine,
85
and other amino
acids.
83,86,87
4. Glyceraldehyde-3-Phosphate Dehydrogenase
and the Generation of ATP in Fermentation
Reactions
The oxidation of an aldehyde to a carboxylic acid,
a highly exergonic process, often proceeds through a
thioester intermediate whose cleavage can then be
coupled to synthesis of ATP. This sequence is of central
importance to the energy metabolism of cells (Chapters
10 and 17) and is shown in Fig. 15-6.
The best known enzyme catalyzing the first step
of this reaction sequence is glyceraldehyde 3-phosphate
dehydrogenase which functions in the glycolytic
sequence (steps a and b of Fig. 10-3). It is present in
both prokaryotes and eukaryotes as a tetramer of
identical 36 - to 37-kDa subunits. Three-dimensional
structures have been determined for enzyme from
several species, including lobster,
47,88
E. coli,
89
the
thermophilic bacterium Bacillus stearothermophilus
(Fig. 2-13),
9090a
and trypanosomes.
91
Recall that alde-
hydes are in equilibrium with their covalent hydrates
(Eq. 15-8, step a). Dehydrogenation of such a hydrate
yields an acid (Eq. 15-8, step b) but such a mechanism
offers no possibility of conserving the energy available
from the reaction. However, during catalysis by glycer-
aldehyde-phosphate dehydrogenase the SH group of
Cys 149, in the first step (step a, Fig. 15-6), adds to the
substrate carbonyl group to form an adduct, a thio-
hemiacetal. This adduct is oxidized by NAD
+
to a
thioester, an S-acyl enzyme (step b), which is then
cleaved by the same enzyme through a displacement
on carbon by an oxygen atom of P
i
(phosphorolysis;
step c). The sulfhydryl group of the enzyme is released
simultaneously and the product, the acyl phosphate
1,3-bisphosphoglycerate, is formed. The imidazole
group of His 176 may catalyze both steps a and b.
89,92
A separate enzyme then transfers the phospho group
from the 1 position of 1,3-bisphosphoglycerate to ADP
to form ATP and 3-phosphoglycerate (step d). The
overall sequence of Fig. 15-6 is the synthesis of one
mole of ATP coupled to the oxidation of an aldehyde
R C
H
O
R C
H
OH
OH
R C
OH
O
H
2
O +
a
b
Covalent
hydrate 2[H]
to a carboxylic acid and the conversion of NAD
+
to
NADH.
In green plants and in some bacteria an NADP
+
-
dependent cytoplasmic glyceraldehyde 3-phosphate
dehydrogenase does not use inorganic phosphate to
form an acyl phosphate intermediate but gives 3-phos-
phoglycerate with a free carboxylate as in Eq. 15-8.
93,93a
Because it doesnt couple ATP cleavage to the dehy-
drogenation, it drives the [NADPH] / [NADP
+
] ratio
to a high value favorable to biosynthetic processes (see
Chapter 17).
(15-8)
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
Figure 15-6 Generation of ATP coupled to oxidation of an
aldehyde to a carboxylic acid. The most important known
example of this sequence is the oxidation of glyceraldehyde
3-phosphate to 3-phosphoglycerate (Fig. 10-3, steps a and b).
Other important sequences for substrate-level phosphory-
lation are shown in Eq. 14-23 and in Fig. 15-16.
R C
H
O R C
H
S
OH
E
R C
S
O
E
R C
O
O
P
R C
O
O

P
i
Dehydrogenase
Thioester
E SH
NAD
+
NADH
ADP
ATP
Phospho-
transferase
C
OH
CH
2
OP R=
H
for glyceraldehyde
3-phosphate dehydrogenase
(An SH group from
CoA or some other
compound may replace
ESH in some instances)
b
c
a
d
Acyl phosphate
776 Chapter 15. Coenzymes of OxidationReduction Reactions
Animal tissues also contain aldehyde dehydrogen-
ases of a nonspecific type which are thought to act to
remove toxic aldehydes from tissues.
94,95
Like glycer-
aldehyde 3-phosphate, these enzymes form thioester
intermediates as in Fig. 15-6 but which are hydrolyzed
rather than being converted to acyl phosphates. A
mutation (E487K) in the mitochondrial enzyme occurs
in about 50% of the Asian population. Although the
structural alteration is not at the active site, the enzyme
activity is low. Individuals carrying the mutation are
healthy but have an aversion to alcohol, whose con-
sumption causes an elevated blood level of acetalde-
hyde, facial flushing, dizziness, and other symptoms.
A similar effect is exerted by the drug disulfiram
(Antabuse), which has been used to discourage drink-
ing and whose metabolites are thought to inhibit alde-
hyde dehydrogenase.
95
Alcohol dehydrogenases also oxidize aldehydes,
probably most often as the geminal diol forms, accord-
ing to Eq. 15-8. No ATP is formed. The same enzymes
can catalyze the dismutation of aldehydes, with equal
numbers of aldehyde molecules going to carboxylic
acid and to the alcohol.
96 98
5. Reduction of Carboxyl Groups
The last two reaction steps in Fig. 15-6, steps c and
d, are in essence the reverse of the sequence used for
synthesis of a thioester such as a fatty acyl coenzyme
A. Thus, the chemistry by which ATP is generated
during glycolysis and that by which it is utilized in
biosynthesis is nearly the same. Furthermore, a stan-
dard biochemical method for reduction of carboxyl
groups to aldehyde groups is conversion, in an ATP-
requiring process, to a thioester followed by reduction
of the thioester (Table 15-1, reaction type C). For example,
the sequence of Fig. 15-6 is reversed during gluconeo-
genesis (see Fig. 17-17). The carboxyl group of the
side chain of aspartate can be reduced in two steps to
form the alcohol homoserine (Figs. 11-3; 24-13).
The aldehyde generated by reduction of a
thioester is not always released from the enzyme but
may be converted on to the alcohol in a second reduc-
tion step.
99
This is the case for 3-hydroxy-3-methyl-
glutaryl-CoA reductase (HMG-CoA reductase), a
large 887-residue protein that synthesizes mevalonate
(Eq. 15-9).
100 102
This highly regulated enzyme controls
the rate of synthesis of cholesterol and is a major target
of drugs designed to block cholesterol synthesis. The
structure of a smaller 428-residue bacterial enzyme is
known.
103,104
Aspartate 766 is a probable proton donor
in both reduction steps and Glu 558 and His 865 may
act as a catalytic pair that protonates the sulfur of
coenzyme A.
100
A related oxidation reaction is catalyzed by
glucose-6-phosphate dehydrogenase, the enzyme
that originally attracted Warburgs attention and led to
the discovery of NADP
+
. The substrate, the hemiacetal
ring form of glucose, is oxidized to a lactone which is
then hydrolyzed to 6-phosphogluconate (Eq. 15-10).
104a
This oxidation of an aldehyde to a carboxylic acid is
not linked directly to ATP synthesis as in Fig. 15-6.
The ring-opening step ensures that the reaction goes
to completion. This reaction is a major supplier of
NADPH for reductive biosynthesis and the large Gibbs
energy decrease for the overall reaction ensures that
the ratio [NADPH] / [NADP
+
] is kept high within
cells. This is the only source of NADPH for mature
erythrocytes and a deficiency of glucose 6-phosphate
dehydrogenase is a common cause of drug- and food-
OOC CH
2
C
CH
3
OH
CH
2
C
O
S CoA
2 NADPH + 2H

+
2 NADPH
+
CoASH
OOC CH
2
C
CH
3
OH
CH
2
CH
2
OH

(15-9)
(15-10)
HO
O
HO
CH
2
OH
OH
H
O P
O O
HO
CH
2
O P
OH
HO
C
C H
C
OH
HO H
C OH H
C H
CH
2
OP
Glucose 6-phosphate
OH
O O

6-Phosphogluconate
NADP
+
NADPH + H
+
H
2
O
Lactone
777
induced hemolytic anemia in human beings. About
400 variant forms of this enzyme are known.
105
Like
the sickle cell trait (Box 7-B) some mutant forms of
glucose-6-phosphate dehydrogenase appear to confer
resistance to malaria.
6. Reduction of CarbonCarbon Double
Bonds
Neither NADP
+
nor NAD
+
is a strong enough
oxidant to carry out the dehydrogenation of an acyl-CoA
(reaction type D of Table 15-1). However, NADPH or
NADH can participate in the opposite reaction. Thus,
NADPH transfers a hydride ion to the -carbon of an
unsaturated acyl group during the biosynthesis of
fatty acids (Chapter 17) and during elongation of
shorter fatty acids (Chapter 21).
106107b
A discovery
of medical importance is that isonicotinyl hydrazide
(INH), the most widely used antituberculosis drug,
forms an adduct (of an INH anion or radical) with
NAD
+
of long-chain enoyl-acyl carrier protein
reductase (enoyl-ACP reductase).
133a,b
This enzyme
utilizes NADH in reduction of a C = C double bond
during synthesis of mycolic acids. The same enzyme
is blocked by triclosan, an antibacterial compound
used widely in household products such as antiseptic
soaps, toothpastes, cosmetics, fabrics, and toys.
133c e
Less frequently NADPH is used to reduce an
isolated double bond. An example is the hydrogenation
of desmosterol by NADPH (Eq. 15-11), the final step
in one of the pathways of biosynthesis of cholesterol
(Fig. 22-7). In this and in other reactions of the same
type, hydrogen transfer has been shown to be from the
pro-S position in NADPH directly to C-25 of the sterol.
The proton introduced from the medium (designated
by the asterisks in Eq. 15-11) enters trans to the H

ion
from NADPH. The proton always adds to the more
electron-rich terminus of the double bond, i.e., it follows
the Markovnikov rule. This result suggests that proto-
nation of the double bond may precede H

transfer.
108
Additional pyridine nucleotide-dependent dehy-
drogenases include glutathione reductase (Figs.
15-10, 15-12), dihydrofolate reductase (Fig. 15-19),
isocitrate dehydrogenase, sn-glycerol-3-phosphate
dehydrogenase (Chapter 21), L-3-hydroxyacyl-CoA
dehydrogenase (Chapter 21), retinol dehydrogenase
(Chapter 23), and a bacterial quinone oxidoreduc-
tase.
109
Some of these also contain a flavin coenzyme.
7. Transient Carbonyl Groups in Catalysis
Some enzymes contain bound NAD
+
which oxi-
dizes a substrate alcohol to facilitate a reaction step
and is then regenerated. For example, the malolactic
enzyme found in some lactic acid bacteria and also in
Ascaris decarboxylates L-malate to lactate (Eq. 15-12).
This reaction is similar to those of isocitrate dehydro-
genase,
110 112
6-phosphogluconate dehydrogenase,
113
and the malic enzyme (Eq. 13-45)
114
which utilize free
NAD
+
to first dehydrogenate the substrate to a bound
oxoacid whose carbonyl group facilitates decarboxy-
lation. Likewise, the bound NAD
+
of the malolactic
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
O
OH
Cl Cl Cl
Triclosan
N
N
C
O
NH
2
H C
O
H
Adduct of isonicotinyl hydrazide
anion with NAD
+

OOC CH
2
C OH
COO

H
H
3
C C OH
COO

H
H
+
CO
2
+ +
l-Malate l-Lactate
(15-12)
HO
H
H
H
CH
3
H
CH
3
N
H H
S
C
O
NH
2
Desmosterol
*
25
*
NADPH
Cholesterol
+
(15-11)
778 Chapter 15. Coenzymes of OxidationReduction Reactions
enzyme apparently dehydrogenates the malate to a
bound oxaloacetate which is decarboxylated to pyru-
vate.
115
The latter remains in the active site and is
reduced by the bound NADH to lactate which is released
from the enzyme.
Another reason for introducing a carbonyl group
is to form a symmetric intermediate in a reaction that
inverts the configuration about a chiral center. An
example is UDP-galactose 4-epimerase, an enzyme
that converts UDP-galactose to UDP-glucose (Eq. 15-13;
Chapter 20) and is essential in the metabolism of galac-
tose in our bodies. The enzyme contains bound NAD
+
and forms a transient 4-oxo intermediate and bound
NADH. Rotation of the intermediate allows nonstereo-
specific reduction by the NADH, leading to epimeriza-
tion.
116117a
The enzyme is a member of the short-chain dehy-
drogenase group with a catalytic TyrLys pair in the
active site.
118
Another way that formation of an oxo
group can assist in epimerization of a sugar is through
enolization with nonstereospecific return of a proton
to the intermediate enediol. A third possible mechanism
of epimerization is through aldol cleavage followed
by aldol condensation, with inversion of configuration.
In each case the initial creation of an oxo group by
dehydrogenation is essential.
Bound NAD
+
is also present in S-adenosylhomo-
cysteine hydrolase,
119,120
which catalyzes the irrevers-
ible reaction of Eq. 15-14. Transient oxidation at the 3
position of the ribose ring facilitates the reaction. The
reader can doubtless deduce the function that has
been established for the bound NAD
+
in this enzyme.
However, the role of NAD in the urocanase reaction
(Eq. 15-15) is puzzling. This reaction, which is the
second step in the catabolism of histidine, following
Eq. 14-44, appears simple. However, there is no obvious
COO

N
NH
COO

N
NH
O
H
2
O
Urocanate
CH
2
O
H H
OH OH
H H
HO CH
2
O
OH OH
+
S
C

OOC
H
3
N H
SH
C

OOC
H
3
N H
H
2
O
Adenine
Adenine
H H
H H
S-Adenosyl-l-homocysteine
Adenosine
l-Homocysteine
+
3'
3'
(15-14)
(15-15)
mechanism and no obvious role for NAD
+
. See Frey
for a discussion.
117
8. ADP Ribosylation and Related Reactions
of NAD
+
and NADP
+
The linkage of nicotinamide to ribose in NAD
+
and NADP
+
is easily broken by nucleophilic attack on
C-1 of ribose. In Chapter 11 enzyme-catalyzed ADP
ribosylation, which can be shown as in Eq. 15-16, is
discussed briefly. The nucleophilic group Y from an
ADP-ribosyltransferase carries the ADP-ribosyl group
which can then be transferred by a second displace-
ment onto a suitable nucleophilic acceptor group.
121,122
Hydrolysis (Eq. 15-16, step a) gives free ADP-ribose.
123
Other known products of enzymatic action are indicated
in steps cf. Poly-(ADP ribosylation) is discussed
in Chapter 27. The structure of cyclic ADP-ribose
(cADPR)
124
is shown in Chapter 11, Section E,2. The
acceptor nucleophile is N-1 of the adenine ring which
is made more nucleophilic by electron donation from
the amino group. A similar reaction with ADP ribose
(Eq. 15-16, step e) produces a dimeric ADP-ribose
(ADPR)
2
125
while reaction with free nicotinic acid
(step f) yields, in an overall base exchange, nicotinic
acid adenine dinucleotide (NAADP
+
).
126,127
Some
of these compounds, e.g., cADPR, NAADP
+
, and
(ADPR)
2
, are involved in signaling with calcium
ions.
124
H
O
HO
CH
2
OH
OH
OH
O
P
O
P
O

O O

O O
Uridine
HO
O
HO
CH
2
H
OH
OH
O
P
O
P
O

O O

O O
Uridine
(15-13)
779
the cyanide adduct, whose absorption maximum
is at 327 nm, has been used to introduce deuterium
into the para position of the pyridine nucleotides.
In the adduct, the hydrogen adjacent to the highly
polarized C N bond is easily dissociated as a proton.
Thiolate anions and bisulfite also add. Dithionite ion,
S
2
O
4
2
, can lose SO
2
and acquire a proton to form the
sulfoxylate ion HSO
2

which also adds to the 4 position


of the NAD
+
ring.
130
The resulting adduct is unstable
and loses SO
2
to give NADH + H
+
. Addition can also
occur at the two ortho positions. The adducts of HO

to the 4 position of NAD


+
are stable but those to the
2 position undergo ring opening (Eq. 15-18) in base-
catalyzed reactions which are followed by further
degradation.
128,131
Another base-catalyzed reaction is the addition of
enolate anions derived from ketones to the 4 position
of the pyridine nucleotides (Eq. 15-19). The adducts
undergo ring closure and in the presence of oxygen
are converted slowly to fluorescent materials. While
forming the basis for a useful analytical method for
determination of NAD
+
(using 2-butanone), these
reactions also have created a troublesome enzyme
inhibitor from traces of acetone present in commercial
NADH.
132
N
R
C
O
NH
2
N
R
C
O
NH
2
Y H
Nucleophile, Y

This proton is
easily dissociated
in the CN

adduct
+
Adduct
(15-17)
9. The Varied Chemistry of the Pyridine
Nucleotides
Despite the apparent simplicity of their structures,
the chemistry of the nicotinamide ring in NAD
+
and
NADP
+
is surprisingly complex.
128,129
NAD
+
is ex-
tremely unstable in basic solutions, whereas NADH
is just as unstable in slightly acidic media. These prop-
erties, together with the ability of NAD
+
to undergo
condensation reactions with other compounds, have
sometimes caused serious errors in interpretation of
experiments and may be of significance to biological
function.
Addition to NAD
+
and NADP
+
. Many nucleo-
philic reagents add reversibly at the para (or 4) position
(Eq. 15-17) to form adducts having structures resem-
bling those of the reduced coenzymes. Formation of
N
R
C
O
NH
2
N
R
C
O
NH
2
OH
H
OH
OH

(15-18)
N
R
C
O
NH
2
N
R
C
H
O
CH
3
NH
2
O
N
R
C
N
CH
3
O
R
C
N
CH
3
N
O
2
C
O
CH
2
CH
3
+
+
Fluorescent
OH

(15-19)
A. Pyridine Nucleotide Coenzymes and Dehydrogenases
Nicotinamide
ADP-ribosyl-Y-Enzyme
H
2
O
ADP-ribose
Mono-ADP ribosylation
(Chapter 11)
Poly-(ADP ribosylation)
(Chapter 27)
Cyclic ADP-ribose
(cADPR, Chapter 11)
Dimeric
ADP-ribose
(ADPR)
2
Nicotinic acid
Base exchange
NAADP
(Nicotinic
acid adenine
dinucleotide)
b
c
e
a
d
f
ADP-
ribose
+
N
C
O
NH
2
CH
2
O P P Ribose
Adenine
O
HO OH
+
Y-Enzyme
(ADP-ribosyl
transferase
or cyclase)
NAD
+
(15-16)
780 Chapter 15. Coenzymes of OxidationReduction Reactions
The reactions of Eq. 15-19 occur nonenzymatically
only under the influence of strong base but dehydro-
genases often catalyze similar condensations relatively
rapidly and reversibly. Pyruvate inhibits lactate dehy-
drogenase, 2-oxoglutarate inhibits glutamate dehydro-
genase, and ketones inhibit a short-chain alcohol
dehydrogenase in this manner.
133,69a
Modification of NADH in acid. Reduced pyri-
dine nucleotides are destroyed rapidly in dilute HCl
and more slowly at pH 7 in reactions catalyzed by buffer
acids.
128,131,134
Apparently the reduced nicotinamide
ring is first protonated at C-5, after which a nucleophile
Y

adds at the 6 position (Eq. 15-20). The nucleophile


may be OH

, and the adduct may undergo further


reactions. For example, water may add to the other
double bond and the ring may open on either side of
the nitrogen. The glycosidic linkage can be isomerized
from to or can be hydrolytically cleaved. The early
steps in the modification reaction are partially revers-
ible, but the overall sequence is irreversible. One of
the products, which has been characterized by crystal
structure determination, is shown in Eq. 15-20.
128,135
It
can arise if the group Y of Eq. 15-20 is the C-2' hydroxyl
of the ribose ring and if the configuration of the glyco-
sidic linkage is inverted (anomerized).
The foregoing reactions have attracted interest
because glyceraldehyde-3-phosphate dehydrogenase,
in a side reaction, converts NADH to a substance
referred to as NADH-X which has been shown to be
the 6(R) adduct of Eq. 15-20, where Y is OH. In an
ATP-dependent reaction an enzyme from yeast recon-
verts NADH-X to NADH.
136
Mercury (II) ions can add in place of H
+
in the first
step of Eq. 15-20 and subsequent reactions similar to
those promoted by acid can occur.
137
Other reactions of pyridine nucleotides. Alka-
line hexacyanoferrate (III) oxidizes NAD
+
and NADP
+
to 2-,4-, and 6-pyridones. The 6-pyridone of N-methyl-
nicotinamide is a well-known excretion product of
nicotinic acid in mammals. Reoxidation of NADH and
NADPH to NAD
+
and NADP
+
can be accomplished
with hexacyanoferrate (III), quinones, and riboflavin
but not by H
2
O
2
or O
2
. However, O
2
does react at
neutral pH with uptake of a proton to form a peroxide
derivative of NADH.
138
When heated in 0.1 N alkali at
100C for 5 min, NAD
+
is hydrolyzed to nicotinamide
and adenosine-diphosphate-ribose.
Treatment of NAD
+
with nitrous acid deaminates
the adenine ring. The resulting deamino NAD
+
as
well as synthetic analogs containing the following
groups in place of the carboxyamide have been used
widely in enzyme studies. In fact, almost every part
of the coenzyme molecule has been varied systemati-
cally and the effects on the chemical and enzymatic
properties have been investigated.
139 141
Caged
NAD
+
and NADP
+
have also been made.
142
These
compounds do not react as substrates until they are
released (uncaged) by photolytic action of a laser
beam (see Chapter 23).
B. The Flavin Coenzymes
Flavin adenine diphosphate (FAD, flavin adenine
dinucleotide) and riboflavin 5'-monophosphate
(FMN, flavin mononucleotide), whose structures are
shown in Fig. 15-7, are perhaps the most versatile of all
N
H H
C
O
NH
2
N
H H
C
O
NH
2
H
H
N
Adduct
H H
C
O
NH
2
H
H
H
H
+
+
Y

Y
AMP P
O
O
H
OH
H
N
O
H
CONH
2
H
H
H
H
H
O
O
An acid modification product from NADH

(15-20)
N O
C NH
2
O
6-Pyridone
R
C CH
3
,
O
C H,
O
C O

,
O
C NH
2
S
and
781
the oxidation coenzymes. The name flavin adenine
dinucleotide is not entirely appropriate because the
D-ribityl group is not linked to the riboflavin in a
glycosidic linkage. Hemmerich suggested that FAD
be called flavin adenine diphosphate.
143
The attention of biochemists was first attracted to
flavins as a result of their color and fluorescence. The
study of spectral properties of flavins (Fig. 15-8) has
been of importance in understanding these coenzymes.
The biochemical role of the flavin coenzymes was
first recognized through studies of the old yellow
enzyme
144,145
which was shown by Theorell to contain
riboflavin 5'-phosphate. By 1938, FAD was recognized
as the coenzyme of a different yellow protein, D-amino
acid oxidase of kidney tissue. Like the pyridine nucle-
otides, the new flavin coenzymes were reduced by
dithionite to nearly colorless dihydro forms (Figs. 15-7
and 15-8) revealing the chemical basis for their function
as hydrogen carriers.
Flavins are also among the natural light receptors
and display an interesting and much studied photo-
chemistry (Fig. 3-5).
143
Flavins may function in some
photoresponses of plants, and they serve as light
emitters in bacterial bioluminescence (Chapter 23).
146,147
Three facts account for the need of cells for both
the flavin and pyridine nucleotide coenzymes: (1)
Flavins are usually stronger oxidizing agents than is
NAD
+
. This property fits them for a role in the electron
transport chains of mitochondria where a sequence
of increasingly more powerful oxidants is needed and
makes them ideal oxidants in a variety of other dehy-
drogenations. (2) Flavins can be reduced either by
one- or two-electron processes. This enables them to
participate in oxidation reactions involving free radicals
and in reactions with metal ions. (3) Reduced flavins
Figure 15-7 The flavin
coenzymes flavin adenine
dinucleotide (FAD) and
riboflavin 5'-phosphate
(FMN). Dotted lines en-
close the region that is
altered upon reduction.
AMP Riboflavin 5-phosphate or FMN
N
N
NH
N
H
2
C
H
3
C
H
3
C
O
O
C
C
C
H
2
C
H
HO
H
H
OH
OH
N
N
NH
N
R
O
O
O N
N
N
N
NH
2
1
2
3
4
OH OH
5
6
O
P O
O
O

7
8
9
10
H
P O
O
O

H
+ 2[H]
Reduced form FADH
2
or FMNH
2
Oxidized form FAD or FMN
5'
Riboflavin
10a
4a
1'
Figure 15-8 Absorption spectrum of neutral, uncharged
riboflavin (A), the riboflavin anion (B), and reduced to the
dihydro form (Fig. 15-7) by the action of light in the presence
of EDTA (C). A solution of 1.1 x 10
4
M riboflavin containing
0.01 M EDTA was placed 11.5 cm from a 40-W incandescent
lamp for 30 min.
Wave number, cm
1
10
-3
Wavelength, nm
18 22 26 30 34 38 42 46
500 400 350 300 250 220
24
20
16
12
8
4
0
28
32
A
B
C
M
o
l
a
r

e
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t
,


1
0
-
3
B. The Flavin Coenzymes
782 Chapter 15. Coenzymes of OxidationReduction Reactions
are autooxidizable, i.e., they can be reoxidized directly
and rapidly by O
2
, a property shared with relatively
few other organic substances. For example, NADH
and NADPH are not spontaneously reoxidized by
oxygen. Autooxidizability allows flavins of some
enzymes to pass electrons directly to O
2
and also pro-
vides a basis for the functioning of flavins in hydroxy-
lation reactions.
1. Flavoproteins and Their Reduction
Potentials
Flavin coenzymes are usually bound tightly to
proteins and cycle between reduced and oxidized states
while attached to the same protein molecule. In a free
unbound coenzyme the redox potential is determined
by the structures of the oxidized and reduced forms of
the couple. Both riboflavin and the pyridine nucleotides
contain aromatic ring systems that are stabilized by
resonance. Part of this resonance stabilization is lost
upon reduction. The value of E depends in part upon
the varying amounts of resonance in the oxidized and
reduced forms. The structures of the coenzymes have
apparently evolved to provide values of E appropriate
for their biological functions.
The relative strengths of binding of oxidized
and reduced flavin coenzymes to a protein also have
strong effects upon the reduction potential of the
coenzyme.
148
If the oxidized form is bound weakly,
but the reduced form is bound tightly, a bound flavin
will have a greater tendency to stay in the reduced
form than it did when free. The reduction potential
E will be less negative than it is for the free flavin-
dihydroflavin couple. On the other hand, if the oxi-
dized form of the flavin is bound more tightly by the
protein than is the reduced form, E will be more
negative and the flavoenzyme will be a less powerful
oxidizing agent. In fact, the values of E at pH 7 for
flavoproteins span a remarkably wide range from 0.49
to + 0.19 V. The state of protonation of the reduced
flavin when bound to the enzyme will also have a
major effect on the oxidationreduction potential.
For example, acyl-CoA dehydrogenases are thought to
form an anionic species of reduced FAD (FADH

)
which is tightly bound to the protein.
149
Every flavoprotein accepts electrons from the
substrate that it oxidizes and passes these electrons
on to another substrate, an oxidant. In the following
sections we will consider for several enzymes how the
electrons may get into the flavin from the oxidizable
substrate and how they may flow out of the flavin into
the final electron acceptor.
2. Typical Dehydrogenation Reactions
Catalyzed by Flavoproteins
The functions of flavoprotein enzymes are numer-
ous and diversified.
151 153a
A few of them are shown
in Table 15-2 and are classified there as follows: (A)
oxidation of hemiacetals to lactones, (B) oxidation
of alcohols to aldehydes or ketones, (C) oxidation of
amines to imines, (D) oxidation of carbonyl compounds
or carboxylic acids to ,-unsaturated compounds,
(E) oxidation of NADH and NADPH in electron trans-
port chains, and (F) oxidation of dithiols to disulfides
or the reverse reaction. Three-dimensional structures
are known for enzymes of each of these types.
Reactions of types AC could equally well be
catalyzed by pyridine nucleotide-requiring dehydro-
genases. Recall that D-glucose-6-phosphate dehydro-
genase uses NADP
+
as the oxidant (Eq. 15-10). The
first product is the lactone which is hydrolyzed to
6-phosphogluconic acid. The similar reaction of free
glucose (Table 15-2, reaction type A) is catalyzed by
fungal glucose oxidase, a 580-residue FAD-containing
enzyme.
154155
A bacterial cholesterol oxidase has a
similar structure.
156
The important plant enzyme
glycolate oxidase is a dimer containing riboflavin
5'-phosphate.
157 159
It catalyzes a reaction of type B,
which plays an important role in photorespiration
(Chapter 23). Amino acid oxidases (reaction type C)
are well-known. The peroxisomal D-amino acid oxidase
from kidney was the source from which Warburg first
isolated FAD and has been the subject of much investi-
gation of mechanism and structure.
153,160 163
Many
snake venoms contain an active 140-kDa L-amino-acid
oxidase which contains FAD. Flavin-dependent amine
oxidases, important in the human body, catalyze the
related reaction with primary, secondary, or tertiary
amines and in which a carboxyl group need not be
present.
164 166a
Reduced flavin produced by all of
these oxidases is reoxidized with molecular oxygen
and hydrogen peroxide is the product. Nature has
chosen to forego the use of an electron transport chain
(Fig. 10-5), giving up the possible gain of ATP in favor
of simplicity and a more direct reaction with oxygen.
In some cases there is specific value to the organism in
forming H
2
O
2
(see Chapter 18).
In contrast to the flavin oxidases, flavin dehydro-
genases pass electrons to carriers within electron
transport chains and the flavin does not react with O
2
.
Examples include a bacterial trimethylamine dehy-
drogenase (Fig. 15-9) which contains an ironsulfur
cluster that serves as the immediate electron accep-
tor
167 169
and yeast flavocytochrome b
2
, a lactate
dehydrogenase that passes electrons to a built-in heme
group which can then pass the electrons to an external
acceptor, another heme in cytochrome c.
170 173
Like
glycolate oxidase, these enzymes bind their flavin
coenzyme at the ends of 8-stranded barrels similar
783 B. The Flavin Coenzymes
BOX 15-B RIBOFLAVIN
The bright orange-yellow color and brilliant green-
ish fluorescence of riboflavin first attracted the attention
of chemists. Blyth isolated the vitamin from whey in
1879 and others later obtained the same fluorescent,
yellow compound from eggs, muscle, and urine. All of
these substances, referred to as flavins because of their
yellow color, were eventually recognized as identical.
The structure of riboflavin was established in 1933 by R.
Kuhn and associates, who had isolated 30 mg of the
pure material from 30 kg of dried albumin from 10,000
eggs. The intense fluorescence assisted in the final stages
of purification. The vitamin was synthesized in 1935 by
P. Karrer.
a
Riboflavin, a yellow solid, has a low solubility of
~ 100 mg / l at 25C. Three crystalline forms are known.
One of these, the readily soluble form, is ten times
more soluble than the others and can be used to prepare
metastable solutions of higher concentration. One crys-
talline form is platelike and occurs naturally in the
tapetum (Box 13-C) of the nocturnal lemur.
Discovery of the role of riboflavin in biological
oxidation was an outgrowth of biochemists interest in
respiration. In the 1920s Warburg provided evidence
that oxygen reacted with an iron-containing respiration
catalyst and it was shown that the dye methylene blue
could often substitute for oxygen as an oxidant (Box 18-A).
Oxidation of glucose 6-phosphate by methylene blue
within red blood cells required both a ferment (en-
zyme) and a coferment, later identified as NADP
+
.
A yellow protein, isolated from yeast, was found to
have the remarkable property of being decolorized by
the reducing system of glucose 6-phosphate plus the
protein and coferment from red blood cells.
Warburg and Christian showed that the color of this
old yellow enzyme came from a flavin and proposed
that its cyclic reduction and reoxidation played a role
in cellular oxidation. When NADP
+
was isolated the
proposal was extended to encompass a respiratory
chain. The two hydrogen carriers NADP
+
and flavin
would work in sequence to link dehydrogenation of
glucose to the iron-containing catalyst that interacted
with oxygen. While we still do not know the physiolog-
ical function of the old yellow enzyme,
b
the concept of
respiratory chain was correct.
Human beings require about 1.5 mg of riboflavin
per day. Because of its wide distribution in food, a
deficiency, which affects skin and eyes, is rarely seen.
Riboflavin is produced commercially in large quantities
by fungi such as Eremothecium asbyii which, apparently
because of some metabolic anomaly, produce the vita-
min in such copious amounts that it crystallizes in the
culture medium.
When taken up by the body, riboflavin is converted
into its coenzyme forms (Chapter 25) and any excess is
quickly excreted in the urine. Urine also contains smaller
amounts of metabolites. The ribityl group may be cut
by the action of intestinal bacteria acting on riboflavin
before it is absorbed. The resulting 10-hydroxyethyl
flavin may sometimes be a major urinary product.
c,d
The related 10-formylmethyl flavin is also excreted,
c
as
are small amounts of 7- and 8- hydroxyriboflavins,
apparently formed in the body by hydroxylation. These
may be degraded farther to the 7- and 8- carboxylic
acids of lumichrome (riboflavin from which the ribityl
side chain is totally missing).
e
A riboflavin glucoside
has also been found in rat urine.
f
The choroid layer of the eye (behind the retina) in
many animals contains a high concentration of free
riboflavin. Cats eyes also contain a large amount of
7 -hydroxyriboflavin (nekoflavin), as do their livers.
g
Nekoflavin is also present in human blood.
h
Hen egg
white contains a 219-residue riboflavin-binding protein
whose functions are thought to be storage of the vitamin
and delivery to the developing embryo.
i k
Most of the
riboflavin in human blood is bound to proteins such
as albumin and immunoglobulins. However, during
pregnancy a riboflavin-binding protein similar to that
of the chicken appears, apparently to carry riboflavin to
the fetus.
j
Riboflavin is stable to heat but is extremely sensitive
to light, a fact of some nutritional significance. Do not
leave bottled milk in the sunshine (see Fig. 15-8)! Many
products of photolysis are formed (Fig. 3-5). Among
them is lumichrome.
a
Yagi, K. (1990) in Flavins and Flavoproteins (Curti, B., Ronchi, S.,
and Zanetti, G., eds), pp. 3 16, Walter de Gruyter, Berlin
b
Kohli, R. M., and Massey, V. (1998) J. Biol. Chem. 273, 32763
32770
c
Owen, E. C., West, D. W., and Coates, M. E. (1970) Br. J. Nutr. 24,
259267
d
Roughead, Z. K., and McCormick, D. B. (1991) European Journal
of Clinical Nutrition 45, 299307
e
Ohkawa, H., Ohishi, N., and Yagi, K. (1983) J. Biol. Chem. 258,
56235628
f
Ohkawa, H., Ohishi, N., and Yagi, K. (1983) J. Nutr. Sci. Vitami-
nol. 29, 515522
g
Matsui, K., and Kasai, S. (1996) J. Biochem. 119, 441447
h
Zempleni, J., Galloway, J. R., and McCormick, D. B. (1995) Int. J.
Vitamins Nutr. Res. 66, 151157
i
Matsui, K., Sugimoto, K., and Kasai, S. (1982) J. Biochem. 91,
469475
j
Miura, R., Tojo, H., Fujii, S., Yamano, T., Miyake, Y., (1984) J.
Biochem. 96, 197206
k
Monaco, H. L. (1997) EMBO J. 16, 14751483
H
3
C
H
3
C N
O
NH
N N
CH
2
O
CH
2
OH
H
C
HO
OH
C
C
H
HO
H
784 Chapter 15. Coenzymes of OxidationReduction Reactions
B.
2[H]
Glycolate oxidase
HOOC CH
2
OH HOOC CH O
A. d-Glucose
2[H]
Glucose oxidase
Gluconolactone
H
2
O
d-Gluconic acid
(See Eq. 17-12 for structures in a closely related reaction.)
D. R CH
2
CH
2
C S CoA
O
Acyl-CoA dehydrogenases
2[H]
R CH CH C S CoA
O
C. R C
H
NH
2
COOH
2[H]
R C COOH
NH
H
2
O
R C COOH
O
+ NH
3
E. NADH (or NADPH) + H
+
NAD
+
(NADP
+
)
2[H]
E.
C
N
O
HS SH
R
H
C
N
O
R
H
S S
2[H]
GSH
Amino acid oxidases
Dihydrolipoic acid amide
TABLE 15-2
Some Dehydrogenation Reactions Catalyzed by Flavoproteins
a
a
These are shown as removal of two H atoms [H] and may occur by transfer of H

, H
+
+ e

, or 2H
+
+ 2e

. They represent reaction type 9C of the


table inside the back cover.
to that of triose phosphate isomerase (Fig. 2-28). Flav-
ocytochrome b
2
has an additional domain which car-
ries the bound heme. Two additional domains
of trimethylamine dehydrogenase have a topology
resembling that of the FAD- and NADH-binding
domains of glutathione reductase shown in Fig. 15-10.
A bacterial mandelate dehydrogenase is structurally
and mechanistically closely related to the glycolate
oxidase family.
173a
Reaction type D of Table 15-2, the dehydrogenation
of an acyl-coenzyme A (CoA), could not be accomplished
by a pyridine nucleotide system because the reduction
potential (E, pH 7 = 0.32 V) is inappropriate. The
more powerfully oxidizing flavin system is needed.
(However, the reverse reaction, hydrogenation of a
C = C bond, is often carried out biologically with a
reduced pyridine nucleotide.) Dehydrogenation
reactions of this type are important in the energy
metabolism of aerobic cells. For example, the first
oxidative step in the oxidation of fatty acids (Fig. 17-1)
is the , dehydrogenation of fatty acyl-CoA deriva-
tives. The pro-R hydrogen atoms are removed from
both the - and -carbon atoms to create the double
bond (Table 15-2, type C).
F
C
D
A
E
B
Figure 15-9 Stereoscopic view of the
large domain (residues 1 383) of tri-
methylamine dehydrogenase from a
methylotrophic bacterium. The helices
and strands of the ()
8
barrel are drawn
in heavy lines as are the FMN (center) and
the Fe
4
S
4
ironsulfur cluster at the lower
right edge. The / loop to which it is
bound is drawn with dashed lines. The
733-residue protein also contains two other
structural domains. From Lim et al.
150
Courtesy of F. S. Mathews.
785
Figure 15-10 The three-
dimensional structure of
glutathione reductase. Bound
FAD is shown. NAD
+
binds
to a separate domain below
the FAD. The two cysteine
residues forming the reduc-
ible disulfide loop are indicat-
ed by dots. From Thieme et
al.
182
Animal mitochondria contain several different
acyl-CoA dehydrogenases with differing preferences
for chain length or branching pattern.
174177
A related
reaction that occurs in the citric acid cycle is dehydro-
genation of succinate to fumarate by succinate dehy-
drogenase (Eq. 15-21).
177a
The dehydrogenation also
involves trans removal of one of the two hydrogens,
one pro-S hydrogen and one pro-R.
178
The enzyme has
a large 621-residue flavoprotein subunit and a smaller
27-kDa ironsulfur protein subunit.
179 181
Neither the acyl-CoA dehydrogenases nor succinate
dehydrogenase react with O
2
. Acyl-CoA dehydroge-
nases pass the electrons removed from substrates to
another flavoprotein, a soluble electron transferring
flavoprotein (p. 794), which carries the electrons to
an ironsulfur protein embedded in the inner mito-
chondrial membrane where they enter the electron-
transport chain. Succinate dehydrogenase as well as
NADH dehydrogenase (Table 15-2, reaction E)
183
are
embedded in the same membrane and also pass their
electrons to ironsulfur clusters and eventually to
oxygen through the electron transport chain of the mito-
chondria (Chapter 18). Fumarate reductase
184
has
properties similar to those of succinate dehydrogenase
but catalyzes the opposite reaction in anaerobic respi-
ration (Chapter 18),
184
as do similar reductases of
bacteria
185
and of some eukaryotes.
186
Dihydrolipoyl dehydrogenase (lipoamide dehy-
drogenase), glutathione reductase, and human
thioredoxin reductase
187 190
belong to a subclass
of flavoproteins that act on dithiols or disulfides.
The reaction catalyzed by the first of these is illustrated
in Table 15-2 (reaction type F). The other two enzymes
usually promote the reverse type of reaction, the re-
duction of a disulfide to two SH groups by NADPH
(Eq. 15-22). Glutathione reductase splits its substrate
into two halves while reduction of the small 12-kDa
protein thioredoxin (Box 15-C) simply opens a loop
in its peptide chain. The reduction of lipoic acid opens
the small disulfide-containing 5-membered ring in that
molecule. Each of these flavoproteins also contains
within its structure a reducible disulfide group that
participates in catalysis.
Each 50-kDa chain of the dimeric glutathione
reductase is organized into three structural domains
(Fig. 15-10).
191 193
Two of the domains each contain
a nucleotide-binding motif resembling those of the
NAD
+
-dependent dehydrogenases. One of these
domains binds NADPH and the other FAD. The latter
domain also contains the reducible disulfide which is
formed from Cys 58 and Cys 63. It serves as an inter-
mediate hydrogen carrier which can in turn reduce
oxidized glutathione. A trypanothione reductase
from trypanosomes and related flagellated protozoa
has a similar structure and acts on trypanothione,
which replaces glutathione in these organisms.
188,194-196
Because it is unique to trypanosomes, this enzyme is
a target for design of drugs against these organisms
G S S G
Oxidized glutathione
G SH 2
Glutathione
(See Box 11-B for structure)
NADPH + H
+
NADP
+
Glutathione reductase
B. The Flavin Coenzymes
(15-22)
C

OOC
COO

H
S
H
S
H
R
H
R
H
H COO

OOC
Succinate
dehydrogenase
2[H]
(15-21)
786 Chapter 15. Coenzymes of OxidationReduction Reactions
BOX 15-C THIOREDOXIN AND GLUTAREDOXIN
The small proteins thioredoxin and glutaredoxin
are present in relatively high concentrations in bac-
teria, plants, and animals. For example, thioredoxin
has a concentration of 15 M in E. coli. Both proteins
were discovered by their role as reducing agents in
conversion of the ribonucleotides AMP, GMP, CMP,
and UMP to the corresponding 2-deoxyribonucleo-
tides which are needed for synthesis of DNA:
a,b
The redox group in thioredoxin is a disulfide
loop located on a protrusion at one end of the mole-
cule. In the 108-residue E. coli thioredoxin, as well
as in the 105-residue human thioredoxin,
c
it is formed
by cysteines 32 and 35 which are present in the
conserved sequence CGPC. The SH groups of
these two interacting cysteines have pK
a
values of
~ 6.9 and 7.5, the former belonging predominantly
to the more exposed Cys 32.
d
The buried Asp 26
carboxyl group, which may be a proton donor to
Cys 35 during reduction of the disulfide form,
e
forms a salt bridge with the Lys 57 NH
3
+
and
has a high pK
a
of ~ 7.4.
e g
The pK
a
assignments, which have been contro-
versial, are discussed in Chapter 7. This disulfide
loop is reduced by NADPH through the action of
the flavoprotein enzyme thioredoxin reductase.
NAD(P)H + H
+
Thioredoxin reductase
NAD(P)
+
Thioredoxin-(SH)
2
Thioredoxin-S
2
2-Deoxy-AMP
AMP
The resulting thiol pair of the reduced thioredoxin
is the reductant used for ribonucleotide reductase
(Chapter 16). The standard redox potential E of
E. coli thioredoxin is 0.27 V, appropriately low for
coupling to the NADPH / NADP
+
system.
Reduced thioredoxin has a variety of functions.
h
It is the reductant for conversion of methionine sulf-
oxide to methionine in bacteria, for reduction of
sulfate in yeast, and for additional specific enzymatic
reactions.
b,h
However, its major function may be to
reduce disulfide linkages in various proteins.
i
Several
photosynthetic enzymes are activated by reduction
of disulfide linkages via photosynthetically generated
reduced ferredoxin and thioredoxin
j,k
(Chapter 23).
Reduced thioredoxin may also play a similar role
in nonphotosynthetic cells. It may reduce mixed
disulfides such as those formed between glutathione
and proteins (Box 11-B).
l n
Thioredoxin may partici-
pate in regulation of the level of nitric oxide (NO)
in tissues
m
and it is needed in the assembly of fila-
mentous bacteriophages.
b,o
For reasons that are
not clear, thioredoxin is also an essential subunit
for a virus-induced DNA polymerase formed in
E. coli following infection by bacteriophage T7 (see
Chapter 27).
It was a surprise to discover that a mutant of
E. coli lacking thioredoxin can still reduce ribonu-
cleotides. In the mutant cells thioredoxin is replaced
by glutaredoxin, whose active site disulfide linkage
is reduced by glutathione rather than directly by
NADPH. Oxidized glutathione is, in turn, reduced
by NADPH and glutathione reductase. Thus, the
end result is the same with respect to ribonucleotide
reduction.
However, the two proteins have significantly differ-
ent specificities and functions. The disulfide loop
in glutaredoxin, whose eukaryotic forms are often
called thioltransferases,
p
has the sequence CPYC.
Although glutaredoxins are weaker reductants of
mixed disulfides of proteins with glutathione than
are thioredoxins,
q s
they are more specific.
Both thioredoxin and glutaredoxin are members
of a larger group of thiol:disulfide oxidoreductases
which are found in all known organisms. In E. coli
there are one thioredoxin, three different gluta-
redoxins,
h,t
and the periplasmic protein disulfide
NADPH + H
+
Glutathione reductase
NAD(P)
+
Glutaredoxin-(SH)
2
2GSH
GSSG
Glutaredoxin-S
2
Oxidized E. coli thioredoxin. From Langsetmo et al.
f
787
BOX 15-C (continued)
a
Holmgren, A. (1981) Trends Biochem. Sci. 6, 2629
b
Holmgren, A. (1989) J. Biol. Chem. 264, 1396313966
c
Forman-Kay, J. D., Clore, G. M., Wingfield, P. T., and
Gronenborn, A. M. (1991) Biochemistry 30, 26852698
d
Dyson, H. J., Jeng, M.-F., Tennant, L. L., Slaby, I., Lindell, M.,
Cui, D.-S., Kuprin, S., and Holmgren, A. (1997) Biochemistry 36,
26222636
e
Jeng, M.-F., and Dyson, H. J. (1996) Biochemistry 35, 16
f
Langsetmo, K., Fuchs, J. A., and Woodward, C. (1991)
Biochemistry 30, 76037609
g
Ladbury, J. E., Wynn, R., Hellinga, H. W., and Sturtevant, J. M.
(1993) Biochemistry 32, 75267530
h
Gvakharia, B. O., Hanson, E., Koonin, E. K., and Mathews, C.
K. (1996) J. Biol. Chem. 271, 1530715310
i
Thomas, J. A., Poland, B., and Honzatko, R. (1995) Arch.
Biochem. Biophys. 319, 19
j
Capitani, G., Markovic-Housley, Z., DelVal, G., Morris, M.,
Jansonius, J. N., and Schrmann, P. (2000) J. Mol. Biol. 302,
135154
k
Buchanan, B. B., Schrmann, P., Decottignies, P., and Lozano,
R. M. (1994) Arch. Biochem. Biophys. 314, 257260
l
Wynn, R., Cocco, M. J., and Richrds, F. M. (1995) Biochemistry
34, 1180711813
m
Nikitovic, D., and Holmgren, A. (1996) J. Biol. Chem. 271,
1918019185
n
Prinz, W. A., slund, F., Holmgren, A., and Beckwith, J. (1997)
J. Biol. Chem. 272, 15661 15667
o
Russel, M., and Model, P. (1985) Proc. Natl. Acad. Sci. U.S.A. 82,
2933
p
Srinivasan, U., Mieyal, P. A., and Mieyal, J. J. (1997)
Biochemistry 36, 31993206
q
Katti, S. K., Robbins, A. H., Yang, Y., and Wells, W. W. (1995)
Protein Sci. 4, 19982005
r
Gravina, S. A., and Mieyal, J. J. (1993) Biochemistry 32,
3363376
s
Nikkola, M., Gleason, F. K., and Eklund, H. (1993) J. Biol.
Chem. 268, 38453849
t
slund, F., Nordstrand, K., Berndt, K. D., Nikkola, M.,
Bergman, T., Ponstingl, H., Jrnvall, H., Otting, G., and
Holmgren, A. (1996) J. Biol. Chem. 271, 67366745
u
Freedman, R. B., Hirst, T. R., and Tuite, M. F. (1994) Trends
Biochem. Sci. 19, 331335
v
Rietsch, A., Belin, D., Martin, N., and Beckwith, J. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1304813053
w
Darby, N. J., and Creighton, T. E. (1995) Biochemistry 34,
1677016780
x
Kanaya, E., Anaguchi, H., and Kikuchi, M. (1994) J. Biol. Chem.
269, 42734278
y
Derman, A. I., Prinz, W. A., Belin, D., and Beckwith, J. (1993)
Science 262, 17441747
z
Chivers, P. T., Prehoda, K. E., and Raines, R. T. (1997)
Biochemistry 36, 40614066
aa
Jacobi, A., Huber-Wunderlich, M., Hennecke, J., and
Glockshuber, R. (1997) J. Biol. Chem. 272, 2169221699
bb
Wells, W. W., Xu, D. P., Yang, Y., and Rocque, P. A. (1990)
J. Biol. Chem. 265, 1536115364
cc
Bischoff, R., Lepage, P., Jaquinod, M., Cauet, G., Acker-Klein,
M., Clesse, D., Laporte, M., Bayol, A., Van Dorsselaer, A., and
Roitsch, C. (1993) Biochemistry 32, 725734
dd
Hwang, C., Sinskey, A. J., and Lodish, H. F. (1992) Science 257,
14961502
ee
Ruoppolo, M., Freedman, R. B., Pucci, P., and Marino, G.
(1996) Biochemistry 35, 1363613646
ff
Couprie, J., Vinci, F., Dugave, C., Qumneur, E., and
Mourtiez, M. (2000) Biochemistry 39, 67326742
which cause such terrible diseases as African sleeping
sickness and Chagas disease.
195,197
Another flavoprotein constructed on the glutathione
reductase pattern is the bacterial plasmid-encoded
mercuric reductase which reduces the highly toxic
Hg
2+
to volatile elemental mercury, Hg
0
. A reducible
disulfide loop corresponding to that in glutathione
reductase is present in this enzyme but there is also
a second pair of cysteines nearby. All of these may
participate in binding and reduction of Hg
2+
.
198199a
B. The Flavin Coenzymes
isomerases DsbA and DsbC, which were discussed
in Chapter 10.
u w
Similar enzymes are found in the
endoplasmic reticulum of eukaryotic cells.
u,x
Like
thioredoxins, they contain disulfide loops which
contain the sequences CGHC and CGYC as well as
CPHC. Thioredoxin reductase itself also can keep
cytoplasmic bacterial proteins reduced.
y
Redox
potentials vary within this family of proteins:
n,s,z,aa
E

, pH 7
Thioredoxins .27 V
Glutaredoxins .20 to .23 V
Protein disulfide isomerase .18 V
DsbA .09 to .12 V
These differences are correlated with differing func-
tions. The intracellular reduced thioredoxins are
thermodynamically the best reductants of disulfide
linkages in proteins and they help keep intracellular
proteins reduced. Glutaredoxin can be reduced
efficiently by reduced glutathione or by NADPH
and glutathione reductase and can, in turn, reduce
cysteine and the oxidized form of vitamin C,
dehydroascorbic acid (Box 18-D).
bb,cc
The peri-
plasmic bacterial proteins DsbA and DsbC have the
highest redox potentials and an unusually low first
pK
a
for the dithiol pair in their thioredoxinlike
domains.
aa
The basis for these properties has been
hard to understand but is consistent with their role
in assisting formation of disulfide bridges in extra-
cellular proteins and with the role of the related
protein disulfide isomerase in the ER.
u,ddff
These
disulfide exchanges are nucleophilic displacement
reactions (Chapter 12).
788 Chapter 15. Coenzymes of OxidationReduction Reactions
has been isolated from a bacterial electron-transferring
flavoprotein.
225
Commercial FAD may contain some
riboflavin 5'-pyrophosphate which activates some
flavoproteins and inhibits others.
226
Methanogenic bacteria contain a series of unique
coenzymes (Section F) among which is coenzyme
F
420
, a 5-deazaflavin substituted by H at position 7
and OH at position 8 (8-hydroxy-7,8-didemethyl-5-
deazariboflavin).
227,228
This unique redox catalyst links the oxidation of
H
2
or of formate to the reduction of NADP
+229
and also
serves as the reductant in the final step of methane
biosynthesis (see Section E).
228
It resembles NAD
+
in
having a redox potential of about 0.345 volts and
the tendency to be only a two-electron donor. More
recently free 8-hydroxy-7,8-didemethyl-5-deazaribo-
flavin has been identified as an essential light-absorbing
chromophore in DNA photolyase of Methanobacterium,
other bacteria, and eukaryotic algae.
230
Roseoflavin
is not a coenzyme but an antibiotic from Streptomyces
davawensis.
231
Many synthetic flavins have been used
in studies of mechanisms and for NMR
232
and other
forms of spectroscopy.
NH
N N
N
O
O
H
3
C
H
2
C N N
CH
2
HC
HN
C O
ribitylPPadenosine
Peptide
chain
Covalently bound modified FAD
of succinate dehydrogenase
1
2
(as in FAD)
3. More Flavoproteins
Flavoproteins function in virtually
every area of metabolism and we have
considered only a small fraction of the
total number. Here are a few more.
Flavin-dependent reductases use hydrogen
atoms from NADH or NADPH to reduce
many specific substances or classes of com-
pounds. The FAD-containing ferredoxin:
NADP
+
oxidoreductase catalyzes the reduc-
tion of free NADP
+
by reduced ferredoxin generated in
the chloroplasts of green leaves.
200,201
Similar enzymes,
some of which utilize reduced flavodoxins, are found in
bacteria.
202,203
The FMN-containing subunit of NADH:
ubiquinone oxidoreductase is an essential link in the
mitochondrial electron transport chain for oxidation of
NADH in plants and animals
183,204,205
and for related
processes in bacteria. Glutamate synthase, a key
enzyme in the nitrogen metabolism of plants and
microorganisms, uses electrons from NADPH to reduce
2-oxoglutarate to glutamate in a complex glutamine-
dependent process (see Fig. 24-5). The enzyme contains
both FMN and FAD and three different iron sulfur
clusters.
205a
Flavin reductases use NADH or NADPH
to reduce free riboflavin, FMN, or FAD needed for
various purposes
206,206a
including emission of light by
luminous bacteria.
207
They provide electrons to many
enzymes that react with O
2
such as the cytochromes
P450
208,209
and nitric oxide synthase (Chapter 18).
An example is adrenodoxin reductase (see chapter
banner, p. 764), which passes electrons from NADPH
to cytochrome P450 via the small redox protein adreno-
doxin. This system functions in steroid biosynthesis as is
indicated in Fig. 22-7.
209a,b
Other flavin-dependent reduc-
tases have protective functions catalyzing the reduction
of ascorbic acid radicals,
210,211
toxic quinones,
212 214
and peroxides.
215 218
4. Modified Flavin Coenzymes
Mitochondrial succinate dehydrogenase, which
catalyzes the reaction of Eq. 15-21, contains a flavin
prosthetic group that is covalently attached to a histi-
dine side chain. This modified FAD was isolated and
identified as 8-(N
2
-histidyl)-FAD.
219
The same pros-
thetic group has also been found in several other dehy-
drogenases.
220
It was the first identified member of
a series of modified FAD or riboflavin 5'-phosphate
derivatives that are attached by covalent bonds to
the active sites of more than 20 different enzymes.
219
These include 8-(N
2
-histidyl)-FMN,
221
8-
(N
1
-histidyl)-FAD,
222
8-(O-tyrosyl-FAD),
223
and 6-
(S-cysteinyl)-riboflavin 5'-phosphate, which is found
in trimethylamine dehydrogenase (Fig. 15-9).
224
An
8-hydroxy analog of FAD ( OH in place of the 8-CH
3
)
N
NH
N O
O
H
HO
CH
2
C
C
C
CH
2
H
HO
HO
H
OH
H
O
P
O
N
O
CH
3
O CO
2

O
N
O CO
2
H
H

OOC
Coenzyme F
420
(oxidized form)
789
To avoid this problem, Brustlein and Bruice used a
5-deazaflavin to oxidize NADH nonenzymatically.
237
When this reaction was carried out in
2
H
2
O, no
2
H
entered the product at C-5, indicating that a hydrogen
atom (circled in Eq. 15-24) had been transferred directly
from NADH to the C-5 position. Similar direct transfer
of hydrogen to C-5 of 5-deazariboflavin 5'-phosphate
is catalyzed by flavoproteins such as N-methylglutamate
synthase
238
and acyl-CoA dehydrogenase.
237 239
However, these experiments may not have established
a mechanism for natural flavoprotein catalysis because
the properties of 5-deazaflavins resemble those of NAD
+
more than of flavins.
239
Their oxidationreduction
potentials are low, they do not form stable free radicals,
and their reduced forms dont react readily with O
2
.
Nevertheless, for an acyl-CoA dehydrogenase the rate
of reaction of the deazaflavin is almost as fast as that
of natural FAD.
238
For these enzymes a hydride ion
transfer from the CH (reaction type D of Table 15-1)
is made easy by removal of the -H of the acyl-CoA to
form an enolate anion intermediate.
The three-dimensional structure of the medium
chain acyl-CoA dehydrogenases with bound substrates
and inhibitors is known.
174,175,240
A conserved glutamate
side chain is positioned to pull the pro-R proton from
the carbon to create the initial enolate anion.
174,175,241
The pro-R C H lies by N-5 of the flavin ring seem-
ingly ready to donate a hydride ion as in Eq. 15-23.
NMR spectroscopy has been carried out with
13
C or
15
N in each of the atoms of the redox active part of the
FAD. The results show directly the effects of strong
hydrogen bonding to the protein at N-1, N-3, and N-5
and also suggest that the bound FADH
2
is really FADH

with the negative charge localized on N-1 by strong


hydrogen bonding.
149
Many mutants have been made,
242
substrate analogs have been tested,
176,243
kinetic
isotope effects have been measured,
242a
and potentio-
metric titrations have been done.
243
All of the results
are compatible with the enolate anion hydride-transfer
N
N
NH
N O
O
R H
+
5
H

N
N
NH
N O
O
R
H
H
(15-23)
5. Mechanisms of Flavin Dehydrogenase
Action
The chemistry of flavins is complex, a fact that is
reflected in the uncertainity that has accompanied efforts
to understand mechanisms. For flavoproteins at least
four mechanistic possibilities must be considered.
153a,233
(a) A reasonable hydride-transfer mechanism can be
written for flavoprotein dehydrogenases (Eq. 15-23).
The hydride ion is donated at N-5 and a proton is
accepted at N-1. The oxidation of alcohols, amines,
ketones, and reduced pyridine nucleotides can all be
visualized in this way. Support for such a mechanism
came from study of the nonenzymatic oxidation of
NADH by flavins, a reaction that occurs at moderate
speed in water at room temperature. A variety of
flavins and dihydropyridine derivatives have been
studied, and the electronic effects observed for the
reaction are compatible with the hydride ion mecha-
nism.
234 236
According to the mechanism of Eq. 15-23, a hydride
ion is transferred directly from a carbon atom in a sub-
strate to the flavin. However, a labeled hydrogen
atom transferred to N-5 or N-1 would immediately
exchange with the medium, rapid exchange being
characteristic of hydrogens attached to nitrogen.
B. The Flavin Coenzymes
2
H
2
O
N
N
N
CH
3
H O
O
N
N
N
CH
3
O
O
CH
3
N
H H
CONH
2
R
N
R
CONH
2
2
H
CH
3
+
H H
(15-24)
N
N
NH
N O
O
CH
2
CH)
3
CH
2
OH
H
3
C
N
H
3
C
H
3
C
Roseoflavin
(HO
790 Chapter 15. Coenzymes of OxidationReduction Reactions
4a adduct mechanism are the reduced
flavin and an aldehyde, the same as
would be obtained by the hydride ion
mechanism. However, in Eq. 15-26,
both hydrogens in the original substrate
(that on oxygen and that on carbon)
have dissociated as protons, the electrons
having moved as a pair during the
cleavage of the adduct. Hamilton argued
that an isolated hydride ion has a large
diameter while a proton is small and
mobile; for this reason dehydrogenation
may often take place by proton transfer
mechanisms.
Experimental support for the mech-
anism of Eq. 15-26 has been obtained
using D-chloroalanine as a substrate
for D-amino acid oxidase.
252 254
Chloro-
pyruvate is the expected product, but
under anaerobic conditions pyruvate
was formed. Kinetic data obtained with -
2
H and
-
3
H substrates suggested a common intermediate
for formation of both pyruvate and chloro-pyruvate.
This intermediate could be an anion formed by loss
of H
+
either from alanine or from a C-4a adduct. The
anion could eliminate chloride ion as indicated by the
dashed arrows in the following structure. This would
lead to formation of pyruvate without reduction
of the flavin. Alternatively, the electrons from the
carbanion could flow into the flavin (green arrows),
reducing it as in Eq. 15-26. A similar mechanism
has been suggested for other flavoenzymes.
249,255
Objections to the carbanion mechanism are the expected
N
N
NH
N
R
O
O
H
R C COOH
NH
2
N
N
NH
N
R
O
O
R C COOH
NH
2
H
R C COO
NH
2
N
N
NH
N
R
O
O

+
+
+
H
H
Reduced flavin
H
5 4a
(15-25)
mechanism. Questions about the acidity of the -H
and the mechanism of its removal to form the enolate
anion
242a
are similar to those discussed in Chapter 13,
Section B.
A peculiarity of several acyl-CoA dehydrogenases
is a bright green color with an absorption maximum at
710 nm. This was found to result from tightly bound
coenzyme A persulfide (CoA S S

).
244,245
(b) A second possible mechanism of flavin reduc-
tion is suggested by the occurrence of addition reactions
involving the isoalloxazine ring of flavins. Sulfite adds
to flavins by forming an NS bond at the 5 position and
nitroethane, which is readily dissociated to the carban-
ion H
3
C CH

NO
2,
acts as a substrate for D-amino
acid oxidase.
246,247
This fact suggested a carbanion
mechanism according to which normal D-amino acid
substrates form carbanions by dissociation of the H
(Eq. 15-25). Ionization would be facilitated by binding
of the substrate carboxylate to an adjacent arginine
side chain and the carbanion could react at N-5 of the
flavin as in Eq. 15-25. Similar mechanisms have been
suggested for other flavin enzymes.
248,249
(c) The adducts with nitroethane and other com-
pounds
250
pointed to reaction at N-5, but Hamilton
251
suggested that a better position for addition of nucleo-
philes is carbon 4a, which together with N-5 forms a
cyclic Schiff base. He argued that other electrophilic
centers in the flavin molecule, such as carbons 2, 4,
and 10a, would be unreactive because of their involve-
ment in amide or amidine-type resonance but an amine,
alcohol, or other substrate could add to a flavin at
position 4a (Eq. 15-26, step a). Cleavage of the newly
formed CO bond could then occur by movement of
electrons from the alcohol part of the adduct into the
flavin as indicated in step b. The products of this
R C OH
H
H
N
N
NH
N O
O
H
+
O
C H H
R
H
R
4a
H C
O
R
+ H
+
N
N
NH
N O
O
H
R
H
Flavin
coenzyme
Adduct
a
b
(15-26)
791 B. The Flavin Coenzymes
This H removed as H
+
in rate-limiting step
N
N
NH
N O
O
R
H
3
C
H
3
C
H
H
2
N
+
C H
2
C Cl COO

H
H
+
4a
very high pK
a
for loss of the -H to form the car-
banion
256
and the observed formation of only chloro-
alanine and no pyruvate in the reverse reaction of
chloropyruvate, ammonia, and reduced flavoprotein.
A long-known characteristic of D-amino acid oxi-
dase is its tendency to form charge-transfer complexes
with amines, complexes in which a nonbonding electron
has been transferred partially to the flavin. Complete
electron transfer would yield a flavin radical and a
substrate radical which could be intermediates in a
free radical mechanism, as discussed in the next
section.
256
The three-dimensional structure of the complex
of D-amino acid oxidase with the substrate analog
benzoate has been determined. The carboxyl group
of the inhibitor is bound by an arginine side chain
(Fig. 15-11) that probably also holds the amino acid
substrate. There is no basic group nearby in the enzyme
that could serve to remove the -H atom in Eq. 15-26
but the position is appropriate for a direct transfer of
the hydrogen to the flavin as a hydride ion as in Eq.
15-23.
161,162,257
In spite of all arguments to the contrary
the hydride ion mechanism could be correct! However,
an adduct mechanism is still possible.
Experimental evidence supports a 4a adduct
mechanism for glutathione reductase and related
enzymes
191,258,258a
(Fig. 15-12). In this figure the reac-
tion sequence is opposite to that in Eq. 15-26. The
enzyme presumably functions as follows. NADPH
binds next to the bound FAD and reduces it by transfer
of the 4-pro-S hydrogen of the NADPH (Fig. 15-12,
step a). The sulfur atom of Cys 63 is in van der Waals
contact with the bound FAD at or near carbon atom 4a.
In step b the nucleophilic center on atom C-4a of FADH
2
attacks a sulfur atom of the disulfide loop between
cysteines 58 and 63 in the protein to create a C-4a
adduct of a thiolate ion with oxidized FAD and to
cleave the S S linkage in the loop. In step c the thiol
group of cysteine 63 is eliminated, after which the thiol
of Cys 58 attacks the nearer sulfur atom of the oxidized
glutathione in a nucleophilic displacement (step d)
to give one reduced glutathione (GSH) and a mixed
disulfide of glutathione and the enzyme (G-S-S-Cys 58).
The thiolate anion of Cys 63, which is stabilized by
interaction with the adjacent flavin ring, then attacks
this disulfide (step e) to regenerate the internal disulfide
and to free the second molecule of reduced glutathione.
The imidazole group of the nearby His 467 of the second
subunit presumably participates in catalysis as may
some other side chains.
191
The disulfide exchange
reactions are similar to those discussed in Chapter 12.
A variation is observed for E. coli thioredoxin
reductase. The reducible disulfide and the NADPH
binding site are both on the same side of the flavin
rather than on opposite sides as in Fig. 15-12.
190,259
Mercuric reductase also uses NADPH as the reductant
transferring the 4S hydrogen. The Hg
2+
presumably
binds to a sulfur atom of the reduced disulfide loop
and there undergoes reduction. The observed geometry
of the active site is correct for this mechanism.
6. Half-Reduced Flavins
A possible mechanism of flavin dehydrogenation
consists of consecutive transfer of a hydrogen atom
and of an electron with intermediate radicals being
formed both on the flavin and on the substrate. Such
Figure 15-11 Stereoscopic view
of the benzoate ion in its complex
with D-amino acid oxidase. A
pair of hydrogen bonds binds the
carboxylate of the ligand to the
guanidinium group of R283.
Several hydrogen bonds to the
flavin ring of the FAD are also
indicated. Courtesy of Retsu
Miura.
161
792 Chapter 15. Coenzymes of OxidationReduction Reactions
a mechanism takes full advantage of one of the most
characteristic properties of flavins, their ability to
accept single electrons to form semiquinone radicals.
If the oxidized form Fl of a flavin is mixed with the
reduced form FlH
2
, a single hydrogen atom is trans-
ferred from FlH
2
to Fl to form two

F1H radicals (Eq.


15-27).
The equilibrium represented by this equation is
independent of pH, but because all three forms of the
flavin have different pK
a
values (Fig. 15-13) the appar-
ent equilibrium constants relating total concentrations
of oxidized, reduced, and radical forms vary with
pH.
143,260 262
The fraction of radicals present is greater
at low pH and at high pH than at neutrality. For a
3-alkylated flavin the formation constant K
f
has been
estimated as 2.3 x 10
2
and for riboflavin
260
as 1.5 x 10
2
.
From these values and the pK
a
values in Fig. 15-13, it
is possible to estimate the amount of radical present at
any pH.
Neutral flavin radicals have a blue color (the
wavelength of the absorption maximum,
max
, is ~ 560
nm) but either protonation at N-1 or dissociation of a
proton from N-5 leads to red cation or anion radicals
with
max
at ~ 477 nm. Both blue and red radicals are
Figure 15-12 Probable reaction mechanism for lipo-
amide dehydrogenase and glutathione reductase. See
Pai and Schulz.
191
S
S
C63
C58
FAD
NADPH
N
H
H
N
N
N
N
H
O
O
5
1
N
H
O
H
+
O
N

S
S
C58
N
N
N
N
N
H
O
O
N
H
O
O
N

H
+
H
+
S
N
N
N
N
H
O
O
N
H
O
O
N

H
H
b
c
e
a
d
N
N
N
N
H
O
O
N
H
O
O
N

S
S
H
+
N
N
N
N
H
O
O
N
H
O
O
N

S
S
S
C63
C58
G
S
H
+
GSH
G S S G
GSH
G
G
S
S
C63
C58
S

(15-27)
Fl + FlH
2
K
f
2

FlH
793
Figure 15-13 Properties of oxidized, half-reduced, and fully reduced flavins.
See Mller et al.
263,264
observed in enzymes, with some enzymes favoring
one and some the other. Hemmerich suggested that
enzymes forming red radicals make a strong hydrogen
bond to the proton in the 5 position of the flavin. This
increases the basicity of N-1 leading to its protonation
and formation of the red cation radicals.
It is possible that many flavoprotein oxygenases
and dehydrogenases react via free radicals. For exam-
ple, instead of the mechanism of Eq. 15-26, an electron
could be transferred to the flavin, leaving a radical
pair (at right). The crystallographic structure and
modeling of the substrate complex support this
possibility.
265,265a
In this pair the flavin radical would
be more basic than in the fully oxidized form and the
amino acid radical would be more acidic than in the
uncharged form. A proton transfer as indicated
together with coupling of the radical pair would yield
the same product as the mechanism of Eq. 15-26.
N
N
NH
N O
O
R
N
N
NH
N O
O
R
N
N
NH
N O
O
R
N
N
NH
N O
O
R
Oxidized flavin, Fl (yellow)
Protonation occurs here in oxidized
flavins at very low pH; pK
a
~ 0
pK
a
~ 10.0
Protonation at low pH,
pK
a
~ 2.3 to form cation radical
red
max
~ 490 nm
Dissociates with pK
a
~8.3 8.6 to red
anion radical
max
~477 nm
Fully reduced, dihydroflavin, FlH
2
pK
a
values 6.2, < 0
pK
a
~ 6.6
Unpaired electron
is distributed by
resonance into the
benzene ring

H
+
H
H
H
Angle variable
144
N
NH
O
N
R
O
N
H
H
Half-reduced semiquinone

FlH
blue radical max ~ 560 nm
Another alternative to a radical
pair is a hydrogen atom transfer
followed by a second electron
transfer.
265
If an enzyme binds a flavin
radical much more tightly than the
fully oxidized or reduced forms,
reduction of the flavoprotein will
take place in two one-electron steps.
In such proteins the values of E
for the two steps may be widely
separated. The best known exam-
ples are the small, low-potential
electron-carrying proteins known as
flavodoxins.
266 269a
These proteins,
which carry electrons between
pairs of other redox proteins, have
a variety of functions in anaerobic
and photosynthetic bacteria, cyano-
bacteria, and green algae. Their
functions are similar to those of the
ferredoxins, ironsulfur proteins
that are considered in Chapter 16.
In some bacteria ferredoxin and
flavodoxin are interchangeable and
the synthesis of flavodoxin
is induced if the bacteria become
deficient in iron. Flavodoxins all
contain riboflavin monophosphate, which functions by
cycling between the fully reduced anionic form and a
blue semiquinone radical.
270
The two reduction steps,
from oxidized flavin to semiquinone and from semi-
quinone to dihydroflavin, are well separated. For exam-
ple, the values of E (pH 7) for the flavodoxin from
Megasphaera elsdenii are 0.115 and 0.373 V, while
those of the Azotobacter vinlandii flavodoxin (azoto-
flavin) are + 0.050 and 0.495 V. The latter is the lowest
known for any flavoprotein.
Flavodoxins are small proteins with an / struc-
ture resembling that of the nucleotide binding domain
of dehydrogenases. According to
31
P NMR data, the
phosphate group of the coenzyme bound to flavodoxin
is completely ionized,
271
even though it is deeply
buried in the protein and is not bound to any positively
charged side chain but to the N terminus of an helix
and to four OH groups of serine and threonine side
B. The Flavin Coenzymes
N
N
NH
N O
O
R
H
3
C
H
3
C
H
C
NH
2
R R
Amino
acid
radical
Flavin
anion
radical
+

794 Chapter 15. Coenzymes of OxidationReduction Reactions


chains. The flavin ring is partially buried near the
surface of the 138-residue protein. An aromatic side
chain, from tryptophan or tyrosine, lies against the flavin
on the outside of the molecule. Flavodoxins can be
crystallized in all three forms: oxidized, semiquinone,
and fully reduced. In the crystals the flavin semi-
quinone, like the oxidized flavin, is nearly planar.
The DNA photolyase of E. coli, an enzyme that
participates in the photochemical repair of damaged
DNA (Chapter 23), contains a blue neutral FAD radical
with a 580-nm absorption band and an appropriate
ESR signal.
230,272
In contrast, the mitochondrial
electron-transferring flavoprotein (ETF), a 57-kDa
dimer containing one molecule of FAD, functions
as a single electron carrier cycling between oxidized
FAD and the red anionic semiquinone.
273,274
The re-
duced forms of the acyl-CoA dehydrogenases transfer
their electrons one at a time from their FAD to the FAD
of two molecules of electron-transferring flavoprotein.
Therefore, an intermediate enzyme-bound radical must
be present in the FAD of acyl-CoA dehydrogenase at
one stage of its catalytic cycle. A related ETF from a
methylotrophic bacterium accepts single electrons from
reduced trimethylamine dehydrogenase (Fig. 15-9).
275
Another flavoprotein that makes use of both one-
and two-electron transfer reactions is ferredoxin-
NADP
+
oxidoreductase (Eq. 15-28). Its bound FAD
accepts electrons one at a time from each of the two
2Fd
red
+ NADP
+
+ H
+
2 Fd
ox
+
+ NADPH
reduced ferredoxins (Fd
red
) in chloroplasts and then
presumably transfers a hydride ion to the NADP
+
.
The enzyme is organized into two structural domains,
201
one of which binds FAD and the other NADP
+
. Similar
single-electron transfers through flavoproteins also occur
in many other enzymes. Chorismate mutase, an impor-
tant enzyme in biosynthesis of aromatic rings (Chapter
25), contains bound FMN. Its function is unclear but
involves formation of a neutral flavin radical.
276,277
7. Metal Complexes of Flavins and
Metalloflavoproteins
The presence of metal ions in many flavoproteins
suggested a direct association of metal ions and flavins.
Although oxidized flavins do not readily bind most
metal ions, they form red complexes with Ag
+
and Cu
+
with a loss of a proton from N-3.
278
Flavin semiquinone
radicals also form strong red complexes with many
metals.
264
If the complexed metal ion can exist in more
than one oxidation state, electron transfer between the
flavin and a substrate could take place through the
metal atom. However, chelation by flavins in nature has
not been observed. Metalloflavoproteins probably func-
tion by having the metal centers close enough to the
flavin for electron transfer to occur but not in direct
contact. This is the case for a bacterial trimethylamine
dehydrogenase in which the FeS cluster is bound about
0.4 nm from the alloxazine ring of riboflavin 5'-phosphate
as shown in Fig. 15-9.
Some metalloflavoproteins contain heme groups.
The previously mentioned flavocytochrome b
2
of
yeast is a 230-kDa tetramer, one domain of which
carries riboflavin phosphate and another heme. A
flavocytochrome from the photosynthetic sulfur bacte-
riumChromatium (cytochrome c-552)
279
is a complex
of a 21-kDa cytochrome c and a 46-kDa flavoprotein
containing 8-(S-cysteinyl)-FAD. The 670-kDa sulfite
reductase of E. coli has an
8

4
subunit structure. The
eight chains bind four molecules of FAD and four of
riboflavin phosphate, while the chains bind three or
four molecules of siroheme (Fig. 16-6) and also con-
tain Fe
4
S
4
clusters.
280,281
Many nitrate and some nitrite
reductases are flavoproteins which also contain Mo or
Fe prosthetic groups.
282,283
A group of aldehyde
oxidases and xanthine dehydrogenases also contain
molybdenum as well as iron (Chapter 16). In every case
the metal ions are bound independently of the flavin.
283a
8. Reactions of Reduced Flavins with Oxygen
Free dihydroriboflavin reacts nonenzymatically
in seconds and reduced flavin oxygenases react even
faster with molecular oxygen to form hydrogen perox-
ide (Eq. 15-29).
FlH
2
+ O
2
Fl + H
2
O
2
The reaction is more complex than it appears. As soon
as a small amount of oxidized flavin is formed, it
reacts with reduced flavin to generate flavin radicals

FlH (Eq. 15-27). The latter react rapidly with O


2
each
donating an electron to form superoxide anion radicals

O
2

(Eq. 15-30a) which can then combine with flavin


radicals (Eq. 15-30b).
284

FlH + O
2

O
2

+ H
+
+ Fl

O
2

FlH + H
+
Fl + H
2
O
2
(15-29)
N
N
N
N
R
O
O

M
+
5 4
Proton has
dissociated
(15-28)
(15-30a)
(15-30b)
795
N
N
NH
N O
O
R
H
N
N
NH
N O
O
R
H
4a
O
O H
H
10a
O
2
4a
During the corresponding reactions of reduced
flavoproteins with O
2
, intermediates have been detected.
For example, spectrophotometric studies of the FAD-
containing bacterial p-hydroxybenzoate hydroxylase
(Chapter 18) revealed the consecutive appearance of
three intermediate forms.
285 287
The first, whose absorp-
tion maximum is at 380 390 nm, is thought to be an
adduct at position 4a (Eq. 15-31). That such a 4a perox-
ide really forms with the riboflavin phosphate of the
light-emitting bacterial luciferase (Chapter 23) was
demonstrated using coenzyme enriched with
13
C at
position 4a.
147
A large shift to lower frequency (from
104 to 83 ppm) accompanied formation of the transient
adduct. Comparison with reference compounds
showed that this change agreed with that predicted.
Other structures for O
2
adducts have also been
considered, as has the possibility of rearrangements
among these structures.
288
Nevertheless, the products
observed from many different flavoprotein reactions
can be explained on the basis of a 4a peroxide.
289
(15-31)
C. Lipoic Acid and the Oxidative
Decarboxylation of -Oxoacids
The isolation of lipoic acid in 1951 followed an
earlier discovery that the ciliate protozoan Tetrahymena
geleii required an unknown factor for growth. In inde-
pendent experiments acetic acid was observed to pro-
mote rapid growth of Lactobacillus casei, but it could be
replaced by an unknown acetate replacing factor.
Another lactic acid bacterium Streptococcus faecalis was
unable to oxidize pyruvate without addition of pyru-
vate oxidation factor. By 1949, all three unknown
substances were recognized as identical.
291,291a
After
working up the equivalent of 10 tons of water-soluble
residue from liver, Lester Reed and his collaborators
isolated 30 mg of a fat-soluble acidic material which
was named lipoic acid (or 6-thioctic acid).
292 294
While Tetrahymena must have lipoic acid in its diet,
we humans can make our own, and it is not considered
a vitamin. Lipoic acid is present in tissues in extraor-
dinarily small amounts. Its major function is to partic-
ipate in the oxidative decarboxylation of -oxoacids
but it also plays an essential role in glycine catabolism
in the human body as well as in plants.
295,296
The struc-
ture is simple, and the functional group is clearly the
cyclic disulfide which swings on the end of a long arm.
Like biotin, which is also present in tissues in very small
amounts, lipoic acid is bound in covalent amide linkage
to lysine side chains in active sites of enzymes:
296a
1. Chemical Reactions of Lipoic Acid
The most striking chemical property of lipoic acid
is the presence of ring strain of ~ 17 25 kJ mol
1
in the
cyclic disulfide. Because of this, thiol groups and
cyanide ions react readily with oxidized lipoic acid to
give mixed disulfides (Eq. 15-32a) and isothiocyanates
(Eq. 15-32b), respectively.
Another result of the ring strain is that the reduc-
tion potential E (pH 7, 25C), is 0.30 V, almost the
same as that of reduced NAD ( 0.32V). Thus, reoxida-
tion of reduced lipoic acid amide by NAD
+
is thermo-
dynamically feasible. Yet another property attributed
to the ring strain in lipoic acid is the presence of an
absorption maximum at 333 nm.
C. Lipoic Acid and the Oxidative Decarboxylation of -Oxoacids
S S
C
N
C
H
O
NH
C
H
H
O
Lipoic acid in amide linkage
(lipoamide)
Lysine side
chain
Peptide
chain
~1.5 nm
Formation of H
2
O
2
by flavin oxidases can occur via
elimination of a peroxide anion HOO

from the adduct


of Eq. 15-31 with regeneration of the oxidized flavin.
In the active site of a hydroxylase, an OH group can be
transferred from the peroxide to a suitable substrate
(Eq. 18-42). Although radical mechanisms are likely to
be involved, such hydroxylation reactions can also be
viewed as transfer of OH
+
to the substrate together
with protonation on the inner oxygen atom of the
original peroxide to give a 4a OH adduct. The latter
is a covalent hydrate which can be converted to the
oxidized flavin by elimination of H
2
O. This hydrate
is believed to be the third spectral intermediate identi-
fied during the action of p-hydroxybenzoate hydroxy-
lase.
286,287,290
796 Chapter 15. Coenzymes of OxidationReduction Reactions
2. Oxidative Decarboxylation of Alpha-
Oxoacids
The oxidative cleavage of an -oxoacid is a major
step in the metabolism of carbohydrates and of amino
acids and is also a step in the citric acid cycle. In many
bacteria and in eukaryotes the process depends upon
both thiamin diphosphate and lipoic acid. The oxoacid
anion is cleaved to form CO
2
and the remaining acyl
group is combined with coenzyme A (Eq. 15-33).
NAD
+
serves as the oxidant. The reaction is catalyzed
by a complex of enzymes whose molecular mass varies
from ~ 4 to 10 x 10
6
, depending on the species and
exact substrate.
297
Separate oxoacid dehydrogenase
systems are known for pyruvate,
298300
2-oxoglut-
arate,
301
and the 2-oxoacids with branched side chains
derived metabolically from leucine, isoleucine, and
valine.
302,302a
In eukaryotes these enzymes are located
in the mitochondria. The pyruvate and 2-oxoglut-
arate dehydrogenase complexes of E. coli and Azoto-
bacter vinelandii have been studied most. In both cases
there are three major protein components. The first (E
1
)
is a decarboxylase (also referred to as a dehydrogenase)
for which the thiamin diphosphate is the dissociable
cofactor. The second (E
2
) is a lipoic acid amide-containing
core enzyme which is a dihydrolipoyl transacylase.
The third (E
3
) is the flavoprotein dihydrolipoyl
dehydrogenase, a member of the glutathione reduc-
tase family with a three-dimensional structure and
catalytic mechanism similar to those of glutathione
reductase (Figs. 15-10, 15-12).
303 305
Electron microscopy of the core dihydrolipoyl
transacylase from E. coli reveals a striking octahedral
symmetry which has been confirmed by X-ray diffrac-
tion.
306307a
The core from pyruvate dehydrogenase
has a mass of ~ 2390 kDa and contains 24 identical
99.5-kDa E
2
subunits. The 2-oxoglutarate dehydrogenase
from E. coli has a similar but slightly less symmetric
structure. Each core subunit is composed of three
domains. A lipoyl group is bound in amide linkage to
lysine 42 and protrudes from one end of the domain.
A second domain is necessary for binding to subunits
E
1
and E
3
, while the third major 250-residue domain
contains the catalytic acyltransferase center.
308,309
This
center closely resembles that of chloramphenicol acetyl-
transferase (Chapter 12).
310,311
The lipoyl
301,309,312
and
catalytic
307
domains of the dihydrolipoyl succinyl-
transferase from 2-oxoglutarate dehydrogenase resem-
ble those of pyruvate dehydrogenase and also of the
branched chain oxoacid dehydrogenase. The three
domains of the proteins are joined by long 25- to 30-
residue segments rich in alanine, proline, and ionized
hydrophilic side chains.
309
This presumably provides
flexibility for the lipoyl groups which must move from
site to site. The presence of unexpectedly sharp lines
in the proton NMR spectrum of the core protein may
be a result of this flexibility.
309
To obtain the X-ray structure of the core protein
it was necessary to delete the lipoyl- and E
1
(E
2
)- bind-
ing domains. The resulting 24-subunit structure is
shown in Fig 15-14A,B.
306
It has been assumed for
many years that 12 of the dimeric decarboxylase units
(E
1
) are bound to the 12 edges of the transacetylase
cube, while six (50.6 x 2 kDa) flavoprotein (E
3
) dimers
bind on the six faces of the cube. The active centers of
all three types of subunits are thought to come close
together in the regions where the subunits touch, per-
mitting the sequence of catalytic reactions indicated in
Fig. 15-15 to take place. Eukaryotic as well as some
bacterial pyruvate decarboxylases have a core of 60
subunits in an icosahedral array with 532 symmetry.
This can be seen in the image reconstructions of the
enzyme from Saccharomyces cerevisiae shown in Fig.
15-14C and D. A surprising discovery is that in this
yeast enzyme the E
3
units are not on the outside of
the 5-fold symmetric faces but protrude into the inner
cavity.
299
Each of the 12 E
3
subunits is assisted in
binding correctly to the E
2
core by a molecule of the
47-kDa E
3
-binding protein (BP), also known as pro-
tein X.
313,314
Absence of this protein is associated with
congenital lactic acidosis.
The unique function of lipoic acid is in the oxida-
tion of the thiamin-bound active aldehyde (Fig. 15-15)
in such a way that when the complex with thiamin
breaks up, the acyl group formed by the oxidative
decarboxylation of the oxoacid is attached to the
R C
O
COO

R C
O
S CoA
NAD
+
CoASH
NADH + H
+
CO
2
Thiamin
diphosphate
Lipoamide
FAD
(15-33)
S S
COOH
COOH
S SH S R
COOH
S SH C N
b
CN

+ H
+
a
RSH
(15-32)
797
dihydrolipoyl group at the S-8 position.
315
The lipoic
acid, which is attached to the flexible lipoyl domains
of the core enzyme on a 1.5-nm-long arm, apparently
first contacts the thiamin diphosphate site on one of
the decarboxylase subunits. Bearing the acyl group,
it now swings to the catalytic site on the core enzyme
where CoA is bound. The acyl group is transferred
to CoA producing a dihydrolipoyl group which then
swings to the third subunit where the disulfide loop
and a bound FAD of dihydrolipoamide dehydrogenase
reoxidize the lipoyl group. The reduced flavin-disul-
fide enzyme is then oxidized by NAD
+
(Fig. 15-15) by
the reverse of the mechanism depicted in Fig. 15-12.
Although the direct reaction of a lipoyl group
with the thiamin-bound enamine (active aldehyde)
is generally accepted, and is supported by recent
studies,
315a
an alternative must be considered.
315
Hexacyanoferrate (III) can replace NAD
+
as an oxidant
for pyruvate dehydrogenase and is also able to oxidize
nonenzymatically thiamin-bound active acetaldehyde
Figure 15-14 (A) View of the com-
plex down a fourfold axis through
the face of the cube. (B) Stereoscopic
view of three subunits of the 24-subunit
cubic core of the E. coli pyruvate
dehydrogenase complex. The view
is down the threefold axis through
opposite vertices of the cube. Bound
lipoate and CoA molecules are visible
in each subunit. (A) and (B) from
Knapp et al.
307a
(C) Image of the 60-
subunit core of yeast pyruvate dehydro-
genase reconstructed from electron
micrographs of frozen-hydrated com-
plexes. The five-, three-, and twofold
symmetries can be seen. The view is
along the threefold axis. 1. Truncated
E
2
; 2. Truncated E
2
plus binding
proteins; 3. Truncated E
2
+ binding
protein + E
3
. The reconstructions
show that the E
3
and BP components
are inside the truncated E
2
scaffold.
The morphologies of E
3
and BP are
not correctly represented since they
are not bound with icosahedral sym-
metry. (D) Stereoscopic image of
complex shown in B,3 with the front
half cut away to show the interior.
The conical objects protruding in are
the E
3
molecules. The arrow points
to a binding site for attachment of the
binding protein to the core. Courtesy
of J. K. Stoops et al.
299
C. Lipoic Acid and the Oxidative Decarboxylation of -Oxoacids
1 2 3
A B
C
D
10 nm
5 nm
10 nm
798 Chapter 15. Coenzymes of OxidationReduction Reactions
to 2-acetylthiamin, a compound in which the acetyl
group has a high group transfer potential (Eq. 14-22).
Thus, the lipoyl group could first oxidize the active
aldehyde to a thiamin-bound acyl derivative, and then
in the second step accept the acyl group by a nucleo-
philic displacement reaction. This mechanism fails
to explain the unique role of lipoic acid in oxidative
decarboxylation. However, as we will see in the next
section, oxidative decarboxylation does not always
require lipoic acid and acetylthiamin is probably an
intermediate whenever lipoamide is not utilized.
Within many tissues the enzymatic activities of the
pyruvate and branched chain oxoacid dehydrogenases
complexes are controlled in part by a phosphorylation
dephosphorylation mechanism (see Eq. 17-9). Phos-
phorylation of the decarboxylase subunit by an ATP-
dependent kinase produces an inactive phosphoenzyme.
A phosphatase reactivates the dehydrogenase to com-
plete the regulatory cycle (see Eq. 17-9 and associated
discussion). The regulation is apparently accomplished,
in part, by controlling the affinity of the protein for
CO
2
Thiamin
diphosphate
Thiamin-bound
active aldehyde
Core active center
dihydrolipoyl
acyltransferase
NADH + H
+
NAD
+
Lipoamide
dehydrogenase
Oxoacid
decarboxylase
S S
C O
C
O
C
O
SH
S C
O
HS
HS
H
+
C HO
R
S
N
R
CoA

S
O C
R
COO

SH
S
C
R
O H
S
N
+
R C
O
S CoA
Figure 15-15 Sequence
of reactions catalyzed by
-oxoacid dehydrogenases.
The substrate and product
are shown in boxes, and the
path of the oxidized oxoacid
is traced by the heavy arrows.
The lipoic acid head is
shown rotating about the
point of attachment to a core
subunit. However, a whole
flexible domain of the core is
also thought to move.
thiamin diphosphate.
315b
The lipoamide dehydrogenase
component of all three dehydrogenase complexes
appears to be the same.
3. Other Functions of Lipoic Acid
In addition to its role in the oxidative decarboxyla-
tion of 2-oxoacids, lipoic acid functions in the human
body as part of the essential mitochondrial glycine-
cleavage system described in Section E. It may also
participate in bacterial glycine reductase (Eq. 15-61)
and other enzyme systems. Dihydrolipoamide dehydro-
genase binds to G4-DNA structures of telomeres and
may have a biological role in DNA-binding.
315c
Lipoic
acid is being utilized as a nutritional supplement and
appears to help in maintaining cellular levels of gluta-
thione.
316
799
4. Additional Mechanisms of Oxidative
Decarboxylation
Pyruvate is a metabolite of central importance
and a variety of mechanisms exist for its cleavage.
The pyruvate dehydrogenase complex is adequate for
strict aerobes and is very important in aerobic bacteria
and in facultative anaerobes such as E. coli. Lactic acid
bacteria, which lack cytochromes and other heme
proteins, are able to carry out limited oxidation with
flavoproteins such as pyruvate oxidase.
317
However,
strict anaerobes have to avoid accumulation of reduced
pyridine nucleotides and use a nonoxidative cleavage
of pyruvate by pyruvate formate-lyase. The pyruvate
dehydrogenase complex of our bodies generates NADH
which can be oxidized in mitochondria to provide
energy to our cells. However, NADH is a weak reduc-
tant. Some cells, such as those of nitrogen-fixing
bacteria, require more powerful reductants such as
reduced ferredoxin and utilize pyruvate: ferredoxin
oxidoreductase. Escherichia coli, a facultative anaerobe,
is adaptable and makes use of all of these types of
pyruvate cleavage.
318
Pyruvate oxidase. The soluble flavoprotein
pyruvate oxidase, which was discussed briefly in
Chapter 14 (Fig. 14-2, Eq. 14-22), acts together with a
membrane-bound electron transport system to convert
pyruvate to acetyl phosphate and CO
2
.
319
Thiamin
diphosphate is needed by this enzyme but lipoic acid
is not. The flavin probably dehydrogenates the thiamin-
bound intermediate to 2-acetylthiamin as shown in
Eq. 15-34. The electron acceptor is the bound FAD
and the reaction may occur in two steps as shown
with a thiamin diphosphate radical intermediate.
319a
Reaction with inorganic phosphate generates the
energy storage metabolite acetyl phosphate.
S
N
C
CH
3
CH
2
CH
2
O-P-P
Pyrimidine
O
H
3
C
H
S
N
C
CH
3
CH
2
CH
2
O-P-P
Pyrimidine
O
H
3
C
S
N
C
CH
2
CH
2
O-P-P
Pyrimidine
O
H
3
C
e

Thiamin diphosphate radical Enamine


+
2-Acetyl-TDP
H
+
e

Thiamin diphosphate
H
3
C C
O
OPO
3
H

Acetyl phosphate
H
2
PO
3

(15-34)
Pyruvate:ferredoxin oxidoreductase. Within
clostridia and other strict anaerobes this enzyme
catalyzes reversible decarboxylation of pyruvate
(Eq. 15-35). The oxidant used by clostridia is the low-
potential ironsulfur ferredoxin.
320,320a
Clostridial
ferredoxins contain two FeS clusters and are therefore
two-electron oxidants. Ferredoxin substitutes for NAD
+
in Eq. 15-33 but the Gibbs energy decrease is much less
( 16.9 vs 34.9 kJ / mol. for oxidation by NAD
+
).
The enzyme does not require lipoic acid. It seems
likely that a thiamin-bound enamine is oxidized by an
ironsulfide center in the oxidoreductase to 2-acetyl-
thiamin which then reacts with CoA. A free radical
intermediate has been detected
318,321
and the proposed
sequence for oxidation of the enamine intermediate is
that in Eq. 15-34 but with the FeS center as the elec-
tron acceptor. Like pyruvate oxidase, this enzyme
transfers the acetyl group from acetylthiamin to coen-
zyme A. Cleavage of the resulting acetyl-CoA is used
to generate ATP. An indolepyruvate: ferredoxin oxi-
doreductase has similar properties.
322
In methanogenic bacteria
320
the low-potential
5-deazaflavin coenzyme F
420
serves as the reductant
in a reversal of Eq. 15-35. A similar enzyme, 2-oxo-
glutarate synthase, apparently functions in synthesis
of 2-oxoglutarate from succinyl-CoA and CO
2
by
photosynthetic bacteria.
323
Either reduced ferredoxin
or, in Azotobacter, reduced flavodoxin is generated in
nitrogen-fixing bacteria (Chapter 24) by cleavage of
pyruvate and is used in the N
2
fixation process.
Oxidative decarboxylation
by hydrogen peroxide. The
nonenzymatic oxidative decarb-
oxylation of -oxoacids by H
2
O
2
is well-known. The first step is
the formation of an adduct, an
organic peroxide, which breaks
up as indicated in Eq. 15-36. An
enzyme-catalyzed version of this
reaction is promoted by lactate
monooxygenase, a 360-kDa octa-
meric flavoprotein obtained from
Mycobacterium smegmatis
324
and a
member of the glycolate oxidase
family. Under anaerobic condi-
tions, the enzyme produces pyru-
vate by a simple dehydrogenation.
However, the pyruvate dissociates
slowly and in the presence of
C. Lipoic Acid and the Oxidative Decarboxylation of -Oxoacids
Pyruvate

+ CoA Acetyl-CoA + CO
2
Ferredoxin (ox) Ferredoxin (red)
G (pH 7) = 16.9 kJ mol
1
(15-35)
800 Chapter 15. Coenzymes of OxidationReduction Reactions
oxygen it forms acetic acid, with one of the oxygen
atoms of the carboxyl group coming from O
2
.
324,325
Hydrogen peroxide is the usual product formed from
oxygen by flavoprotein oxidases, and it seems likely
that with lactate monooxygenase the hydrogen peroxide
formed immediately oxidizes the pyruvate according
to Eq. 15-36. The -oxoacids formed by amino acid
oxidases in vitro are also oxidized by accumulating
hydrogen peroxide. However, if catalase (Chapter 16)
is present it destroys the H
2
O
2
and allows the oxoacid
to accumulate.
Pyruvate formate-lyase reaction. Anaerobic
cleavage of pyruvate to acetyl-CoA and formate
(Eq. 15-37) is essential to the energy economy of many
cells, including those of E. coli. No external oxidant is
needed, and the reaction does not require lipoic acid.
The reaction has sometimes been called the phosphoro-
clastic reaction because the product acetyl-CoA usually
reacts further with inorganic phosphate to form acetyl
phosphate (see Fig. 15-16). The latter can then transfer
its phospho group to ADP to form ATP.
The mechanism of the cleavage of the pyruvate
in Eq. 15-37 is not obvious. Thiamin diphosphate is
not involved, and free CO
2
is not formed. The first
identified intermediate is an acetyl-enzyme containing
a thioester linkage to a cysteine side chain. This is
cleaved by reaction with CoA-SH to give the final
product. A clue came when it was found by Knappe
and coworkers that the active enzyme, which is rapidly
inactivated by oxygen, contains a long-lived free
radical.
326
Under anaerobic conditions cells convert
the inactive form E
i
to the active form E
a
by an enzy-
matic reaction with S-adenosylmethionine and reduced
flavodoxin Fd(red) as shown in Eq. 15-38.
327 329
A
deactivase reverses the process.
330
Figure 15-16 Two systems for oxidative decarboxylation of -oxoacids and for substrate-level phosphorylation. The value
of G = +34.5 kJ mol
1
(Table 6-6) was used for the synthesis of ATP
4
from ADP
3
and HPO
4
2
in computing the values of G
given.
CH
3
COO

R C
O
COO

R C
O
S CoA
R C
O
O

H
3
C C
O
COO

CH
3
C
O
S CoA
CH
3
C
O
O PO
3
2
A Oxidative decarboxylation of an -oxoacid with thiamin diphosphate
2 Fd (ox)
or
NAD
+
2 Fd (red)
or
NADH
CoASH CO
2
Thiamin
lipoic acid
Pi
+
ADP(GDP) ATP(GTP)
CoASH
1. with lipoic acid and NAD
+
as oxidants
2. with ferredoxin as oxidant
G (pH 7, for pyruvate) = 35.5 kJ/mol overall
G (pH 7, for pyruvate) = 13.9 kJ/mol overall
The pyruvate formate-lyase reaction B
CoASH HCOO

CoASH
P
i
ADP ATP
G (pH 7) = 13.5 kJ/mol overall
Optional ATP-generating system
Optional ATP-generating system
R C
O
COO

R C
O
O

R C
OH
O
C
O
O

HO
H
+
H
2
O
2
H
2
O + CO
2
(15-36)
CH
3
C
COO

O
CoA SH
CH
3
C
O
S CoA
H C
O

O
G' (pH 7) = 12.9 kJ mol
1
(15-37)
801
The activating enzyme, which is allosterically
activated by pyruvate, is an ironsulfur (Fe
4
S
4
) protein
(Chapter 16).
331,331a
Formation of the observed radical
may proceed via a 5'-deoxyadenosyl radical as has
been proposed for lysine 2,3-aminomutase (Eq. 16-42).
332
The activation reaction also resembles the free radical-
dependent reactions of vitamin B
12
which are discussed
in Chapter 16. When subjected to O
2
of air at 25C
pyruvate formate-lyase is destroyed with a half-life
of ~ 10 s. The peptide chain is cleaved and sequence
analysis and mass spectrometry of the resulting frag-
ments show that the specific sequence Ser-Gly-Tyr at
positions 733 755 is cut with formation of an oxalyl
group on the N-terminal tyrosine of one fragment
(Eq. 15-39). Various
13
C-containing amino acids were
supplied to growing cells of E. coli and were incorpo-
rated into the proteins of the bacteria. Pyruvate formate-
lyase containing
13
C in carbon-2 of glycine gave an
EPR spectrum with hyperfine splitting arising from
coupling of the unpaired electron of the radical with
the adjacent
13
C nucleus.
333,333a
This experiment,
together with the results described by Eq. 15-39,
suggested that the radical is derived from Gly 734.
The activating enzyme will also generate radicals
from short peptides such as Arg-Val-Ser-Gly-Tyr-Ala-
Val, which corresponds to residues 731 737 of the
pyruvate formate-lyase active site. If Gly 734 is replaced
by L-alanine, no radical is formed, but radical is formed
if D-alanine is in this position. This suggests that the
pro-S proton of Gly 734 is removed by the activating
Ser C
O
NH CH C
O
NH Tyr

OOC C
O
NH Tyr
735 734 733
O
2
735
Glycine
radical
(15-39)
C. Lipoic Acid and the Oxidative Decarboxylation of -Oxoacids
enzyme
329
as illustrated in Eq. 15-40. It has been sug-
gested that the peptide exists in the -bend conforma-
tion shown.
How does the enzyme work? The -CH proton
of the glycyl radical of the original pro-R proton under-
goes an unexpected exchange with the deuterium of
2
H
2
O. The exchange is catalyzed by the thiol group
of Cys 419, suggesting that it is close to Gly 734 in the
three-dimensional structure.
334
The mutants C419S
and C418S are inactive but still allow formation of the
Gly 734 radical.
334335a
The mechanism has been pro-
posed in Eq. 15-41.
H
3
C S
CH
2
(adenosyl)
CH
2
CH
2
CH COO

5'
NH
3
+
+
e

(From flavodoxin H
2
)
Fe(II) in activating enzyme
Methionine
CH
2
(adenosyl)
Inactive pyruvate
formate lyase (E
i
)
Radical form
active enzyme (E
a
)
H
3
C (adenosyl)
5-Deoxyadenosine
(15-38)
(15-40)
N
C
C
O
H
S H
R
H
N
C
C
O
H CH
3
H
HO
N
C
C
O
N
O
H
H(CH
3
)
N H OH
O
O H
N H
Ser 733
Val
Tyr 735
Radical
Activating
enzyme
Activating
enzyme
d-Alanine
C
H H

OOC C CH
3
O
S
O
C
S
CH
3

O
C
C
H H
O

C
O
G734 C419
C
S
O CH
3
C419
G734 C419
C
H H
G734
S
H
C418
S
H
H
C
S
H
G734 C419 C418
HCOO CoA S C CH
3
O
CoA S

(15-41)
802 Chapter 15. Coenzymes of OxidationReduction Reactions
BOX 15-D FOLIC ACID (PTEROYLGLUTAMIC ACID)
In 1931, Lucy Wills, working in India, observed
that patients with tropical macrocytic anemia, a
disease in which the erythrocytes are enlarged but
reduced in numbers, were cured by extracts of yeast
or liver. The disease could be mimicked in monkeys
fed the local diet, and a similar anemia could be
induced in chicks. By 1938 it had also been shown
that a factor present in yeast, alfalfa, and other
materials was required for the growth of chicks.
Isolation of the new vitamin came rapidly after it
was recognized that it was also an essential nutrient
for Lactobacillus casei and Streptococcus faecalis R.
a,b
Spinach was a rich source of the new compound,
and it was named folic acid (from the same root as
the word foliage).
The microbiological activity attributed to folic
acid in extracts of natural materials was largely that
of di- and triglutamyl derivatives, one of the facts
that has led to the description of the history of folic
acid as the most complicated chapter in the story of
the vitamin B complex.
a
Metabolic functions for
folic acid were suggested by the observations that
the requirement for Streptococcus faecalis could be
replaced by thymine plus serine plus a purine base.
Folic acid is required for the biosynthesis of all of
these substances. A function in the interconversion
of serine and glycine was suggested by the observa-
tion that certain mutants of E. coli required either
serine or glycine for growth. Isotopic labeling experi-
ments established that in the rat as well as in the
yeast Torulopsis serine and glycine could be inter-
converted. It was also shown that the amount of
interconversion decreased in the folate-deficient rat.
Deficiency of folic acid is a common nutritional
problem of worldwide importance.
b
A recommended
daily intake is 0.2 mg, but because of the association
between low folic acid intake and neural tube defects
in infants, women of child-bearing age should have
0.4 mg / day.
c e
l-Glutamic
acid
para-Aminobenzoic
acid
N
N
HN
N
CH
2
N C
N
O
C
COOH
H
CH
2
CH
2
COOH
H
2
N
O
H
H
1
2
4
5
8
7
9
10
a
Wagner, A. F., and Folkers, E. (1964) Vitamins and Coenzymes,
Wiley (Interscience), New York
b
Jukes, T. H. (1980) Trends Biochem. Sci. 5, 112113
c
Cziezel, A. E., and Duds, I. (1992) N. Engl. J. Med. 327, 1832
1835
d
Jukes, T. H. (1997) Protein Sci. 6, 254256
e
Rosenquist, T. H., Ratashak, S. A., and Selhub, J. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1522715232
5. Cleavage of -Oxoacids and Substrate-Level
Phosphorylation
The -oxoacid dehydrogenases yield CoA deriva-
tives which may enter biosynthetic reactions. Alterna-
tively, the acyl-CoA compounds may be cleaved with
generation of ATP. The pyruvate formate-lyase system
also operates as part of an ATP-generating system for
anaerobic organisms, for example, in the mixed acid
fermentation of enterobacteria such as E. coli (Chapter
17). These two reactions, which are compared in Fig.
15-16, constitute an important pair of processes both
of which accomplish substrate-level phosphorylation.
They should be compared with the previously consid-
ered examples of substrate level phosphorylation
depicted in Eq. 14-23 and Fig. 15-16.
D. Tetrahydrofolic Acid and Other Pterin
Coenzymes
In most organisms reduced forms of the vitamin
folic acid serve as carriers for one-carbon groups at three
different oxidation levels corresponding to formic acid,
formaldehyde, and the methyl group and also facili-
tate their interconversion.
336,337
Moreover, folic acid
is just one of the derivatives of the pteridine ring
system that enjoy a widespread natural distribution.
338
One of these derivatives functions in the hydroxylation
of aromatic amino acids and in nitric oxide synthase
and yet another within several molybdenum-containing
enzymes. Pteridines also provide coloring to insect
wings and eyes and to the skins of amphibians and
fish. They appear to act as protective filters in insect
eyes and may function as light receptors. An example is
a folic acid derivative found in some forms of the DNA
photorepair enzyme DNA photolyase (Chapter 23).
1. Structure of Pterins
Because of the prevalence of its derivatives, 2-amino-
4-hydroxypteridine has been given the trivial name
pterin. Its structure resembles closely that of guanine,
the 5-membered ring of the latter having been expanded
to a 6-membered ring. In fact, pterins are derived
803
biosynthetically from guanine. The two-ring system of
pterin is also related structurally and biosynthetically
to that of riboflavin (Box 15-B).
Interest in pteridines began with Frederick G.
Hopkins, who in 1891 started his investigation of the
yellow and white pigments of butterflies. Almost 50
years and a million butterflies later, the structures of
the two pigments, xanthopterin and leucopterin
(Fig. 15-17), were established.
339
These pigments are
produced in such quantities as to suggest that their
synthesis may be a means of deposition of nitrogenous
wastes in dry form.
Among the simple pterins isolated from the eyes
of Drosophila
340
is sepiapterin (Fig. 15-17), in which
the pyrazine ring has been reduced in the 7,8 position
and a short side chain is present at position 6. Reduc-
tion of the carbonyl group of sepiapterin with NaBH
4
followed by air oxidation produces biopterin, the
most widely distributed of the pterin compounds.
First isolated from human urine, biopterin (Fig. 15-17)
is present in liver and other tissues where it functions
in a reduced form as a hydroxylation coenzyme
(see Chapter 18).
338
It is also present in nitric oxide
synthase (Chapter 18).
341,342
Other functions in oxida-
tive reactions, in regulation of electron transport, and
in photosynthesis have been proposed.
343
Neopterin,
found in honeybee larvae, resembles biopterin but has
a D-erythro configuration in the side chain. The red eye
pigments of Drosophila, called drosopterins, are com-
plex dimeric pterins containing fused 7-membered
rings (Fig. 15-17).
344,345
In the pineal gland, as well as in
the retina of the eye, light-sensitive
pterins may be photochemically
cleaved to generate such products
as 6-formylpterin, a compound
that could serve as a metabolic
regulator.
346
Another pterin acts as
a chemical attractant for aggregation
of the ameboid cells of Dictyostelium
lacteum
347
(Box 11-C). Molybdopterin
(Fig. 15-17) is a component of several
N
N N
CH
2
N
H
2
N
O
N
C
H
O
HN
H
H
COO

10
H
NH
O
5
H pK
a
= 10.5
COO

A proton adds here


with pK
a
= 4.82
The active site
H
S configuration
C
O
N
H
COO

OOC
H
Tetrahydrofolic acid (THF) or tetrahydropteroylglutamic acid (H
4
PteGlu)
Additional -linked glutamates
can be added to form tetrahydrofolyl-
triglutamic acid (THF-triglutamic
acid or H
4
PteGlu
3
) as shown
here or higher polyglutamates
H
NH
O
H
HN
COO

COO

OOC
N
N
Mo-containing enzymes discussed in Chapter 16,
348,349
and methanopterin is utilized by methanogenic
bacteria.
350 353
2. Folate Coenzymes
The coenzymes responsible for carrying single-
carbon units in most organisms are derivatives of
5,6,7,8-tetrahydrofolic acid (abbreviated H
4
PteGlu,
H
4
folate or THF). However, in methanogenic bacteria
the tetrahydro derivative of the structurally unique
methanopterin (Fig. 15-17) is the corresponding single-
carbon carrier.
353
Naturally occurring tetrahydrofolates
contain a chiral center of the S configuration.
354,355
They
exhibit negative optical rotation at 589 nm. The folate
coenzymes are present in extremely low concentrations
and the reduced ring is readily oxidized by air.
In addition to the single L-glutamate unit present
in tetrahydrofolic acid, the coenzymes occur to the
greatest extent as conjugates called folyl polyglutamates
in which one to eight or more additional molecules
of L-glutamic acid have been combined via amide
linkages.
356357a
The first two of the extra glutamates
are always joined through the (side chain) carboxyl
groups but in E. coli the rest are joined through their
carboxyls.
358
The distribution of the polyglutamates
varies from one organism to the next. Some bacteria
contain exclusively the triglutamate derivatives, while
in others almost exclusively the tetraglutamate or
octaglutamate derivatives predominate.
359
The serum
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
HN
N N
N
H
2
N
O
HN
N H
2
N
O
N
N
N
N N
N
Pterin: 2-amino-4-hydroxypteridine
H
Guanine
1
2
3
4
4a
5
6
7
8
8a
Pteridine
804 Chapter 15. Coenzymes of OxidationReduction Reactions
Figure 15-17 Structures of several biologically important pterins.
HN
N N
H
N O
H
2
N
O
Xanthopterin (yellow)
HN
N N
H
H
N O
H
2
N
O
Leucopterin (white)
O
HN
N N
H
N C
H
2
N
O
7
8
Sepiapterin
H
OH
O
CH
3
HN
N N
H
N
H
2
N
O
Drosopterin and isodrosopterin
H
H
3
C
NH
N
H
H
H
H
H
N
N NH
2
O
HN
N N
N
O
H
2
N CH
3
CH
HN
CH
3
O
HO
HO
H
H
OH
H
O
HO HO
O
P
O
O

O
C

OOC
COO

H
Methanopterin
HN
N N
N
H
2
N
O
l-erythro-Biopterin
HO H
H
OH
CH
3
Blue-green algae contain glycosides
with glucose, galactose, xylose, etc.,
attached possibly here
HN
N N
N
H
2
N
O
Urothione
S
S
OH
H
OH
CH
3
A urinary product thought to
be derived from molybdopterin
HN
N N
N
H
2
N
O
O
O
H
N
HO
OH

S
Mo
S

7
Molybdopterin
S
Mo
S
H
H
H
7

H
H
?
?
of many species contains only derivatives of folic acid
itself but pteroylpentaglutamate is the major folate
derivative present in rodent livers.
360,361
A large fraction
of these pentaglutamates consists of the 5-methyl-
THF derivative, while at the heptaglutamate level
most consists of the free THF derivative. Folyl poly-
glutamate in its oxidized form is a component of
some DNA photolyases (Chapter 23).
362
3. Dihydrofolate Reductase
Folic acid and its polyglutamyl derivatives can be
reduced to the THF coenzymes in two stages: the first
step is a slow reduction with NADPH to 7,8-dihydro-
folate (step a, Fig. 15-18). The same enzyme that cata-
lyzes this reaction rapidly reduces the dihydrofolates
805
to tetrahydrofolates (step b, Fig. 15-18). Again, NADPH
is the reducing agent and the enzyme has been given
the name dihydrofolate reductase. An unusual fact
is that bacteriophage T4 not only carries a gene for its
own dihydrofolate reductase but also the enzyme is a
structural unit in the phage baseplate.
363
Inhibitors. Aside from its role in providing reduced
folate coenzymes for cells, this enzyme has attracted a
great deal of attention because it appears to be a site of
action of the important anticancer drugs methotrexate
(amethopterin) and aminopterin.
293,364,365
These com-
pounds inhibit dihydrofolate reductase in concentra-
tions as low as 10
8
to 10
9
M. Methotrexate is also
widely used as an immunosuppresant drug and in
the treatment of parasitic infections.
Another dihydrofolate inhibitor trimethoprim is
an important antibacterial drug, usually given together
with a sulfonamide. Although it is not as close a
structural analog of folic acid as is methotrexate, it is
a potent inhibitor which binds far more tightly to the
enzyme from bacteria than to that from humans.
366 368
The inhibitors pyrimethamine and cycloguanil are
effective antimalarial drugs.
369,370
An enormous number of synthetic compounds have
been prepared in the hope of finding still more effec-
tive inhibitors. By 1984, over 1700 different inhibitors
of dihydrofolate reductase had been studied.
367
However, methotrexate and trimethoprim remain
outstanding. Methotrexate has been in clinical use for
nearly 50 years and is very effective against leukemia
and some other cancers.
371
Before 1960 persons with
acute leukemia lived no more than 3 6 months. How-
ever, with antifolate treatment some have lived for 5
years or more and complete cures of the relatively rare
choriocarcinoma have been achieved. New methods
of chemotherapy use the antifolates in combination
with other drugs.
Folate coenzymes are required in the biosynthesis
of both purines and thymine. Consequently, rapidly
growing cancer cells have a high requirement for
activity of this enzyme. However, since all cells require
the enzyme the antifolates are toxic and cannot be used
for prolonged therapy. An even more
serious problem is the development of
resistance to the drug by cancer cells,
often through amplification of the
dihydrofolate reductase gene
365,372 374
as discussed in Chapter 27. Cells may
also become resistant to methotrexate
and other antifolates as a result of
mutations that prevent efficient uptake
of the drug,
375
cause increased action
of effux pumps in the cell,
376
reduce
the affinity of dihydrofolate reductase
for the drug,
377
or interfere with conversion of the
drug to polyglutamate derivatives which are better
inhibitors than free methotrexate.
378,379
Cell surface
folate receptors, present in large numbers in some
tumor cells, can be utilized to bring suitably designed
antifolate drugs or even unrelated cytotoxic com-
pounds into tumor cells.
379a
Some tumor cells are
more active than normal cells in generating folyl poly-
glutamates, contributing to the effectiveness of meth-
otrexate.
379
Resistance of E. coli cells to trimethoprin
may result from acquisition by the bacteria of a new
form of dihydrofolate reductase carried by a plasmid.
380
Structure and mechanism. Dihydrofolate reduc-
tase from E. coli is a small 159-residue protein with a
central parallel stranded sheet,
381 383
while that from
higher animals is 20% larger. The three-dimensional
structures of the enzymes from Lactobacillus,
384
chicken,
mouse, and human are closely similar. The NADP
binds at the C-terminal ends of strands as in other
dehydrogenases. In Fig. 15-19 the reduced nicotinamide
end of NADP
+
is seen next to a molecule of bound
dihydrofolate.
381
The side chain carboxylate of Asp 27
makes a pair of hydrogen bonds to the pterin ring as
shown on the right-hand side of Eq. 15-42. As can be
seen from Fig. 15-19, the nicotinamide ring of NADP
+
is correctly positioned to have donated a hydride ion to
C-6 to form THF with the 6S configuration. Notice that
in Eq. 15-42 the NH hydrogen atom at the 3' position
N
N N
N
H
2
N
NH
2
N C
N
C
COO

COO

H
O
H
H
Aminopterin (4-amino-4-deoxyfolic acid)
Methotrexate contains CH
3
here
N
N
NH
2
H
2
N
CH
2
O-CH
2
CH
2
-OCH
3
OCH
3
OCH
3
Trimethoprim
Cl
N
N
NH
2
H
2
N CH
2
CH
3
Pyrimethamine
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
806 Chapter 15. Coenzymes of OxidationReduction Reactions
Figure 15-18 Tetrahydrofolic acid and its one-carbon derivatives.
HN
N N
N
HN
R
H
2
N
O
HN
N N
N
HN
R
H
2
N
O
H
H
H
*
C
O
N
C
COO

H COO

H
HN
N N
N
H
2
N
O
H
CH
3
HN
R
H C
O

O
a
b
c
NADPH
DHF
NADPH
Serine
Glycine
Gly
CO
2
c
H C
H
O
or
(nonenzymatic)
2-Oxopantoate
2-Oxovalerate
Folate
or polyglutamates
NADPH NADP
+
DHF
Thymine (thymidylate)
Hydroxymethylcytosine
(hydroxymethyldeoxy CMP) d
NADPH
NADP
+
NH
3
Purines Histidine
Formimino-Gly Formimino-Glu
5-Formimino-THF 5,10-Methenyl-THF 5-Formyl-THF (leucovorin)
h
e
H
2
O
ADP
+ P
i
ATP
OH

g
ADP
+ P
i
ATP
e
n
Purines
Formylmethionine
o
NADPH
NADP
+
CO
p
H
2
O
g
l
m
f
k
j
i
5-Methyl-THF
Homocysteine
Methionine
Methane
R=
Formate
Tetrahydrofolate
5,10-Methylene-THF
10-Formyl-THF CO
2
HN
N N
N
H
2
N
O
H
H
6
N
H
2
C
R
HN
N N
N
H
2
N
O
H
CH HN R
HN
HN
N N
N
H
2
N
O
H
N
HC
R
+
HN
N N
N
H
2
N
O
H
5
HN R HC
O
HN
N N
N
H
2
N
O
H
10
N R
HC
O
H
5
807
of the pterin makes a hydrogen bond to
Asp 27. However, X-ray studies have shown
that the pteridine ring of methotrexate is
turned 180 so that its 4-NH
2
group forms
hydrogen bonds to backbone carbonyls of
Ala 97 and Leu 4 at the edge of the central
pleated sheet while a protonated N-1
interacts with the side chain carboxylate
of Asp 27.
Because of its significance in cancer
therapy and its small size, dihydrofolate
reductase is one of the most studied of all
enzymes. Numerous NMR studies
385 387
and investigations of catalytic mechanism
and of other properties
388
have been
conducted. Many mutant forms have been
created.
389391
For example, substitution
of Asp 27 (Asp 26 in L. casei) of the E. coli
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
N
N N
N H
H
N
R
O
H
H O
H
H
O
O Asp 27
N
N N
N H
H
N
R
O
H

H
O
O Asp 27
3
1
H
Tautomerized dihydropterin ring
pK
a
=6.5
H
H
H
O
H H
5
6
8
Tetrahydrofolate
Figure 15-19 Drawings of the active site of E. coli dihydrofolate reductase showing the bound ligands NADP
+
and tetrahydro-
folate. Several key amino acid side chains are shown in the stereoscopic views on the right. The complete ribbon structures
are on the left. (A) Closed form. (B) Open form into which substrates can enter and products can escape. From Sawaya and
Kraut.
381
Courtesy of Joseph Kraut. Molscript drawings (Kraulis, 1991).
(15-42)
Closed
Open
A
B
808 Chapter 15. Coenzymes of OxidationReduction Reactions
enzyme by Asn reduced the specific cata-
lytic activity to 1/300 that of native enzyme
indicating that this residue is important for
catalysis.
390
The value of k
cat
for reduction
of dihydrofolate is highest at low pH and
varies around a pK
a
of ~ 6.5.
392 394
One
interpretation is that this pK
a
belongs to the
Asp 27 carboxyl group and another is that it
belongs to an N-5 protonated species of the
coenzyme. As we have seen (Chapter 6) it is
often impossible to assign a pK
a
to a single
group because there may be a mixture of
interacting tautomeric species. Despite the
enormous amount of study, we still dont
know quite how the proton gets to N-5 in
this reaction. Only one possibility is illus-
trated in Eq. 15-42. Asp 27 is protonated,
the pterin ring is enolized, and a buried
water molecule serves to relay a proton to
N-5. The X-ray crystallographic studies
on the E. coli enzyme show that after the
binding of substrates a conformational
change closes a lid over the active site (Fig.
15-19). This excludes water from N-5 but it
may permit intermittent access, allowing
transfer of a proton from a water molecule
bound to O-4 as shown in Eq. 15-42.
381
During the reduction of folate to dihydro-
folate a different mechanism of proton dona-
tion must be followed to allow protonation at C-7.
4. Single-Carbon Compounds and Groups in
Metabolism
Some single-carbon compounds and groups impor-
tant in metabolism are shown in Table 15-3 in order
(from left to right) of increasing oxidation state of the
carbon atom. Groups at three different oxidation
levels, corresponding to formic acid, formaldehyde,
and the methyl group, are carried by tetrahydrofolic
acid coenzymes. While the most completely reduced
compound, methane, cannot exist in a combined form,
its biosynthesis depends upon reduced methanopterin,
as does that of carbon monoxide. Figure 15-18 sum-
marizes the metabolic interrelationships of the com-
pounds and groups in this table.
Serine as a C1 donor. For many organisms, from
E. coli to higher animals, serine is a major precursor of
C-1 units.
395 397
The -carbon of serine is removed as
formaldehyde through direct transfer to tetrahydro-
folate with formation of methylene-THF and glycine
(Eq. 14-30, Fig. 15-18, step c). This is a stereospecific
transfer in which the pro-S hydrogen on C-3 of serine
enters the pro-S position also in methylene-THF.
398
The glycine formed in this reaction can, in turn, yield
another single-carbon unit by loss of CO
2
under the
influence of the THF and the PLP-requiring glycine
cleavage system which is discussed in the next para-
graph. Free formaldehyde in a low concentration can
also combine with THF to form methylene-THF (Fig.
15-18, step c and Eq. 15-43).
399
The glycine decarboxylasesynthetase system.
Glycine is cleaved reversibly within mitochondria of
CH
4
Methane
H
3
C Y
Methyl groups
bound to O, N, S
H C
O
H
Formaldehyde
H C
O
H
Formic acid
CO
2
H
2
CO
3
CH
2
OH
Hydroxymethyl
group
C
O
H
Formyl group
CH
2
Methylene group Methenyl group
Formimino group
Carbon monoxide
C
H
C O
C
H
NH
TABLE 15-3
Single-Carbon Compounds in Order of Oxidation State of
Carbon
(15-43)
CH
2
N
N
HO
H
10
5
H
2
C
N
N
H
C
N
N
THF
H
2
CO
+
HO

H
+
H H
809
plants and animals and also by bacteria to CO
2
, NH
3,
and a methylene group which is carried by tetrahydro-
folic acid
296,400 403
(Fig. 15-20). Four proteins are required.
The P-protein consists of two identical 100-kDa subunits,
each containing a molecule of PLP. This protein is a
glycine decarboxylase which, however, replaces the
lost CO
2
by an electrophilic sulfur of lipoate rather than
by a proton. Serine hydroxymethyltransferase can also
catalyze this step of the sequence.
404
The lipoate is
bound to a second protein, the H-protein. A third
protein, the T-protein, carries bound tetrahydrofolate
which displaces the aminomethyl group from the
dihydrolipoate and converts it to N
5
,N
10
-methylene
Schiff base
PLP bound
to P-protein
CO
2
Glycine
H-protein
NADH + H
+
Lipoamide
dehydrogenase
NAD
+
N
5
, N
10
-Methylene
THF
NH
3
P-protein
H
H
HN
O H
N C
CH
2
OP
S S
C O
C
O
C
O
N
N
H
SH
S CH
2
NH
3
+
H
H
HS
HS
H
+
THF bound
to T-protein
5
N
N
N
N
H
2
C N O
H
2
N
NH
3
+
H
H
H
H
10
Figure 15-20 The reversible glycine cleavage system of mitochondria. Compare with Fig. 15-15.
tetrahydrofolate with release of ammonia. The
dihydrolipoate is then reoxidized by NAD
+
and dihy-
drolipamide dehydrogenase (Fig. 15-20). Glycine can
be oxidized completely in liver mitochondria with the
methylene group of methylene-THF being converted
to CO
2
through reaction steps d, e, and f of Fig. 15-18.
The glycine cleavage system is reversible and is used
by some organisms to synthesize glycine.
Whether it arises from the hydroxymethyl group
of serine or from glycine, the single-carbon unit of
methylene-THF (which is at the formaldehyde level of
oxidation) can either be oxidized further to 5,10-meth-
enyl-THF and 10-formyl-THF (steps d and e, Fig. 15-18)
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
810 Chapter 15. Coenzymes of OxidationReduction Reactions
or it can be reduced to methyl-THF (step i). The reac-
tions of folate metabolism occurs in both cytoplasm and
mitochondria at rates that are affected by the length of
the polyglutamate chain. The many complexities of
these pathways are not fully understood.
395,397
Starting with formate or carbon dioxide. Most
organisms can, to some extent, also utilize formate as
a source of single-carbon units. Human beings have
a very limited ability to metabolize formate and the
accumulation of formic acid in the body following
ingestion of methanol is often fatal. However, many
bacteria are able to subsist on formate as a sole carbon
source. Archaea may generate the formate by reduction
of CO
2
. Utilization of formate begins with formation
of 10-formyl-THF (Fig. 15-18, step g, lower left corner).
10-Formyl-THF can be reduced to methylene-THF and
the single-carbon unit can be transferred to glycine to
form serine. In some bacteria three separate enzymes
are required to convert formate to methylene-THF:
10-formyl-THF synthetase (Fig. 15-18, step g, presumably
via formyl phosphate),
405
methenyl-THF cyclohydrolase
(step e, reverse), and methylene-THF dehydrogenase
(step d, reverse). NADH is used by the acetogens but
in E. coli and in most higher organisms NADPH is the
reductant. In some bacteria and in some tumor cells,
two of the three enzymes are present as a bifunctional
enzyme.
406407a
In most eukaryotes all three of the
enzymatic activities needed for converting formate to
methylene-THF are present in a single large (200-kDa)
dimeric trifunctional protein called formyl-THF
synthetase.
407,408
N
10
-Formyl-THF serves as a biological formylating
agent needed for two steps in the synthesis of pu-
rines
409410b
(Chapter 25) and, in bacteria and in mito-
chondria and chloroplasts, for synthesis of formyl-
methionyl-tRNA
411
which initiates synthesis of all
polypeptide chains in bacteria and in these organelles
(Chapter 29).
N
5
-Formyl-THF (leucovorin). The N
5
-formyl
derivative of THF (5-formyl-THF) is a growth factor for
Leuconostoc citrovorum. Following this discovery in 1949
it was called the citrovorum factor or leucovorin.
Its significance in metabolism is not clear. Perhaps it
serves as a storage form of folate in cells that have a
dormant stage, e.g., of seeds or spores.
412
It may also
have a regulatory function.
413
In some ants and in
certain beetles, it is stored and hydrolyzed to formic
acid. The carabid beetle Galerita lecontei ejects a defen-
sive spray that contains 80% formic acid.
414
5-Formyl-THF can arise by transfer of a formyl
group from formylglutamate and there is an enzyme
that converts 5-formyl-THF to 5,10-methenyl-THF
with concurrent cleavage of ATP.
412
5-Formyl-THF is
sometimes used in a remarkable way to treat certain
highly malignant cancers. Following surgical removal
of the tumor the patient is periodically given what
would normally be a lethal dose of methotrexate, then,
about 36 h later, the patient is rescued by injection of
5-formyl-THF. The mechanism of rescue is thought
to depend upon the compounds rapid conversion to
10-formyl-THF within cells. Tumor cells are not rescued,
perhaps because they have a higher capacity than
normal cells for synthesis of polyglutamates of metho-
trexate.
379
This observation also suggests that the major
anti-cancer effect of methotrexate may not be on dihy-
drofolate reductase but on enzymes of formyl group
transfer that are inhibited by polyglutamate derivatives
of methotrexate.
Catabolism of histidine and purines. Another
source of single-carbon units in metabolism is the
degradation of histidine which occurs both in bacteria
and in animals via formiminoglutamate. The latter
transfers the CH = NH group to THF forming 5-form-
imino-THF, which is in turn converted (step h, Fig. 15-18)
to 5,10-methenyl-THF and ammonia. In bacteria that
ferment purines, formiminoglycine is an intermediate.
Again, the formimino group is transferred to THF and
deaminated, the eventual product being 10-formyl-THF.
In these organisms, the enzyme 10-formyl-THF synthase
also has a very high activity.
415
It probably operates
in reverse as a mechanism for synthesis of ATP in
this type of fermentation. In other organisms excess
10-formyl-THF may be oxidized to CO
2
via an NADP
+
-
dependent dehydrogenase (step f, Fig. 15-16), providing
a mechanism for detoxifying formic acid. In some
organisms excess 10-formyl-THF may simply be hydro-
lyzed with release of formate (step q).
415a
Thymidylate synthase. Methylene-THF serves
as the direct precursor of the 5-methyl group of thy-
mine as well as of the hydroxymethyl groups of
hydroxymethylcytosine
416
and of 2-oxopantoate, an
intermediate in the formation of pantothenate and coen-
zyme A.
417
The latter is a simple hydroxymethyl trans-
fer reaction (Eq. 15-44) that is related to an aldol
condensation and which may proceed through an
imine of the kind shown in Eq. 15-43.
OOC CH
3
O

H
3
C H
N
N
H
2
C
5
10
OOC CH
2
OH
O

H
3
C CH
3
N
5,10
- Methylene- THF (15-44)
811
During thymine formation the coenzyme is oxi-
dized to dihydrofolate, which must be reduced by
dihydrofolate reductase to complete the catalytic cycle.
A possible mechanistic sequence for thymidylate
synthase, an enzyme of known three-dimensional
structure,
354,418421a
is given in Fig. 15-21. In the first step
(a) a thiolate anion, from the side chain of Cys 198 of
the 316-residue Lactobacillus enzyme, adds to the 5
position of the substrate 2'-deoxyuridine monophosphate
Figure 15-21 Probable mechanism of action of thymidylate synthase. After Huang and Santi
422
and Hyatt et al.
354
a
b
d
f
dUMP
Methylene-THF
Thymidylate (dTMP)
c
SEnz
HB
SEnz
AH
e
SEnz
SEnz
H
2
folate
HN
N
O
CH
3
O

HN
N
O
CH
2
O
HN NH
R H
HN
N
O
CH
3
O
H
A
HN
N
HO
CH
2
O
N NH
R H
+
H
A
N NH
R H
CH
2 HN
N
H
O
O
SEnz
H
B
AH
HN
N
HO
H
O SEnz
N NH
R H
+
CH
2
A
HN
N
O
H
O
SEnz
O
OH
O P
H O H199
H
O
C
E160
O
H O
H
N
C
O
N229
H

(dUMP). As a consequence the 6 position becomes a


nucleophilic center which can combine with methylene-
THF as shown in step b of Fig. 15-21. After tautomer-
ization (step c) this adduct eliminates THF (step d) to
give a 5-methylene derivative of the dUMP. The latter
immediately oxidizes the THF by a hydride ion transfer
to form thymidylate and dihydrofolate.
422,422a
In protozoa thymidylate synthase and dihydro-
folate reductase exist as a single bifunctional protein.
D. Tetrahydrofolic Acid and Other Pterin Coenzymes
A:, AH, B:; catalytic basic or acidic groups
812 Chapter 15. Coenzymes of OxidationReduction Reactions
Consequently, in methotrexate-resistant strains of
Leishmania (and also in cancer cells) both enzyme
activities are increased equally by gene amplification.
423
This presents a serious problem in the treatment of
BOX 15-E THYMIDYLATE SYNTHASE, A TARGET ENZYME IN CANCER CHEMOTHERAPY
a
If an animal or bacterial cells are deprived of
thymine they can no longer make DNA. However,
synthesis of proteins and of RNA continues for
some time. This can be demonstrated experimen-
tally with thymine-requiring mutants. However,
sooner or later such cells lose their vitality and die.
The cause of this thymineless death
b
is not entirely
clear. Perhaps thymine is needed to repair damage
to DNA and if it is not available transcription
eventually becomes faulty. Chromosome breakage
is also observed.
c
Whatever the cause of death, the
phenomenon provides the basis for some of the
most effective chemotherapeutic attacks on cancer.
Rapidly metabolizing cancer cells are especially
vulnerable to thymineless death. Consequently,
thymidylate synthase is an important target enzyme
for inhibition. One powerful inhibitor is the mono-
phosphate of 5-fluoro-2'-deoxyuridine. The
inhibition was originally discovered when 5-fluoro-
uracil was recognized as a useful cancer chemo-
therapeutic agent.
Fluorouracil has many effects in cells, including
incorporation into RNA,
b
but the inhibition of
thymidylate synthase by the reduction product may
be the most useful effect in chemotherapy. In fact,
5-fluoro-2'-deoxyuridine is a much less toxic and
5-Fluorouridine 5-phosphate Triphosphate RNA
Reduction (via diphosphate)
5-F-2-deoxyuridine
5-Fluoro-2-
deoxyuridine
5-phosphate
5-Fluorouracil
dUMP
dTMP
Thymidylate
synthetase
Methylene-THF
THF
Dihydrofolate
Dihydrofolate
reductase
NADP
Antifolates
NADPH

+
HN
N
O
F
O
H
more potent drug than 5-fluoro-
uracil. It binds into the active
site of thymidylate synthase
and reacts in the initial steps of
catalysis. However, the 5-H of
2'-deoxyuridine, the normal
substrate, must be removed as
H
+
(step c of Fig. 15-21) in order
for the reaction to continue. The
5-F atom cannot be removed in
this way and a stable adduct is
formed.
d,e
A crystal structure
containing both the bound
5-fluorodeoxyuridine and
methylene-THF supports this
conclusion.
e
Thymidylate synthase
requires methylene tetrahydro-
folate as a reductant and the
reduction of dihydrofolate is also
an important part of the process.
In protozoa dihydrofolate reduc-
tase and thymidylate synthase occur as a single-
chain bifunctional enzyme.
f
As has been pointed
out in the main text, such folic acid analogs as
methotrexate are among the most useful anticancer
drugs. By inhibiting dihydrofolate reductase they
deprive thymidylate synthase of an essential
substrate.
Because 5-fluorouracil acts on normal cells as
well as cancer cells, its usefulness is limited. Know-
ledge of the chemistry and three-dimensional
structure of thymidylate synthase complexes is
being used in an attempt to discover more specific
and effective drugs that attack this enzyme.
g,h
a
Friedkin, M. (1973) Adv. Enzymol. 38, 235292
b
Sahasrabudhe, P. V., and Gmeiner, W. H. (1997) Biochemistry 36,
59815991
c
Ayusawa, D., Shimizu, K., Koyama, H., Takeishi, K., and Seno,
T. (1983) J. Biol. Chem. 258, 1244812454
d
Huang, X. F., and Arvan, P. (1995) J. Biol. Chem. 270, 20417
20423
e
Hyatt, D. C., Maley, F., and Montfort, W. R. (1997) Biochemistry
36, 45854594
f
Ivanetich, K. M., and Santi, D. V. (1990) FASEB J. 4, 15911597
g
Shoichet, B. K., Stroud, R. M., Santi, D. V., Kuntz, I. D., and
Perry, K. M. (1993) Science 259, 14451449
h
Weichsel, A., Montfort, W. R., Ciesla, J., and Maley, F. (1995)
Proc. Natl. Acad. Sci. U.S.A. 92, 34933497
protozoal parasitic diseases for which few suitable
drugs are known.
Cells of E. coli, when infected with T-even bacterio-
phage, convert dCMP to 5-hydroxymethyl-dCMP
423a
813
and suitably infected cells of Bacillus
subtilis form 5-hydroxymethyl-
dUMP.
424,425
These products can be
formed by attack of OH

on quinonoid
intermediates of the type postulated for
thymidylate synthetase (formed in step
d of Fig. 15-21).
416
A related reaction
is the posttranscriptional conversion
of a single uracil residue in tRNA mole-
cules of some bacteria to a thymine ring
(a ribothymidylic acid residue). In this
case, FADH
2
is used as a reducing agent
to convert the initial adduct of Fig. 15-21
to the ribothymidylic acid and THF.
426
Synthesis of methyl groups. The
reduction of methylene-THF to 5-methyl-
THF within all living organisms from
bacteria to higher plants and animals
provides the methyl groups needed in
biosynthesis.
426a
These are required for
formation of methionine and, from it,
S-adenosylmethionine. The latter is used to modify
proteins, nucleic acids, and other biochemicals through
methylation of specific groups. In the methanogens,
reduction of a corresponding methylene derivative of
methanopterin gives rise to methane (Section F).
Mammalian methylene-THF reductase is a FAD-
containing flavoprotein that utilizes NADPH for the
reduction to 5-methyl-THF.
427,428
Matthews
429
suggested
that the mechanism of this reaction involves an internal
oxidationreduction reaction that generates a 5-methyl-
quinonoid dihydro-THF (Eq. 15-45). Methylene-THF
reductase of acetogenic bacteria is also a flavoprotein
but it contains FeS centers as well. The 237-kDa
4

4
oligomer contains two molecules of FAD and four to
six of both Fe and S
2
ions.
430,431
The methyl group of methyl-THF is incorporated
into methionine by the vitamin B
12
-dependent methio-
nine synthase which is discussed in Chapter 16. Mat-
thews suggested that methionine synthase may also
make use of the 5-methyl-quinonoid-THF of Eq. 15-45.
An initial reduction step would precede transfer of the
methyl group. Methyl-THF is a precursor to acetate in
HN
N N
N
H
2
C
CH
2
H
2
N
O N
5
10
R
H
+
HN
N N
N
H
2
C
CH
2
H
2
N
O
N R
+
H
H
+
H
HN
N N
N
CH
3
CH
2
NHR
HN
O
H
+
Methylene- THF
Internal redox
reaction
5- Methyl - quinonoid- THF
HN
N N
N
CH
3
CH
2
NHR
H
2
N
O
H
Methyl - THF
FAD
NADPH NADP
+
H
(15-45)
the acetogenic bacteria.
432
This corrinoid coenzyme-
dependent process is also considered in Chapter 16.
E. Specialized Coenzymes of Methanogenic
Bacteria
Methane-producing bacteria
433,434
obtain energy
by reducing CO
2
with molecular hydrogen:
CO
2
+ 4H
2
CH
4
+ 2H
2
O
G

= 131 kJ / mol
Some species are also able to utilize formate or formal-
dehyde as reducing agents.
435
These compounds are
oxidized to CO
2
, the reducing equivalents formed being
used to reduce CO
2
to methane. Carbon monoxide can
also be converted to CO
2
.
CO + H
2
O CO
2
+ H
2
G

= 20 kJ / mol
E. Specialized Coenzymes of Methanogenic Bacteria
(15-47)
(15-46)
HS CH
2
CH
2
SO
3

Coenzyme M
HS (CH
2
)
6
C
O
N
H
C
C
7-Mercaptoheptanoylthreonine phosphate
H
CH
3
O PO
3
2
COO
H

OOC
CH
CH N
H
CH
H
N
CH N
H
COO

COO

O COO

O COO

O
O CH
2
O
CH
2
NH
2
Methanofuran
O
R
CH
2
H
N C
O
H
Formylmethanofuran
814 Chapter 15. Coenzymes of OxidationReduction Reactions
Figure 15-22 Tentative scheme for reduction of carbon dioxide to
methane by methanogens. After Rouvire et al.
352
and Thauer et al.
435
a
b
c
d
e
f
g
H
2
X
X + H
2
O
H
2
MFR
5-Formyl-H
4
MPT
H
2
O
5,10-Methenyl-H
4
MPT
5,10-Methylene-H
4
MPT
5-CH
3
-MPT
CoM-SH
H-S-HTP
CoM-S-S-HTP
CH
4
F
430
[Ni]
See Fig. 16-27
X
H
2
X
CO
2
H
2
F
420
H
2
F
420
H
2
F
420
H
2
F
420
H
2
h
O
NH
3
R
+
Methanofuran
MFR
O
N
R
Carbamate
C
O
O
H

O
N
R
Formyl-MFR
C
O
H H
HN
N N
N
O
H
2
N
R'
H
H
5
H
4
-Methanopterin
H
4
MPT
H
3
C S
SO
3

Methyl-CoM
i
By using the combined reactions of Eqs.
15-46 and 15-47 the bacteria can subsist
on CO alone (Eq. 15-48):
4CO + 2 H
2
O 3CO
2
+ CH
4
G

= 211 kJ / mol of
methane formed
Some species reduce methanol to methane
via Eqs. 15-49 or 15-50.
CH
3
OH + H
2
CH
4
+ H
2
O
G

= 113 kJ / mol
4CH
3
OH 3CH
4
+ CO
2
+ 2 H
2
O
G

= 107 kJ / mol of
methane formed
To accomplish these reactions a
surprising variety of specialized cofac-
tors are needed.
351,352,434
The first of
these, coenzyme M, 2-mercaptoethane
sulfonate, was discovered in 1974.
436
It
is the simplest known coenzyme. Later,
the previously described 5-deazaflavin
F
420
(Section B), a nickel tetrapyrrole
F
430
(Chapter 16), methanopterin (Fig.
15-17),
437
the carbon dioxide reduction
factor called methanofuran,
352,438
and
7-mercaptoheptanoylthreonine
phosphate
439,440
were also identified.
A sketch of the metabolic pathways
followed in methane formation is given
in Fig. 15-22.
352,435
In the first step (a)
the amino group of methanofuran is
thought to add to CO
2
to form a carbam-
ate which is reduced to formylmethano-
furan by H
2
and an intermediate carrier
H
2
X in step b. The formyl group is then
transferred to tetrahydromethanopterin
(H
4
MPT) (step c)
440a
and is cyclized and
reduced in two stages in steps d, e, and f.
The reductant is the deazaflavin F
420
and
the reactions parallel those for conver-
sion of formyl-THF to methyl-THF
(Fig. 15-18).
431,440b,441
The methyl group
of methyl-H
4
MPT is then transferred to
the sulfur atom of the thiolate anion of
coenzyme M, from which it is reduced
off as CH
4
. This is a complex process
requiring the nickel-containing F
430
,
FAD, and 7-mercaptoheptanoylthreo-
nine phosphate (HS-HTP).
The HS-HTP may be the 2-electron
donor for the reduction. The mixed-disulfide
CoM-S-S-HTP appears to be an intermediate and also
an allosteric effector for the first step, the reduction of
(15-50)
(15-48)
(15-49)
CO
2
to formylmethanofuran. A complex of proteins
that is unstable in oxygen is also needed. The principal
component of the methyl reductase binds two molecules
815
have also been studied intensively. The 450-residue
soluble enzyme from Acinetobacter calcoaceticus is par-
tially homologous to the methanol dehydrogenase.
446a,446b
PQQ and the other quinone prosthetic groups
described here all function in reactions that would be
possible for pyridine nucleotide or flavin coenzymes.
All of them, like the flavins, can exist in oxidized, half-
reduced semiquinone and fully reduced dihydro
forms. The questions to be asked are the same as we
asked for flavins. How do the substrates react? How
is the reduced cofactor reoxidized? In nonenzymatic
reactions alcohols, amines, and enolate anions all add
at C-5 of PQQ to give adducts such as that shown for
methanol in Eq. 15-51, step a.
444,449,449a
Although many
additional reactions are possible, this addition is a
reasonable first step in the mechanism shown in Eq.
15-51. An enzymatic base could remove a proton as is
indicated in step b to give PQQH
2
. The pathway for
reoxidation (step c) might involve a cytochrome b,
cytochrome c, or bound ubiquinone.
445,446
Although the soluble PQQ-dependent glucose dehydro-
genase forms, with methylhydrazine, an adduct similar
to that depicted in Eq. 15-51,
449b
the structure of a
glucose complex of the PPQH
2
- and Ca
2+
-containing
enzyme at a resolution of 0.2 nm suggests a direct
hydride ion transfer. The only base close to glucose
C1 is His 144. The C1-H proton lies directly above the
PQQH
2
C5 atom. Oubrie et al.
446a
propose a deproto-
nation of the C1-OH by His 144 and H

transfer to
C5 of PQQ followed by tautomerization (Eq. 15-52).
Theoretical calculations by Zheng and Bruice
449c
favor
a simpler mechanism with hydride transfer to the
oxygen atom of the C4 carbonyl (green arrows). In
either case, the Ca
2+
would assist by polarizing the C5
carbonyl group.
F. Quinones, Hydroquinones, and Tocopherols
O
O
5
4
PQQ
HO
OH
c
a
CH
3
OH
b
H
2
CO
Reoxidation
O
H

HO O
C
H
H H B
of the nickel-containing F
430
. It also binds two moles of
CoM-SH noncovalently. The binding process requires
ATP as well as the presence of other proteins, notably
those of the hydrogenase system.
Coenzyme M, previously found only in methano-
genic archaeobacteria, has recently been discovered in
both gram-negative and gram-positive alkene-oxidizing
eubacteria. It seems to function in cleavage of epoxy
rings and as a carrier of hydroxyalkyl groups.
441a
See
also Chapter 17.
F. Quinones, Hydroquinones, and Tocopherols
1. Pyrroloquinoline Quinone (PQQ)
Bacteria that oxidize methane or methanol
(methylotrophs) employ a periplasmic methanol dehy-
drogenase that contains as a bound coenzyme, the
pyrroloquinoline quinone designated PQQ or meth-
oxatin (Eq. 15-51).
442 444
This fluorescent ortho-quinone
is released when the protein is denatured. Some other
bacterial alcohol dehydrogenases
445,445a
and glucose
dehydrogenases
446-446d
contain the same cofactor.
PQQ-containing methanol dehydrogenase from
Methylophilus is a dimer of a 640-residue protein con-
sisting of two disulfide-linked peptide chains with
one noncovalently bound PQQ.
447
The large subunit
contains a propellor (Fig. 15-23), which is similar to
that in the protein G
i
shown in Fig. 11-7. The PQQ
binding site lies above the propellor and is formed by
a series of loops. The coenzyme interacts with several
polar protein side chains and with a bound calcium
ion.
448
Bacterial PQQ-dependent glucose dehydrogenases
N
N
H
COOH
HOOC O
O
COOH
1
N
N
H
COOH
HOOC OH
OH
COOH
7
6
9
5
3
2
4
8
PQQ (oxidized)
PQQH
2
(reduced)
(15-51)
816 Chapter 15. Coenzymes of OxidationReduction Reactions
Figure 15-23 (A) Stereoscopic view of the H subunit
of methanol dehydrogenase. Eight four-stranded anti-
parallel sheets, labeled W1W8, form the base of the
subunit. Several helices and two additional -sheet
structures (labeled x and y) form a cap over the base.
The PQQ is located in a funnel within the cap approxi-
mately on an eight-fold axis of pseudosymmetry.
Courtesy of Xia et al.
447
(B) Schematic view of the
active site. W467 is parallel to the plane of PQQ. All
hydrogen-bond interactions between PQQ and its
surrounding atoms, except for the three water mole-
cules, are indicated. Courtesy of White et al.
448
2. Copper Amine Oxidases
At first it appeared that PQQ had a broad distribu-
tion in enzymes, including eukaryotic amine oxidases.
However, it was discovered, after considerable effort,
that there are additional quinone cofactors that func-
tion in oxidation of amines. These are derivatives of
tyrosyl groups of specific enzyme proteins. Together
with enzymes containing bound PQQ they are often
called quinoproteins.
450 454
Topaquinone (TPQ). Both bacteria and eukary-
otes contain amine oxidases that utilize bound copper
ions and O
2
as electron acceptors and form an aldehyde,
NH
3
, and H
2
O
2
. The presence of an organic cofactor
was suggested by the absorption spectra which was
variously attributed to pyridoxal phosphate or PQQ.
However, isolation from the active site of bovine serum
A
B
O
OH
HO
O HO
O
O
Ca
2+
OH
H
H
O
O
5
Ca
2+
OH
HO
4
5
H
4

His 144
H

H
+
(15-52)
817
amine oxidase established the structure of the reduced
form of the cofactor as a trihydroxyphenylalanyl
group covalently linked in the peptide chain.
455,456
Its oxidized form is called topaquinone:
The same cofactor is present in an amine oxidase from
E. coli
457 460
and in other bacterial
461 463
fungal,
463a
plant,
464
and mammalian
464a
amine oxidases.
Lysine tyrosylquinone (LTQ). Another copper
amine oxidase, lysyl oxidase, which oxidizes side
chains of lysine in collagen and elastin (Eq. 8-8) con-
tains a cofactor that has been identified as having a
lysyl group of a different segment of the protein in
place of the OH in the 2 position of topaquinone.
465
Lysyl oxidase plays an essential role in the crosslinking
of collagen and elastin.
Tryptophan tryptophanylquinone (TTQ). This
recently discovered quinone cofactor is similar to the
lysyl tyrosylquinone but is formed from two trypto-
phanyl side chains.
466
It has been found in methyl-
amine dehydrogenase from methylotrophic gram-
negative bacteria
467 469
and also in a bacterial aromatic
amine dehydrogenase.
470
F. Quinones, Hydroquinones, and Tocopherols
Three-dimensional structures. The TPQ-con-
taining amine oxidase from E. coli is a dimer of 727-
residue subunits with one molecule of TPQ at position
402 in each subunit.
457,458
Methylamine dehydrogenase
is also a large dimeric protein of two large 46.7-kDa
subunits and two small 15.5-kDa subunits. Each large
subunit contains a TTQ cofactor group.
468,471,471a
Reduced TTQ is reoxidized by the 12.5-kDa blue copper
protein amicyanin. Crystal structures have been
determined for complexes of methylamine dehydroge-
nase with amicyanin
471
and of these two proteins with
a third protein, a small bacterial cytochrome c.
472,472a
Mechanisms. Studies of model reactions
473 476
and of electronic, Raman,
466,477,478
ESR,
479,480
and NMR
spectra and kinetics
481
have contributed to an under-
standing of these enzymes.
459,461,464,482,483
For these
copper amine oxidases the experimental evidence
suggests an aminotransferase mechanism.
450,453,474,474a-d
The structure of the E.coli oxidase shows that a single
copper ion is bound by three histidine imidazoles and
is located adjacent to the TPQ (Eq. 15-53). Asp 383 is a
conserved residue that may be the catalytic base in Eq.
15-53.
474b
A similar mechanism can be invoked for LTQ
and TTQ.
How is the reduced cofactor reoxidized? Presum-
ably the copper ion adjacent to the TPQ functions in
this process, passing electrons one at a time to the
next carrier in a chain. There is no copper in the TTQ-
containing subunits. Electrons apparently must jump
about 1.6 nm to the copper ion of amicyanin, then
another 2.5 nm to the iron ion of the cytochrome c.
472
Reoxidation of the aminoquinol formed in Eq. 15-53,
step d, yields a Schiff base whose hydrolysis will release
ammonia and regenerate the TTQ. Intermediate states
with Cu
+
and a TTQ semiquinone radical have been
observed.
483a
O
O
OH
CH
2
CH
N C
O
HO
O
O
CH
2
CH
N C
O
H
1
5
4
6
3
2
H
Topaquinone (TPQ)
N
O
O
CH
2
CH
N C
O
C O
HN
H
H
Lysine tyrosylquinone (LTQ)
N
C
O
N O
O
NH
H
2
C
C
N
O
H
H
H
H
H
Tryptophan tryptophylquinone (TTQ)
818 Chapter 15. Coenzymes of OxidationReduction Reactions
3. Ubiquinones, Plastoquinones, Tocopherols,
and Vitamin K
In 1955, R. A. Morton and associates in Liverpool
announced the isolation of a quinone which they
named ubiquinone for its ubiquitous occurrence.
484,485
It was characterized as a derivative of benzoquinone
attached to an unsaturated polyprenyl (isoprenoid)
side chain (Fig. 15-24). In fact, there is a family of
ubiquinones: that from bacteria typically contains six
prenyl units in its side chain, while most ubiquinones
from mammalian mitochondria contain ten. Ubiqui-
none was also isolated by F. L. Crane and associates
using isooctane extraction of mitochondria. These
workers proposed that the new quinone, which they
called coenzyme Q, might participate in electron
transport. As is described in Chapter 18, this function
has been fully established. Both the name ubiquinone
and the abbreviation Q are in general use. A subscript
indicates the number of prenyl units, e.g., Q
10
.
Ubiquinones can be reversibly reduced to the hydro-
quinone forms (Fig. 15-24), providing a basis for their
function in electron transport within mitochondria and
chloroplasts.
486 490
While vitamin E (Box 15-G) and vitamin K (Box
15-F) are dietary essentials for humans, ubiquinone is
apparently not. Animals are able to make ubiquinones
in quantities adequate to meet the need for this essen-
tial component of mitochondria. However, an extra
dietary supplement may sometimes be of value.
491
A
reduced level of ubiquinone has been reported in gum
tissues of patients with periodontal disease.
492
Ubiquinones are being tested as possible protectants
against heart damage caused by lack of adequate
oxygen or by drugs.
491
N
O
O

Cu O
H
H
C
R
H

N
O
O

Cu O
H
H
C
H
R
H
H
B
d
b
c
a
R-CH
2
NH
3
2e

H
2
O
Aminoquinol
H
2
O
N C
R
H
O C
R
H
N
R
H
H
H
O
O
O

Cu O
H
H
C
NH
2
O
O

Cu O
H
H

NH
2
NH
3
TPQ
+
+
+
H
2+ 2+
2+ 2+
(15-53)
A closely related series of
plastoquinones occur in chloro-
plasts (Chapter 23). In these com-
pounds the two methoxyl groups
of ubiquinone are replaced by
methyl groups (Fig. 15-24). The
most abundant of these com-
pounds, plastoquinone A, con-
tains nine prenyl units.
493,494
Addition of the hydroxyl
group of the reduced ubiqui-
none or plastoquinone to the
adjacent double bond leads to
a chroman-6-ol structure. The
compounds derived from ubiqui-
none in this way are called
ubichromanols (Fig. 15-24).
The corresponding ubichromenol
(Fig. 15-24) has been isolated
from human kidney. Plasto-
chromanols are derived from plastoquinone. That
from plastoquinone A, first isolated from tobacco, is
also known as solanochromene.
A closely related and important family of chro-
manols are the tocopherols or vitamins E (Fig. 15-24,
Box 15-G). Tocopherols are plant products found
primarily in plant oils and are essential to proper
nutrition of humans and other animals. -Tocopherol
is the most abundant form of the vitamin E family;
smaller amounts of the , , and forms occur, as do
a series of tocotrienols which contain unsaturated
isoprenoid units.
495
The configuration of -tocopherol
is 2R,4'R,8'R as indicated in Fig. 15-24. When -toco-
pherol is oxidized, e.g., with ferric chloride, the ring can
be opened by hydrolysis to give tocopherolquinones
(Fig. 15-24), which can in turn be reduced to tocopherol-
hydroquinones. Large amounts of the tocopherol-
quinones have been found in chloroplasts.
Another important family of quinones, related in
structure to those already discussed, are the vitamins
K (Fig. 15-24, Box 15-F). These occur naturally as two
families. The vitamins K
1
(phylloquinones) have
only one double bond in the side chain and that is in
the prenyl unit closest to the ring. This suggests again
the possibility of chromanol formation. In the vitamin
K
2
(menaquinone) series, a double bond is present
in each of the prenyl units. A synthetic compound
menadione completely lacks the polyprenyl side chain
and bears a hydrogen in the corresponding position on
the ring. Nevertheless, menadione serves as a synthetic
vitamin K, apparently because it can be converted in
the body to forms containing polyprenyl side chains.
819
OCH
3
H
3
CO
O
CH
3
O
H
CH
3
OCH
3
H
3
CO
HO
CH
3
OH
H
CH
3
OCH
3
H
3
CO
HO
CH
3
O
CH
3
CH
3
H
CH
3
H
3
C
HO
CH
3
O
CH
3
CH
3
H
H
These two methoxyl
groups are replaced
by CH3 groups in
plastoquinones
n
Ubiquinones (coenzyme Q)
n = 610
n
Reduced ubiquinone
+ 2[H]
n1
Ubichromanol
CH
3
H
3
C
O
CH
3
H
O
HO CH
3
CH
3
A double bond is present
here in ubichromenol
3
1
Tocotrienols contain
double bonds here
2
3
4
5
6
7
O
CH
3
H
O
CH
3
3
Tocopherolquinone of chloroplasts
1'
n = 4,5. In the phylloquinone
(vitamin K1) series only one double
bond is present in the innermost unit
of the isoprenoid side chain.
Vitamins K
8
-Tocopherol contains H in position 7
-Tocopherol contains H in position 5
-Tocopherol contains H in positions 5 and 7
n
-Tocopherol (vitamin E)
Figure 15-24 Structures of the isoprenoid quinones and vitamin E.
these membranes. Within some my-
cobacteria vitamin K apparently
participates in electron transport
chains in the same way (see Chapter
18). Some bacteria contain both
menaquinones and ubiquinones.
The vitamin E derivative
-tocopherolquinone (Fig. 15-24)
can also serve as an electron carrier,
being reversibly reduced to the hydro-
quinone form -tocopherolquinol.
Such a function has been proposed
for the anaerobic rumen bacterium
Butyrovibrio fibrisolvens.
497
When a single hydrogen atom is
removed from a hydroquinone or from
a chromanol such as a tocopherol, a
free radical is formed (Eq. 15-54).
Phenols substituted in the 2, 4, and
6 positions give especially stable
radicals.
Both the presence of methyl substitu-
ents in the tocopherols and their
chromanol structures increase the
ability of these compounds to form
relatively stable radicals.
498,499
This
ability is doubtless probably impor-
tant also in the function of ubiquino-
nes and plastoquinones. Ubiquinone
radicals (semiquinones) are probably
intermediates in mitochondrial elec-
tron transport (Chapter 18) and radi-
cals amounting to as much as 40% of
the total ubiquinone in the NADH-
ubiquinone reductase of heart mito-
chondria have been detected by EPR
measurements.
500,501
The equilibria governing semi-
quinone formation from quinones are similar to those
for the flavin semiquinones which were discussed in
Section B,6. Two consecutive one-electron redox steps
can be defined. Their redox potentials will vary with
pH because of a pK
a
for the semiquinone in the pH 4.5
6.5 region. For ubiquinone this pK
a
is about 4.9 in
water and 6.45 in methanol. A pK
a
of over 13 in the
4. Quinones as Electron Carriers
Ubiquinones function as electron transport agents
within the inner mitochondrial membranes
496
and also
within the reaction centers of the photosynthetic mem-
branes of bacteria (Eq. 23-32).
484,488,494
The plasto-
quinones also function in electron transport within
OH
OH(R)
[H]
O
OH (R)
2
4
6
(15-54)
F. Quinones, Hydroquinones, and Tocopherols
820 Chapter 15. Coenzymes of OxidationReduction Reactions
hydroquinone form
502
will have little effect on redox
potentials near pH 7. The potential for the one-elec-
tron reaction Q + e

is evaluated most readily.


For this reaction E (pH 7) is 0.074, 0.13, 0.17, and
0.23 for 2,3-dimethylbenzoquinone, plastoquinone,
ubiquinone, and phylloquinone, respectively.
Why does this entire family of compounds have
the long polyprenyl side chains? A simple answer is
that they serve to anchor the compounds in the lipid
portion of the cell membranes where they function.
In the case of ubiquinones both the oxidized and the
reduced forms may move freely through the lipid
phase shuttling electrons between carriers.
5. Vitamin K and -Carboxyglutamate
Formation
In higher animals the only known function of
vitamin K is in the synthesis of -carboxyglutamate
(Gla)-containing proteins, several of which are needed
in blood clotting (Box 15-F, Chapter 12). Following the
discovery of -carboxyglutamate, it was shown that liver
microsomes were able to incorporate
14
C-containing
bicarbonate or CO
2
into the Gla of prothrombin and
could also generate Gla in certain simple peptides such
as Phe-Leu-Glu-Val. Three enzymes are required.
All are probably bound to the microsomal mem-
branes.
503 507a
An NADPH-dependent reductase reduces
vitamin K quinone to its hydroquinone form. Conver-
sion of Glu residues to Gla residues requires this reduced
vitamin K as well as O
2
and CO
2
. During the carboxy-
lation reaction the reduced vitamin K is converted into
vitamin K 2,3-epoxide (Eq. 15-55).
508
The mechanism is
uncertain but a peroxide intermediate such as that
shown in Eq. 15-56 is probably involved. This could
be used to generate a hydroxide ion adjacent to the
pro-S -H of the glutamate side chain of the substrate.
This hydrogen could be abstracted by the OH

to form
H
2
O and a carbanion which would be stabilized by the
adjacent carboxyl group
508 511
(Eq. 15-56).
Dowd and coworkers raised doubts that a hydroxide
ion released in the active site in this manner is a strong
enough base to generate the anion shown in Eq. 15-
52.
512,513
They hypothesized a base strength amplifica-
tion mechanism that begins with a peroxide formed at
C-4 followed by ring closure to form a dioxetane and
rearrangement to the following hypothetical strong
base.
507,512,514
The proposal was supported by model
experiments and also by observation
of some incorporation of both atoms
of
18
O
2
into vitamin K epoxide. How-
ever, theoretical calculations support
the simpler mechanism of Eq. 15-56.
The fact that L-threo--fluoro-
glutamate residues, in which the
fluorine atom is in the position corre-
sponding to the pro-S hydrogen, are
not carboxylated but that an erythro-
-fluoroglutamate is carboxylated
OH
OH
CH
3
Phytyl
Glu Gla
CO
2
O
2
H
2
O
Dihydro
vitamin K
Vitamin K-
2R,3S-epoxide
N
N
O
O
COO

H
R
H
H

OOC
O
O
CH
3
O
Phytyl
1
2
3
4
5
6
N
N
O
O
COO

H
S
H
H

H
R
(15-55)
O
O
R
CH
3
O O
H
C
H
S
H
COOH
H
Glu side chain
in protein
Peroxide
1

C
H
COOH
3
4
2
Dihydrovitamin K
O
C
O
O
2
Epoxide H
2
O
(15-56)
O
O
CH
3
Phytyl
O HO

Hypothetical strong base


821
BOX 15-F THE VITAMIN K FAMILY
The existence of an antihemorrhagic factor
required in the diet of chicks to ensure rapid clotting
of blood was reported in 1929 by Henrik Dam at the
University of Copenhagen.
a,b
The fat-soluble material,
later designated vitamin K, causes a prompt (2 6 h)
decrease in the clotting time when administered to
deficient animals and birds. The clotting time for a
vitamin K-deficient chick may be greater than 240 s,
but 6 h after injection of 2 g of vitamin K
3
it falls to
50100 s.
c
Pure vitamin K (Fig. 15-24), a 1,4-naphtho-
quinone, was isolated from alfalfa in 1939. Within a
short time two series, the phylloquinones (vitamin K
1
)
and the menaquinones (vitamin K
2
), were recognized.
The most prominant phylloquinone contains the phytyl
group, which is also present in the chlorophylls. For
a human being a dietary intake of about 30 g per day
is recommended.
d
Additional vitamin K is normally
supplied by intestinal bacteria.
The most obvious effect of a deficiency in vitamin
K in animals is delayed blood clotting, which has been
traced to a decrease in the activity of prothrombin and
of clotting factors VII, IX, and X (Chapter 12, Fig. 12-17).
Prothrombin formed by the liver in the absence of vitamin
K lacks the ability to chelate calcium ions essential for
the binding of prothrombin to phospholipids and to
its activation to thrombin. The structural differences
between this abnormal protein and the normal pro-
thrombin have been pinpointed at the N terminus of
the ~ 560 residue glycoprotein.
e,f
Tryptic peptides from
the N termini differed in electrophoretic mobility. As
detailed in Chapter 12, ten residues within the first 33,
which were identified as glutamate residues by the
sequence analysis on normal prothrombin, are actually
-carboxyglutamate (Gla). The same amino acid is
present near the N termini of clotting factors VII, IX,
and X.
The fact that -carboxyglutamate had not been
identified previously as a protein substituent is ex-
plained by its easy decarboxylation to glutamic acid
during treatment with strong acid. The function of
vitamin K is to assist in the incorporation of the addi-
tional carboxyl group into the glutamate residues of
preformed prothrombin and other blood-clotting
factors
g,h
with a resulting increase in calcium ion
affinity.
Four other plasma proteins designated C, S, M,
and Z contain -carboxyglutamate. The functions of
proteins M and Z are unknown but protein C is a serine
protease involved in regulation of blood coagulation
and protein S is a cofactor that assists the action of
protein C. Other proteins that require vitamin K for
synthesis include the 49-residue bone Gla protein (or
osteocalcin) and the 79-residue matrix Gla protein
found in bone and cartilage.
i,j
These proteins contain
three and five residues of Gla, respectively. Their
possible functions in mineralization are considered
in Chapter 8. At least two additional small human
proline-rich Gla proteins of unknown function are
synthesized in many tissues.
k
Gamma-carboxy-
glutamate also occurs in an invertebrate peptide from
fish-hunting cone snails. This sleeper peptide, which
induces sleep in mice after intracerebral injection, has
the sequence GEE*E*LQE*NQE*LIRE*KSN. Here E*
designates the 5 residues of Gla.
l
An interesting facet of vitamin K nutrition and
metabolism was revealed by the observation that cattle
fed on spoiled sweet clover develop a fatal hemorrhagic
disease. The causative agent is dicoumarol, a com-
pound arising from coumarin, a natural constituent of
clover. Dicourmarol and the closely related synthetic
warfarin are both potent vitamin K antagonists.
Warfarin is used both as a rat poison and in the treat-
ment of thromboembolic disease. As rodenticides
hydroxycourmarin derivatives are usually safe because
a single accidental ingestion by a child or pet does little
harm, whereas regular ingestion by rodents is fatal.
a
Wasserman, R. H. (1972) Ann. Rev. Biochem. 41, 179202
b
Tim Kim, X. (1979) Trends Biochem. Sci. 4, 118119
c
Olson, R. E. (1964) Science 45, 926928
d
Shils, M. E., Olson, J. A., and Shike, M., eds. (1994) Modern
Nutrition in Health and Disease, 8th ed., Vol. 1, Lea & Febiger,
Philadelphia, Pennsylvania (pp. 353355)
e
Friedman, P. A. (1984) N. Engl. J. Med. 310, 14581460
f
Stenflo, J. (1976) J. Biol. Chem. 251, 355363
g
Wood, G. M., and Suttie, J. W. (1988) J. Biol. Chem. 263, 32343239
h
Wu, S.-M., Mutucumarana, V. P., Geromanos, S., and Stafford,
D. W. (1997) J. Biol. Chem. 272, 1171811722
i
Price, P. A., and Williamson, M. K. (1985) J. Biol. Chem. 260,
1497114975
j
Price, P. A., Rice, J. S., and Williamson, M. K. (1994) Protein Sci.
3, 822830
k
Kulman, J. D., Harris, J. E., Haldeman, B. A., Davie, E. W.
(1997) Proc. Natl. Acad. Sci., USA 94, 90589062
l
Prorok, M., Warder, S. E., Blandl, T., and Castellino, F. J. (1996)
Biochemistry 35, 1652816534
H
3
+
N C
H
2
C
COO
H
C
COO

COO

H
-Carboxyglutamate (Gla)
O
CH
2
O
OH
O O
OH
Dicoumarol
F. Quinones, Hydroquinones, and Tocopherols
822 Chapter 15. Coenzymes of OxidationReduction Reactions
suggested the indicated stereospecificity.
515,516
This
was confirmed by observation of a kinetic isotope
effect when
2
H is present in the pro-S position.
517
Addition of the carbanion to CO
2
would generate the
Gla residue. The two glutamates in the following
sequence, in which X may be various amino acids,
are carboxylated if the protein also carries a suitable
N-terminal signal sequence: EXXXEXC. If a suitable
glutamyl peptide is not available for carboxylation the
postulated peroxide intermediate (Eq. 15-55) is still
converted slowly to the 2,3-epoxide of vitamin K.
A third enzyme is required to reduce the epoxide
to vitamin K (Eq. 15-57). The biological reductant is
uncertain but dithiols such as dithiothreitol serve in
the laboratory.
518
See also Eq. 18-47. Protonation of an
intermediate enolate anion would give 3-hydroxy-2,
3-dihydrovitamin K, an observed side reaction product.
Vitamin K epoxide + 2[H] H
2
O + Vitamin K
This reaction is of interest because of its specific inhibi-
tion by such coumarin derivatives as Warfarin:
This synthetic compound, as well as natural cou-
marin anticoagulants (Box 15-F), inhibits both the
vitamin K reductase and the epoxide reductase.
518,519
The matter is of considerable practical importance
because of the spread of warfarin-resistant rats in
Europe and the United States. One resistance muta-
tion has altered the vitamin K epoxide reductase so
that it is much less susceptible to inhibition by war-
farin.
519,520
While glutamate residues in peptides of appropriate
sequence are carboxylated by the vitamin K-depen-
dent system, aspartate peptides scarcely react.
503
Beta-carboxyaspartate is present in protein C of the
blood anticoagulant system (Fig. 12-17)
521
and in
various other proteins containing EGF homology
domains (Table 7-3),
522
but the mechanism of its
formation is unknown.
6. Tocopherols (Vitamin E) as Antioxidants
The major function of the tocopherols is thought to
be the protection of phospholipids of cell membranes
against oxidative attack by free radicals and organic
peroxides. Peroxidation of lipids, which is described
in Chapter 21, can lead to rapid development of ran-
cidity in fats and oils. However, the presence of a small
amount of tocopherol inhibits this decomposition,
presumably by trapping the intermediate radicals in
the form of the more stable tocopherol radicals (Eq.
15-54) which may dimerize or react with other radicals
to terminate the chain. Even though only one mole-
cule of tocopherol is present for a thousand molecules
of phospholipid, it is enough to protect membranes.
523
That vitamin E does function in this way is supported
by the observation that much of the tocopherol require-
ment of some species can be replaced by N,N-diphenyl-
p-phenylenediamine, a synthetic antioxidant (see Table
18-5 for the structure of a related substance). Three
generations of rats have been raised on a tocopherol-
free diet containing this synthetic antioxidant. How-
ever, not all of the deficiency symptoms are prevented.
The antioxidant role of -tocopherol in membranes
is generally accepted.
524 526
It is thought to be critical
to defense against oxidative injury and to help the
body combat the development of tumors and to slow
aging. Gamma-tocopherol may be more reactive than
-tocopherol in removing radicals created by NO and
other nitrogen oxides.
526
Its actions are strongly linked
to those of ascorbic acid (Box 18-D) and selenium. Ascor-
bate may reduce tocopherol semiquinone radicals, while
selenium acts to enhance breakdown of peroxides as
described in the next section.
G. Selenium-Containing Enzymes
In 1957, Schwartz and associates showed that the
toxic element selenium was also a nutritional factor
essential for prevention of the death of liver cells in
rats.
527
Liver necrosis would be prevented by as little
as 0.1 ppm of selenium in the diet. Similar amounts of
selenium were shown to prevent a muscular dystrophy
called white muscle disease in cattle and sheep
grazing on selenium-deficient soil. Sodium selenite
and other inorganic selenium compounds were more
effective than organic compounds in which Se had
replaced sulfur. Keshan disease, an often fatal
heart condition that is prevalent among childen in
Se-deficient regions of China, can be prevented by
supplementation of the diet with NaSeO
3
.
528
Even
the little crustacean water flea Daphnia needs 0.1
part per billion of Se in its water.
529
Selenium has long been known to enhance the
antioxidant activity of vitamin E. Recent work suggests
that vitamin E acts as a radical scavenger, preventing
(15-57)
O
CH
O
HO CH
2
C O
CH
3
Warfarin
823
form with a preference for organic peroxides derived
from phospholipids.
327,538 540
A related selenoprotein
has been found in a human poxvirus.
540a
Selenocys-
teine is present at a position (residues 41 47) near the
N terminus of an helix, in the ~ 180-residue polypep-
tides. A possible reaction mechanism involves attack
by the selenol on the peroxide to give a selenic acid
intermediate which is reduced by glutathione in two
nucleophilic displacement steps (Eq. 15-59).
excessive peroxidation of membrane lipids, while
selenium, in the enzyme glutathione peroxidase,
acts to destroy the small amounts of peroxides that
do form. This was the first established function of
selenium in human beings, but there are others. If we
include proteins from animals and bacteria, at least ten
selenoproteins are known (Table 15-4).
530 534
Seven of
these are enzymes and most catalyze redox processes.
The active sites most often contain selenocysteine,
whose selenol side chain is more acidic (pK
a
~ 5.2) than
that of cysteine and exists as CH
2
Se

at neutral pH.
530
Glutathione peroxidases catalyze the reductive
decomposition of H
2
O
2
or of organic peroxides by
glutathione (G SH) according to Eq. 15-58. At least
three isoenzyme forms have been identified in mam-
mals: a cellular form,
531,535 537
a plasma form, and a
G. Selenium-Containing Enzymes
BOX 15-G VITAMIN E: THE TOCOPHEROLS
Vitamin E was recognized in 1926 as a factor
preventing sterility in rats that had been fed rancid
lipids.
a e
The curative factor, present in high con-
centration in wheat germ and lettuce seed oils, is
a family of vitamin E compounds, the tocopherols
(Fig. 15-24). The first of these was isolated by Evans
and associates in 1936. Vitamin E deficiency in the
rabbit or rat is accompanied by muscular degenera-
tion (nutritional muscular dystrophy; see also
Box 15-A) and a variety of other symptoms that
vary from one species to another. Animals deficient
in vitamin E display obvious physical deterioration
followed by sudden death. Muscles of deficient rats
show abnormally high rates of oxygen uptake, and
abnormalities appear in the membranes of the en-
doplasmic reticulum as viewed with the electron
microscope. It is thought that deterioration of lyso-
somal membranes may be the immediate cause of
death.
The tocopherol requirement of humans is not
known with certainty, but about 5 mg (7.5 IU) / day
plus an additional 0.6 mg for each gram of polyun-
saturated fatty acid consumed may be adequate.
It is estimated that the average daily intake is about
14 mg, but the increasing use of highly refined foods
may lead to dangerously low consumption. Recent
interest
d f
has been aroused by studies that show
that much larger amounts of vitamin E (e.g., 100
400 mg/day) substantially reduce the risk of coro-
nary disease and stroke in both women
g
and men
h
and also decrease oxidative modification of brain
proteins.
i
The decrease in heart attacks and stroke
may be in part an indirect effect of the anticlotting
a
Sebrell, W. H., Jr., and Harris, R. S., eds. (1972) The Vitamins,
Vol. 5, Academic Press, New York
b
DeLuca, H. F., and Suttie, J. W., eds. (1970) The Fat-Soluble
Vitamins, Univ. of Wisconsin Press, Madison, Wisconsin
c
Machlin, L. J., ed. (1980) Vitamin E, Dekker, New York
d
Mino, M., Nakamura, H., Diplock, A. T., and Kayden, H. J.,
eds. (1993) Vitamin E: Its Usefulness in Health and in Curing
Diseases, Japan Scientific Socities Press, Tokyo
e
Packer, L., and Fuchs, J., eds. (1993) Vitamin E in Health and
Disease, Dekker, New York
f
Dowd, P., and Zhend, Z. B. (1995) Proc. Natl. Acad. Sci. U.S.A.
92, 81718175
g
Stampfer, M. J., Hennekens, C. H., Manson, J. E., Colditz, G. A.,
Rosner, B., and Willett, W. C. (1993) N. Engl. J. Med. 328, 1444
1449
h
Rimm, E. B., Stampfer, M. J., Ascherio, A., Giovannucci, E.,
Colditz, G. A., and Willett, W. C. (1993) N. Engl. J. Med. 328,
14501456
i
Poulin, J. E., Cover, C., Gustafson, M. R., and Kay, M. M. B.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 56005603
j
Diplock, A. T., and Lucy, J. A. (1973) FEBS Lett. 29, 205210
action of vitamin E quinone.
f
Plant oils are usually
the richest sources of tocopherols, while animal
products contain lower quantities.
To some extent the vitamin E requirement may
be lessened by the presence in the diet of synthetic
antioxidants and by selenium. Much evidence
supports a relationship between the nutritional
need for selenium and that for vitamin E. Lack of
either causes muscular dystrophy in many animals
as well as severe edema (exudative diathesis) in
chicks. Since vitamin E-deficient rats have a low
selenide (Se
2
) content, it has been suggested that
vitamin E protects reduced selenium from oxida-
tion.
j
Vitamin C (ascorbic acid), in turn, protects
vitamin E.
2 GHSH
Glutathione
GSSG
Oxidized glutathione
H
2
O
2
or R-OOH
2H
2
O or H
2
O + ROH
Glutathione peroxidase
(15-58)
824 Chapter 15. Coenzymes of OxidationReduction Reactions
Three types of iodothyronine deiodinase remove
iodine atoms from thyroxine to form the active thyroid
hormone triiodothyronine and also to inactivate the
hormone by removing additional iodine
531,541 546
(see
also Chapter 25). In this case the CH
2
Se

may attach
the iodine atom, removing it as I
+
to form CH
2
Se I.
The process could be assisted by the phenolic OH
group if it were first tautomerized (Eq. 15-60).
C
O
H
3
C E
B
C
NH
2
+
H
3
C E
B
E
A
SH
Se CH
2
C
O
OH
S

E
C
E
A
S

Se CH
2
C
O
S
E
C
NH
C
CH
2
H
3
C E
B
COO

+
H
3
C C
O
S E
C
H
3
C C
O
OPO
3
2
S
Se
E
A
SH
Se

E
A
NH
4
+
H
2
O
H
+
+ 2e

Acetate
Glycine
H
2
O
b
a
c
f
d
e
g
P
i

S E
C
h
ADP ATP
i
(15-61)
CH
2
Se E I
O CH
2
O
H
I
C
H
N H
O C
I
I
I
H
+
O O
H
I
E CH
2
Se
I
I
H
CH
2
C
H
N H
C C

+
I
Triiodothyronine
(15-60)
OH
O
R H
+
E CH
2
Se
H OR (or H
2
O)
E CH
2
Se OH
H
G S
E CH
2
Se S
G S
G
G S S G
Reduced glutathione
+

H
2
O
(or H
2
O
2
)
(15-59)
A recently discovered human selenoprotein is a
thioredoxin reductase which is present in the T cells
of the immune system as well as in placenta and other
tissues.
189,547 549
The 55-kDa protein has one seleno-
cysteine as the penultimate C-terminal residue. Another
mammalian selenoprotein, of uncertain function, is the
57-kDa selenoprotein P. It contains over 60% of the
selenium in rat plasma and is also present in the human
body. Selenoprotein P contains ten selenocysteine
residues.
550 552a
Some of these may be replaced by
serine in a fraction of the molecules.
553
A smaller
9.6-kDa skeletal muscle protein, selenoprotein W,
contains a single selenocysteine.
554 556
Another seleno-
protein has been found in sperm cells, both in the tail
and in a keratin-rich capsule that surrounds the mito-
chondria in the sperm midpiece.
557
Lack of this protein
may be the cause of the abnormal immotile spermatozoa
observed in Se-deficient rats and of reproductive diffi-
culties among farm animals in Se-deficient regions.
558
Several selenoproteins have been found in certain
bacteria and archaea. A hydrogenase from Methano-
coccus vannielii contains selenocysteine.
559,560
This
enzyme transfers electrons from H
2
to the C-5 si face of
the 8-hydroxy-5-deazaflavin cofactor F
420
(Section B,4).
The same bacterium synthesizes two formate
dehydrogenases (see Fig 15-23), one of which con-
tains Se. Two Se-containing formate dehydrogenases
are made by E. coli. One of them, which is coupled to
a hydrogenase in the formate hydrogen-lyase system
(see Eq. 15-37), is a 715-residue protein containing
selenocysteine at position 140.
561 563
The second has
selenocysteine at position 196 and functions with a
nitrate reductase in anaerobic nitrate respiration.
561
Glycine reductase is a complex enzyme
530,564 566
that catalyzes the reductive cleavage of glycine to
acetyl phosphate and ammonia (Eq. 15-61) with the
825
subsequent synthesis of ATP (Eq. 14-43). Electrons for
reduction of the disulfide that is formed are provided
by NADH. A single selenocysteine residue is present
in the small 12-kDa subunits. The enzyme contains a
dehydroalanine residue (Chapter 14) in subunit B and
a thiol group in subunit C.
566
An acetyl-enzyme deriv-
ative of subunit C, perhaps of its SH group, has been
identified.
546
The mechanism of action is uncertain
but the steps in Eq. 15-61 have been suggested.
567
The subunits are designated E
A
, E
B
, and E
C
. Step e is
particularly hard to understand because formation of
a thioester in this manner is not expected to occur
spontaneously and must be linked in some way to
other steps.
A selenium-containing xanthine dehydrogenase
is present in purine-fermenting clostridia. Like other
xanthine dehydrogenases (Chapter 16), it converts
xanthine to uric acid and contains nonheme iron,
molybdenum, FAD, and an FeS center. The selenium
is probably present as Se
2
bound to Mo as is S
2
in xan-
thine oxidase (Fig. 16-32).
567a
A related reaction is cata-
lyzed by the FeS protein nicotinic acid hydroxylase
(Eq. 15-62) found in some clostridia.
568
Splitting of the
Mo(V) EPR signal when
77
Se is present in the enzyme
shows that the selenium is present as a ligand of molyb-
denum. Another member of the family is a purine
hydroxylase that converts purine 2-hydroxypurine,
or hypoxanthine to xanthine.
567a
TABLE 15-4
Selenium-Containing Proteins
Enzyme Source
Mass
(kDa)
Subunit
composition
Other
cofactors
Glutathione peroxidases None
Cellular
Plasma
Phospholipid
hydroperoxide
Mammals
Humans
Pig, rat
21 x 4
18
Thioredoxin reductase Humans 55 2
Selenoprotein P Mammals 57 None
Selenoprotein W Rat 19.6
Formate dehydrogenase Bacteria - E. coli
Archaea
Heme b
Mo; molybdopterin
600
2

2-4
Hydrogenase Bacteria
Archaea
Some clostridia Glycine reductase
Selenoprotein A
Selenoprotein B
Carbonyl protein C
selenomethionine) Clostridium kluyverii
Thiolase (contains
12
200
250
39 4 None
Fe?
FAD; NiFeSe
2

2
ABC
Iodothyronine deiodinases Vertebrates
Nicotinic acid hydroxylase Clostridia 300 FAD, FeS, Mo
Purine hydroxylase Clostridia FAD, FeS, Mo
dehydrogenase
Oligotropha
carboxidovorans
Carbon monoxide 137 2 FAD; Mo;
molybdopterin

2
G. Selenium-Containing Enzymes
826 Chapter 15. Coenzymes of OxidationReduction Reactions
BOX 15-H GLUTATHIONE PEROXIDASE AND ABNORMALITIES OF RED BLOOD CELLS
The processes by which hemoglobin is kept in
the Fe(II) state and functioning normally within intact
erythrocytes is vital to our health. Numerous hereditary
defects leading to a tendency toward anemia have
helped to unravel the biochemistry indicated in the
accompanying scheme.
a
About 90% of the glucose utilized by erythrocytes
is converted by glycolysis to lactate, but about 10% is
oxidized (via glucose 6-phosphate) to 6-phosphoglu-
conate. The oxidation (reaction a) is catalyzed by glu-
cose-6-phosphate dehydrogenase (Eq.15-10) using
NADP
+
. This is the principal reaction providing the
red cell with NADPH for reduction of glutathione (Box
11-B) according to reaction b. Despite the important
function of glucose-6-P dehydrogenase, ~ 400 million
persons, principally in tropical and Mediterranean
areas, have a hereditary deficiency of this enzyme.
The genetic variations are numerous, with about 400
different ones having been identified. Although most
individuals with this dificiency have no symptoms, the
lack of the enzyme is truly detrimental and sometimes
leads to excessive destruction of red cells and anemia
during some sicknesses and in response to administra-
tion of various drugs.
b
The survival of the defective
genes, like that for sickle cell hemoglobin (Box 7-B) is
thought to result from increased resistance to malaria
parasites.
Other erythrocyte defects that lead to drug sensi-
tivity include a deficiency of glutathione (resulting
from a decrease in its synthesis) and a deficiency of
glutathione reductase (reaction b). The effects of drugs
have been traced to the production of
H
2
O
2
(reaction c) in red blood cells;
catalase, which converts H
2
O
2
into
H
2
O and O
2
, is thought to function
in a similar way. Both enzymes are
probably necessary for optimal health.
An excess of H
2
O
2
can damage
erythrocytes in two ways. It can
cause excessive oxidation of func-
tioning hemoglobin to the Fe(III)-
containing methemoglobin. (Methe-
moglobin is also formed spontane-
ously during the course of the oxygen-
carrying function of hemoglobin. It
is estimated that normally as much
as 3% of the hemoglobin may be
oxidized to methemoglobin daily.)
The methemoglobin formed is reduced
back to hemoglobin through the
action of NADH-methemoglobin
reductase (reaction f). A smaller
fraction of the methemoglobin is
reduced by a similar enzyme requir-
ing NADPH (as indicated by the
colored arrow). A hereditary lack of
the NADH-methemoglobin reductase
is also known.
A second destructive function
of H
2
O
2
is attack on double bonds of
unsaturated fatty acids of the phos-
pholipids in cell membranes. The
resulting fatty acid hydroperoxides
can react further with C C chain cleavage and disrup-
tion of the membrane. This is thought to be the principal
cause of the hemolytic anemia induced by drugs in
susceptible individuals. The selenium-containing gluta-
thione peroxidase is thought to decompose these fatty
acid hydroperoxides. Vitamin E (Box 15-G), acting as
an antioxidant within membranes, is also needed for
good health of erythrocytes.
c,d
a
Chanarin, I. (1970) in Biochemical Disorders in Human Disease,
3rd ed. (Thompson, R. H. S., and Wooton, I. D. P., eds), pp. 163
173, Academic Press, New York
b
Luzzatto, L., and Mehta, A. (1995) in The Metabolic and Molecular
Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 3367 3398, McGraw-
Hill, New York
c
Constantinescu, A., Han, D., and Packer, L. (1993) J. Biol. Chem.
268, 10906 10913
d
Liebler, D. C., and Burr, J. A. (1992) Biochemistry 31, 8278 8284
a b c
d
e
f
g
Autoxidizable
drugs, AH
2
O
2
A
NADP
+
NADPH 6-Phosphogluconate
2 GSH G 6 P
G S S G
H
2
O
2
2 H
2
O
NAD
+
NADH
Hemoglobin(Fe
2+
)
Methemoglobin(Fe
3+
) HO
2
, H
2
O
Nonenzymatic
O
2
O
2

Unsaturated fatty acids


in phospholipids
Fatty acid
hydroperoxides
OOH
827
(15-63)
Athiolase (Eq. 13-35) from Clostridium kluyveri
is one of only two known selenoproteins that contain
selenomethionine.
569
However, the selenomethionine
is incorporated randomly in place of methionine. This
occurs in all proteins of all organisms to some extent
and the toxicity of selenium may result in part from
excessive incorporation of selenomethionine into
various proteins.
Selenium is found to a minor extent wherever
sulfur exists in nature. This includes the sulfur-con-
taining modified bases of tRNA molecules. In addition
to a small amount of nonspecific incorporation of Se
into all S-containing bases there are, at least in bacteria,
specific Se-containing tRNAs. In E. coli one of these is
specific for lysine and one for glutamate. One of the
modified bases has been identified as 5-methyl-amino-
methyl-2-selenouridine.
570
It is present at the first
position of the anticodon, the wobble position.
571
Selenium has its own metabolism. Through the
use of
75
Se as a tracer, normal rat liver has been shown
to contain Se
2
, SeO
3
2
, and selenium in a higher oxida-
tion state.
572
Glutathione may be involved in reduction
of selenite to selenide.
573
The nonenzymatic reduction
of selenite by glutathione yields a selenotrisulfide
derivative (Eq. 15-63). The latter is spontaneously
decomposed to oxidized glutathione and elemental
selenium or by the action of glutathione reductase to
glutathione and selenium. Selenocysteine can be
converted to alanine + elemental selenium (Eq. 14-34).
Some bacteria are able to oxidize elemental Se back to
selenite.
574
Selenium undergoes biological methylation
readily in bacteria, fungi, plants, and animals (Chapter
16).
575,576
This may in some way be related to the
reported effect of selenium in protecting animals
against the toxicity of mercury. Excess selenium may
appear in the urine as trimethylselenonium ions.
577
How is selenium incorporated into selenocysteine-
containing proteins? This element does enter amino
acids to a limited extent via the standard synthetic
pathways for cysteine and methionine. However, the
placement of selenocysteine into specific positions in
selenoproteins occurs by the use of a minor serine-
specific tRNA that acts as a suppressor of chain termi-
nation during protein synthesis.
532,533,578
(This topic is
dealt with further in Chapter 29.) The genes for these
and presumably for other selenocysteine-containing
proteins have the stop codon TGA at the selenocys-
teine positions. However, when present in a suitable
context the minor tRNA, carrying selenocysteine in
place of serine, is utilized to place selenocysteine into
the growing peptide chain. In bacteria, and presum-
ably also in eukaryotes, the selenocysteinyl-tRNA is
formed from the corresponding seryl-tRNA by a PLP-
catalyzed -replacement reaction. The selenium donor
is not Se
2
but selenophosphate Se PO
3
2
in which the
Se P bond is quite weak.
579 581
After addition to the
aminoacrylate intermediate in the PLP enzyme the
Se P bond may be hydrolytically cleaved to HPO
4
2
and selenocysteyl-tRNA.
N
COOH
N
COOH NADP
+
NADPH
O
H
2
O
H
H
+
(15-62)
4 GSH SeO
3
2
pH 7
2 H
+
3 H
2
O
(Selenotrisulfide)
Spontaneous
Glutathione reductase
Se
0
G S S G
G S S G
S Se S G G
S Se H G
Se SH G
G. Selenium-Containing Enzymes
828 Chapter 15. Coenzymes of OxidationReduction Reactions
54. Dunn, M. F., Dietrich, H., MacGibbon, A. K.
H., Koerber, S. C., and Zeppezauer, M. (1982)
Biochemistry 21, 354363
55. Jagodzinski, P. W., and Peticolas, W. L. (1981)
J. Am. Chem. Soc. 103, 234236
56. MacGibbon, A. K. H., Koerber, S. C., Pease,
K., and Dunn, M. F. (1987) Biochemistry 26,
30583067
57. Hennecke, M., and Plapp, B. V. (1983) Biochemis-
try 22, 37213728
58. Plapp, B. V. (1995) Methods Enzymol. 249, 91
119
59. Makinen, M. W., Maret, W., and Yim, M. B.
(1983) Proc. Natl. Acad. Sci. U.S.A. 80, 2584
2588
60. Ryde, U. (1995) Protein Sci. 4, 11241132
61. Ehrig, T., Muhoberac, B. B., Brems, D., and
Bosron, W. F. (1993) J. Biol. Chem. 268, 11721
11726
62. Hurley, T. D., Bosron, W. F., Stone, C. L., and
Amzel, L. M. (1994) J. Mol. Biol. 239, 415429
63. Charlier, H. A., Jr., and Plapp, B. V. (2000)
J. Biol. Chem. 275, 1156911575
64. Xie, P., Parsons, S. H., Speckhard, D. C.,
Bosron, W. F., and Hurley, T. D. (1997) J. Biol.
Chem. 272, 1855818563
65. Cho, H., Ramaswamy, S., and Plapp, B. V.
(1997) Biochemistry 36, 382389
66. Ganzhorn, A. J., Green, D. W., Hershey, A. D.,
Gould, R. M., and Plapp, B. V. (1987) J. Biol.
Chem. 262, 37543761
67. Hake, S., Kelley, P. M., Taylor, W. C., and
Freeling, M. (1985) J. Biol. Chem. 260, 5050
5054
67a. Korkhin, Y., Kalb-Gilboa, A. J., Peretz, M.,
Bogin, O., Burstein, Y., and Frolow, F. (1998) J.
Mol. Biol. 278, 967981
68. Jrnvall, H., Persson, B., Krook, M., Atrian, S.,
Gonzlez-Duarte, R., Jeffery, J., and Ghosh, D.
(1995) Biochemistry 34, 60036013
69. Ribas de Pouplana, L., and Fothergill-Gilmore,
L. A. (1994) Biochemistry 33, 70477055
69a. Benach, J., Atrian, S., Gonzlez-Duarte, R.,
and Ladenstein, R. (1999) J. Mol. Biol. 289,
335355
70. Hawes, J. W., Crabb, D. W., Chan, R. M.,
Rougraff, P. M., and Harris, R. A. (1995)
Biochemistry 34, 42314237
71. Pawlowski, J. E., and Penning, T. M. (1994) J.
Biol. Chem. 269, 1350213510
72. Wilson, D. K., Nakano, T., Petrash, J. M., and
Quiocho, F. A. (1995) Biochemistry 34, 14323
14330
73. Hoog, S. S., Pawlowski, J. E., Alzari, P. M.,
Penning, T. M., and Lewis, M. (1994) Proc.
Natl. Acad. Sci. U.S.A. 91, 25172521
74. Wilson, D. K., Bohren, K. M., Gabbay, K. H.,
and Quiocho, F. A. (1992) Science 257, 8184
75. Rondeau, J.-M., Tte-Favier, F., Podjarny, A.,
Reymann, J.-M., Barth, P., Biellmann, J.-F., and
Moras, D. (1992) Nature (London) 355, 469471
76. Tarle, I., Borhani, D. W., Wilson, D. K.,
Quiocho, F. A., and Petrash, J. M. (1993) J. Biol.
Chem. 268, 2568725693
77. Grimshaw, C. E., Bohren, K. M., Lai, C.-J., and
Gabbay, K. H. (1995) Biochemistry 34, 14374
14384
78. Grimshaw, C. E. (1992) Biochemistry 31,
1013910145
79. Harrison, D. H., Bohren, K. M., Ringe, D.,
Petsko, G. A., and Gabbay, K. H. (1994)
Biochemistry 33, 20112020
80. Maniscalco, S. J., Saha, S. K., Vicedomine, P.,
and Fisher, H. F. (1996) Biochemistry 35, 8994
81. Saha, S. K., Maniscalco, S. J., and Fisher, H. F.
(1996) Biochemistry 35, 1648316488
82. Smith, M. T., and Emerich, D. W. (1993) J. Biol.
Chem. 268, 1074610753
1. Cornish-Bowden, A., ed. (1997) New Beer in an
Old Bottle (Eduard Buchner and the Growth of
Biochemical Knowledge), Valencia, Universitat
de Valncia
1a. Kalckar, H. M. (1969) Biological Phosphoryla-
tions, Prentice-Hall, Englewood Cliffs, New
Jersey (pp. 8697)
2. Rossmann, M. G., Adams, M. J., Buehner, M.,
Ford, G. C., Hackert, M. L., Lentz, P. J., Jr.,
McPherson, A., Jr., Schevitz, R. W., and
Smiley, I. E. (1971) Cold Spring Harbor
Symposia on Quant. Biol. 36, 179191
3. Abad-Zapatero, C., Griffith, J. P., Sussman, J.
L., and Rossmann, M. G. (1987) J. Mol. Biol.
198, 445467
4. Rossmann, M. (1974) New Scientist 61, 266268
5. Eventoff, W., Rossmann, M. G., Taylor, S. S.,
Torff, H.-J., Meyer, H., Keil, W., and Kiltz,
H.-H. (1977) Proc. Natl. Acad. Sci. U.S.A. 74,
26772681
6. Hogrefe, H. H., Griffith, J. P., Rossmann, M.
G., and Goldberg, E. (1987) J. Biol. Chem. 262,
1315513162
7. Iwata, S., and Ohta, T. (1993) J. Mol. Biol. 230,
2127
8. Ostendorp, R., Auerbach, G., and Jaenicke, R.
(1996) Protein Sci. 5, 862873
9. Birktoft, J. J., Rhodes, G., and Banaszak, L. J.
(1989) Biochemistry 28, 6065 6081
10. Gleason, W. B., Fu, Z., Birktoft, J., and Banaszak,
L. (1994) Biochemistry 33, 20782088
11. Goward, C. R., and Nicholls, D. J. (1994)
Protein Sci. 3, 18831888
12. Hall, M. D., Levitt, D. G., and Banaszak, L. J.
(1992) J. Mol. Biol. 226, 867882
13. Kelly, C. A., Nishiyama, M., Ohnishi, Y.,
Beppu, T., and Birktoft, J. J. (1993) Biochemistry
32, 39133922
14. Bellamacina, C. R. (1996) FASEB J. 10, 1257
1269
15. Pullman, M. E., and Colowick, S. P. (1954) J.
Biol. Chem. 206, 129141
16. Fisher, H. F., Conn, E. E., Vennesland, B., and
Westheimer, F. H. (1953) J. Biol. Chem. 202,
687697
17. Westheiner, F. H. (1987) in Pyridine Nucleotide
Coenzymes: Chemical, Biochemical and Medical
Aspects, Vol. A (Dolphin, D., Avramovic, O.,
and Poulson, R., eds), pp. 253322, Wiley
(Interscience), New York
18. You, K.-s, Arnold, L. J., Jr., Allison, W. S., and
Kaplan, N. O. (1978) Trends Biochem. Sci. 3,
265268
19. Nakajima, N., Nakamura, K., Esaki, N.,
Tanaka, H., and Soda, K. (1989) J. Biochem.
106, 515517
20. Esaki, N., Shimoi, H., Nakajima, N., Ohshima,
T., Tanaka, H., and Soda, K. (1989) J. Biol.
Chem. 264, 97509752
21. Anderson, V. E., and LaReau, R. D. (1988) J.
Am. Chem. Soc. 110, 36953697
22. Almarsson, ., and Bruice, T. C. (1993) J. Am.
Chem. Soc. 115, 21252138
23. Oppenheimer, N. J. (1986) J. Biol. Chem. 261,
1220912212
24. Benner, S. A., Nambiar, K. P., and Chambers,
G. K. (1985) J. Am. Chem. Soc. 107, 55135517
25. Young, L., and Post, C. B. (1993) J. Am. Chem.
Soc. 115, 19641970
26. Almarsson, , Karaman, R., and Bruice, T. C.
(1992) J. Am. Chem. Soc. 114, 87028704
27. LaReau, R. D., and Anderson, V. E. (1992)
Biochemistry 31, 41744180
28. Ohno, A., Tsutsumi, A., Kawai, Y., Yamazaki,
N., Mikata, Y., and Okamura, M. (1994) J. Am.
Chem. Soc. 116, 81338137
29. Kallwass, H. K. W., Hogan, J. K., Macfarlane,
E. L. A., Martichonok, V., Parris, W., Kay, C.
M., Gold, M., and Jones, J. B. (1992) J. Am.
Chem. Soc. 114, 10704 10710
30. Klinman, J. P. (1972) J. Biol. Chem. 247, 7977
7987
31. Ostovic, D., Roberts, R. M. G., and Kreevoy,
M. M. (1983) J. Am. Chem. Soc. 105, 76297631
32. Coleman, C. A., Rose, J. G., and Murray, C. J.
(1992) J. Am. Chem. Soc. 114, 97559762
33. Kong, Y. S., and Warshel, A. (1995) J. Am.
Chem. Soc. 117, 62346242
34. Lee, I.-S. H., Jeoung, E. H., and Kreevoy, M.
M. (1997) J. Am. Chem. Soc. 119, 27222728
35. Rucker, J., Cha, Y., Jonsson, T., Grant, K. L.,
and Klinman, J. P. (1992) Biochemistry 31,
1148911499
36. Huskey, W. P., and Schowen, R. L. (1983) J.
Am. Chem. Soc. 105, 5704 5706
36a. Kohen, A., Cannio, R., Bartolucci, S., and
Klinman, J. P. (1999) Nature (London) 399, 496
499
36b. Chin, J. K., and Klinman, J. P. (2000)
Biochemistry 39, 12781284
36c. Northrop, D. B. (1999) J. Am. Chem. Soc. 121,
35213524
36d. Karsten, W. E., Hwang, C.-C., and Cook, P. F.
(1999) Biochemistry 38, 43984402
37. Grau, U. M., Trommer, W. E., and Rossmann,
M. G. (1981) J. Mol. Biol. 151, 289307
38. Li, H., Hallows, W. H., Punzi, J. S., Pank-
iewicz, K. W., Watanabe, K. A., and Goldstein,
B. M. (1994) Biochemistry 33, 1173411744
39. Gawlita, E., Paneth, P., and Anderson, V. E.
(1995) Biochemistry 34, 6050 6058
40. Clarke, A. R., Wilks, H. M., Barstow, D. A.,
Atkinson, T., Chia, W. N., and Holbrook, J. J.
(1988) Biochemistry 27, 16171622
41. Clarke, A. R., Atkinson, T., and Holbrook, J. J.
(1989) Trends Biochem. Sci. 14, 101105
42. Clarke, A. R., Atkinson, T., and Holbrook, J. J.
(1989) Trends Biochem. Sci. 14, 145148
43. Goldberg, J. D., Yoshida, T., and Brick, P.
(1994) J. Mol. Biol. 236, 11231140
44. Cortes, A., Emery, D. C., Halsall, D. J.,
Jackson, R. M., Clarke, A. R., and Holbrook, J.
J. (1992) Protein Sci. 1, 892901
45. Clarke, A. R., Wigley, D. B., Chia, W. N.,
Barstow, D. A., Atkinson, T., and Holbrook, J.
J. (1986) Nature (London) 324, 699702
46. Deng, H., Burgner, J., and Callender, R. (1992)
J. Am. Chem. Soc. 114, 79978003
47. Grau, W. M., Trommer, W. E., and Rossmann,
M. G. (1981) J. Mol. Biol. 151, 289307
48. Pettersson, G. (1987) CRC Critical Review of
Biochemistry 21, 349389
49. Maret, W., and Makinen, M. W. (1991) J. Biol.
Chem. 266, 20636 20644
50. Eklund, H., and Brndn, C.-I. (1987) in
Pyridine Nucleotide Coenzyme: Chemical,
Biochemical and Medical Aspects, Vol. 2,
Part A (Dolphin, D., Poulson, R., and Avram-
ovic, O., eds), pp. 5198, Wiley, New York
51. Adolph, H. W., Kiefer, M., and Cedergren-
Zeppezauer, E. (1997) Biochemistry 36, 8743
8754
51a. Colby, T. D., Bahnson, B. J., Chin, J. K.,
Klinman, J. P., and Goldstein, B. M. (1998)
Biochemistry 37, 92959304
51b. Plapp, B. V., Eklund, H., and Brndn, C.-I.
(1978) J. Mol. Biol. 122, 2332
52. Shearer, G. L., Kim, K., Lee, K. M., Wang, C.
K., and Plapp, B. V. (1993) Biochemistry 32,
1118611194
53. Ramaswamy, S., Eklund, H., and Plapp, B. V.
(1994) Biochemistry 33, 52305237
References
829
83. Delforge, D., Devreese, B., Dieu, M., Delaive,
E., Van Beeumen, J., and Remacle, J. (1997) J.
Biol. Chem. 272, 22762284
84. Sekimoto, T., Fukui, T., and Tanizawa, K.
(1994) J. Biol. Chem. 269, 72627266
85. Brunhuber, N. M. W., Banerjee, A., Jacobs, W.
R., Jr., and Blanchard, J. S. (1994) J. Biol. Chem.
269, 1620316211
86. Britton, K. L., Baker, P. J., Engel, P. C., Rice, D.
W., and Stillman, T. J. (1993) J. Mol. Biol. 234,
938945
87. Scapin, G., Blanchard, J. S., and Sacchettini, J.
C. (1995) Biochemistry 34, 35023512
88. Moras, D., Olsen, K. W., Sabesan, M. N.,
Buehner, M., Ford, G. C., and Rossmann, M.
G. (1975) J. Biol. Chem. 250, 91379162
89. Due, E., Olivier-Deyris, L., Fanchon, E.,
Corbier, C., Branlant, G., and Dideberg, O.
(1996) J. Mol. Biol. 257, 814838
90. Skarzynski, T., Moody, P. C. E., and Wonacott,
A. J. (1987) J. Mol. Biol. 193, 171187
90a. Roitel, O., Sergienko, E., and Branlant, G.
(1999) Biochemistry 38, 1608416091
91. Kim, H., and Hol, W. G. J. (1998) J. Mol. Biol.
278, 511
92. Soukri, A., Mougin, A., Corbier, C., Wonacott,
A., Branlant, C., and Branlant, G. (1989)
Biochemistry 28, 25682592
93. Habenicht, A., Hellman, U., and Cerff, R.
(1994) J. Mol. Biol. 237, 165171
93a. Marchal, S., Rahuel-Clermont, S., and
Branlant, G. (2000) Biochemistry 39, 33273335
94. Sheikh, S., Ni, L., Hurley, T. D., and Weiner,
H. (1997) J. Biol. Chem. 272, 1881718822
95. Steinmetz, C. G., Xie, P., Weiner, H., and
Hurley, T. D. (1997) Structure 5, 701 711
96. Henehan, G. T. M., and Oppenheimer, N. J.
(1993) Biochemistry 32, 735738
97. Henehan, G. T. M., Chang, S. H., and Oppen-
heimer, N. J. (1995) Biochemistry 34, 12294
12301
98. Shearer, G. L., Kim, K., Lee, K. M., Wang, C.
K., and Plapp, B. V. (1993) Biochemistry 32,
1118611194
99. Feingold, D. S., and Franzen, J. S. (1981)
Trends Biochem. Sci. 6, 103105
100. Frimpong, K., and Rodwell, V. W. (1994) J.
Biol. Chem. 269, 1147811483
101. Omkumar, R. V., and Rodwell, V. W. (1994) J.
Biol. Chem. 269, 1686216866
102. Friesen, J. A., and Rodwell, V. W. (1997)
Biochemistry 36, 11571162
103. Lawrence, C. M., Rodwell, V. W., and
Stauffacher, C. V. (1995) Science 268, 17581762
104. Friesen, J. A., Lawrence, C. M., Stauffacher, C.
V., and Rodwell, V. W. (1996) Biochemistry 35,
1194511950
104a. Cosgrove, M. S., Naylor, C., Paludan, S.,
Adams, M. J., and Levy, H. R. (1998) Biochem-
istry 37, 27592767
105. Luzzatto, L., and Mehta, A. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 3367
3398, McGraw-Hill, New York
106. Wakil, S. J. (1989) Biochemistry 28, 45234530
107. Smith, S. (1994) FASEB J. 8, 12481259
107a. Barycki, J. J., OBrien, L. K., Bratt, J. M.,
Zhang, R., Sanishvili, R., Strauss, A. W., and
Banaszak, L. J. (1999) Biochemistry 38, 5786
5798
107b. Fillgrove, K. L., and Anderson, V. E. (2000)
Biochemistry 39, 70017011
108. Watkinson, I. A., Wilton, D. L., Rahimtula, A.
D., and Akhtar, M. M. (1971) Eur. J. Biochem.
23, 16
109. Thorn, J. M., Barton, J. D., Dixon, N. E., Ollis,
D. L., and Edwards, K. J. (1995) J. Mol. Biol.
249, 785799
References
110. Bolduc, J. M., Dyer, D. H., Scott, W. G., Singer,
P., Sweet, R. M., Koshland, D. E., Jr., and
Stoddard, B. L. (1995) Science 268, 13121318
111. Lee, M. E., Dyer, D. H., Klein, O. D., Bolduc, J.
M., Stoddard, B. L., and Koshland, D. E., Jr.
(1995) Biochemistry 34, 378384
112. Schtz, M., and Radler, F. (1973) Arch.
Mikrobiol. 91, 183
113. Hwang, C.-C., and Cook, P. F. (1998) Biochem-
istry 37, 1569815702
114. Karsten, W. E., Gavva, S. R., Park, S.-H., and
Cook, P. F. (1995) Biochemistry 34, 32533260
115. Caspritz, G., and Radler, F. (1983) J. Biol. Chem.
258, 49074910
116. Thoden, J. B., Frey, P. A., and Holden, H. M.
(1996) Biochemistry 35, 51375144
117. Frey, P. A. (1987) in Pyridine Nucleotide
Coenzymes: Chemical, Biochemical and Medical
Aspects, Vol. B (Dolphin, D., Avramovic, O.,
and Poulson, R., eds), pp. 461512, Wiley
(Interscience), New York
117a. Thoden, J. B., Wohlers, T. M., Fridovich-Keil,
J. L., and Holden, H. M. (2000) Biochemistry
39, 56915701
118. Thoden, J. B., Hegeman, A. D., Wesenberg, G.,
Chapeau, M. C., Frey, P. A., and Holden, H.
M. (1997) Biochemistry 36, 62946304
119. Palmer, J. L., and Abeles, R. H. (1979) J. Biol.
Chem. 254, 12171226
120. Yuan, C.-S., Ault-Rich, D. B., and Borchardt,
R. T. (1996) J. Biol. Chem. 271, 2800928016
121. Koch-Nolte, F., Petersen, D., Balasubramani-
an, S., Haag, F., Kahlke, D., Willer, T.,
Kastelein, R., Bazan, F., and Thiele, H.-G.
(1996) J. Biol. Chem. 271, 76867693
122. Takada, T., Iida, K., and Moss, J. (1995) J. Biol.
Chem. 270, 541 544
123. Rising, K. A., and Schramm, V. L. (1997) J. Am.
Chem. Soc. 119, 2737
124. Muller-Steffner, H. M., Augustin, A., and
Schuber, F. (1996) J. Biol. Chem. 271, 23967
23972
125. De Flora, A., Guida, L., Franco, L., Zocchi, E.,
Bruzzone, S., Benatti, U., Damonte, G., and
Lee, H. C. (1997) J. Biol. Chem. 272, 12945
12951
126. Aarhus, R., Graeff, R. M., Dickey, D. M.,
Walseth, T. F., and Lee, H. C. (1995) J. Biol.
Chem. 270, 3032730333
127. Chini, E. N., Beers, K. W., and Dousa, T. P.
(1995) J. Biol. Chem. 270, 32163223
128. Oppenheimer, N. J. (1987) in Pyridine
Nucleotide Coenzymes: Chemical, Biochemical
and Medical Aspects, Vol. A (Dolphin, D.,
Avramovic, O., and Poulson, R., eds), pp.
323365, Wiley (Interscience), New York
129. Everse, J., Anderson, B., and You, K.-S., eds.
(1982) The Pyridine Nucleotide Coenzymes,
Academic Press, New York
130. Blankenhorn, G., and Moore, E. G. (1980) J.
Am. Chem. Soc. 102, 10921098
131. Bernofsky, C. (1987) in Pyridine Nucleotide
Coenzymes: Chemical, Biochemical and Medical
Aspects, Vol. B (Dolphin, D., Avramovic, O.,
and Poulson, R., eds), pp. 105172, Wiley
(Interscience), New York
132. Chaykin, S. (1967) Ann. Rev. Biochem. 36, (I),
149170
133. Everse, J., and Kaplan, N. O. (1973) Adv.
Enzymol. 37, 61133
133a. Rozwarski, D. A., Grant, G. A., Barton, D. H.
R., Jacobs, W. R. J., and Sacchettini, J. C. (1998)
Science 279, 98102
133b. Parikh, S., Moynihan, D. P., Xiao, G., and
Tonge, P. J. (1999) Biochemistry 38, 13623
13634
133c. McMurry, L. M., Oethinger, M., and Levy, S.
B. (1998) Nature (London) 394, 531532
133d. Roujeinikova, A., and 14 other authors. (1999)
J. Mol. Biol. 294, 527 535
133e. Parikh, S. L., Xiao, G., and Tonge, P. J. (2000)
Biochemistry 39, 7645 7650
134. Johnson, R. W., Marschner, T. M., and
Oppenheimer, N. J. (1988) J. Am. Chem. Soc.
110, 22572263
135. Oppenheimer, N. J. (1973) Biochem. Biophys.
Res. Commun. 50, 683690
136. Acheson, S. A., Kirkman, H. N., and Wolfend-
en, R. (1988) Biochemistry 27, 73717375
137. Marshall, J. L., Booth, J. E., and Williams, J. W.
(1984) J. Biol. Chem. 259, 30333036
138. Bernofsky, C., and Wanda, S.-Y. C. (1982) J.
Biol. Chem. 257, 68096817
139. Woenckhaus, C. H. (1974) in Topics in Current
Chemistry (Fortschritte der chemischen Forschung,
Vol. 52 (Boschke, F. L., ed), pp. 209233,
Springer-Verlag, Berlin
140. Anderson, B. M. (1982) in The Pyridine
Nucleotide Coenzymes (Everse, J., Anderson, B.,
and You, K.-S., eds), Academic Press, New
York
141. Woenckhaus, C., and Jeck, R. (1987) in
Pyridine Nucleotide Coenzymes: Chemical,
Biochemical and Medical Aspects, Vol. A
(Dolphin, D., Avramovic, O., and Poulson, R.,
eds), pp. 449568, Wiley (Interscience), New
York
142. Cohen, B. E., Stoddard, B. L., and Koshland,
D. E., Jr. (1997) Biochemistry 36, 9035 9044
143. Hemmerich, P. (1976) in Progress in the
Chemistry of Organic Natural Products, Vol. 33
(Herz, W., Grisebach, H., and Kirby, G. W.,
eds), pp. 451527, Springer-Verlag, New York
144. Fox, K. M., and Karplus, P. A. (1999) J. Biol.
Chem. 274, 9357 9362
145. Niino, Y. S., Chakraborty, S., Brown, B. J., and
Massey, V. (1995) J. Biol. Chem. 270, 19831991
146. Meighen, E. A. (1993) FASEB J. 7, 10161022
147. Fisher, A. J., Raushel, F. M., Baldwin, T. O., and
Rayment, I. (1995) Biochemistry 34, 65816586
148. Van den Berghe-Snorek, S., and Stankovich,
M. T. (1984) J. Am. Chem. Soc. 106, 36853687
149. Miura, R., Nishina, Y., Sato, K., Fujii, S.,
Kuroda, K., and Shiga, K. (1993) J. Biochem.
113, 106113
150. Lim, L. W., Shamala, N., Mathews, F. S.,
Steenkamp, D. J., Hamlin, R., and Xuong, N.
(1986) J. Biol. Chem. 261, 1514015146
151. Massey, V. (1995) FASEB J. 9, 473475
152. Mller, F. (1991) Chemistry and Biochemistry of
Flavoenzymes, Vol. I, CRC Press, Boca Raton,
Florida
153. Curti, B., Ronchi, S., and Zanetti, G., eds.
(1990) Flavins and Flavoproteins, Walter de
Gruyter, Berlin
153a. Fraaije, M. W., and Mattevi, A. (2000) Trends
Biochem. Sci. 25, 126132
154. Kohen, A., Jonsson, T., and Klinman, J. P.
(1997) Biochemistry 36, 2603 2611
154a. Su, Q., and Klinman, J. P. (1999) Biochemistry
38, 85728581
155. Bourdillon, C., Demaille, C., Moiroux, J., and
Savant, J.-M. (1993) J. Am. Chem. Soc. 115, 210
156. Li, J., Vrielink, A., Brick, P., and Blow, D. M.
(1993) Biochemistry 32, 1150711515
157. Lindqvist, Y., and Brndn, C.-I. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 68556859
158. Lindqvist, Y., and Brndn, C.-I. (1989) J. Biol.
Chem. 264, 36243628
159. Stenberg, K., and Lindqvist, Y. (1997) Protein
Sci. 6, 10091015
160. Yagi, K., and Ozawa, T. (1989) Biochim.
Biophys. Acta. 1000, 203206
160a. Curti, B., Ronchi, S., and Simonetta, M. P.
(1990) in Chemistry and Biochemistry of
Flavoenzymes, Vol. III (Mller, F., ed), pp. 69
94, CRC Press, Boca Raton, Florida
830 Chapter 15. Coenzymes of OxidationReduction Reactions
214. Chen, S., Clarke, P. E., Martino, P. A., Dang, P.
S. K., Yeh, C.-H., Lee, T. D., Prochaska, H. J.,
and Talalay, P. (1994) Protein Sci. 3, 12961304
215. Claiborne, A., Ross, R. P., and Parsonage, D.
(1992) Trends Biochem. Sci. 17, 183186
216. Parsonage, D., and Claiborne, A. (1995)
Biochemistry 34, 435441
217. Yeh, J. I., Claiborne, A., and Hol, W. G. J.
(1996) Biochemistry 35, 99519957
218. Poole, L. B. (1996) Biochemistry 35, 6575
219. Mewies, M., McIntire, W. S., and Scrutton, N.
S. (1998) Protein Sci. 7, 7 20
220. Brandsch, R., and Bichler, V. (1991) J. Biol. Chem.
266, 1905619062
221. Willie, A., Edmondson, D. E., and Jorns, M. S.
(1996) Biochemistry 35, 52925299
222. Kenney, W. C., Singer, T. P., Fukuyama, M., and
Miyake, Y. (1979) J. Biol. Chem. 254, 46894690
223. Kim, J., Fuller, J. H., Kuusk, V., Cunane, L.,
Chen, Z.-w, Mathews, F. S., and McIntire, W.
S. (1995) J. Biol. Chem. 270, 3120231209
224. Mewies, M., Basran, J., Packman, L. C., Hille,
R., and Scrutton, N. S. (1997) Biochemistry 36,
71627168
225. Ghisla, S., and Mayhew, S. G. (1976) Eur. J.
Biochem. 63, 373390
226. Hartman, H. A., Edmondson, D. E., and
McCormick, D. B. (1992) Anal. Biochem. 202,
348355
227. Eirich, L. D., Vogels, G. D., and Wolfe, R. S.
(1979) J. Bacterial. 140, 2027
228. Jacobson, F., and Walsh, C. (1984) Biochemistry
23, 979988
229. Jones, J. B., and Stadtman, T. C. (1980) J. Biol.
Chem. 255, 10491053
230. Sancar, A. (1994) Biochemistry 33, 29
231. Otani, S., Takatsu, M., Nakano, M., Kasai, S.,
Miura, R., and Matsui, K. (1974) The Journal of
Antibiotics 27, 8889
232. Murthy, Y. V. S. N., and Massey, V. (1995) J.
Biol. Chem. 270, 2858628594
233. Ghisla, S., and Massey, V. (1989) Eur. J.
Biochem. 2, 243289
234. Powell, M. F., and Bruice, T. C. (1983) J. Am.
Chem. Soc. 105, 10141021
235. Fox, J. L., and Tolin, G. (1966) Biochemistry 5,
38653872
236. Lee, I.-S. H., Ostovc, D., and Kreevoy, M.
(1988) J. Am. Chem. Soc. 110, 39893993
237. Brustlein, M., and Bruice, T. C. (1972) J. Am.
Chem. Soc. 94, 65486549
238. Jorns, M. S., and Hersch, L. B. (1975) J. Biol.
Chem. 250, 36203628
239. Ghisla, S., Thorpe, C., and Massey, V. (1984)
Biochemistry 23, 31543161
240. Kim, J.-J. P., Wang, M., and Paschke, R. (1993)
Proc. Natl. Acad. Sci. U.S.A. 90, 75237527
241. Dakoji, S., Shin, I., Becker, D. F., Stankovich,
M. T., and Liu, H.-w. (1996) J. Am. Chem. Soc.
118, 1097110979
242. Tompkins, L. S., and Falkow, S. (1995) Science
267, 16211622
242a. Vock, P., Engst, S., Eder, M., and Ghisla, S.
(1998) Biochemistry 37, 1848 1860
243. Johnson, B. D., Mancini-Samuelson, G. J., and
Stankovich, M. T. (1995) Biochemistry 34,
70477055
244. Thorpe, C. (1989) Trends Biochem. Sci. 14, 148151
245. Djordjevic, S., Pace, C. P., Stankovich, M. T., and
Kim, J.-J. P. (1995) Biochemistry 34, 21632171
246. Porter, D. J. T., Voet, J. G., and Bright, H. J.
(1973) J. Biol. Chem. 248, 44004416
247. Alston, T. A., Porter, D. J. T., and Bright, H. J.
(1983) J. Biol. Chem. 258, 11361141
248. Gadda, G., Edmondson, R. D., Russell, D. H.,
and Fitzpatrick, P. F. (1997) J. Biol. Chem. 272,
55635570
249. Lehoux, I., and Mitra, B. (1997) FASEB J. 11,
A889
161. Mizutani, H., Miyahara, I., Hirotsu, K.,
Nishina, Y., Shiga, K., Setoyama, C., and
Miura, R. (1996) J. Biochem. 120, 1417
162. Mattevi, A., Vanoni, M. A., Todone, F., Rizzi,
M., Teplyakov, A., Coda, A., Bolognesi, M.,
and Curti, B. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 74967501
163. Vanoni, M. A., Cosma, A., Mazzeo, D.,
Mattevi, A., Todone, F., and Curti, B. (1997)
Biochemistry 36, 56245632
164. Tan, A. K., and Ramsay, R. R. (1993) Biochemis-
try 32, 2137 2143
165. Woo, J. C. G., and Silverman, R. B. (1995) J.
Am. Chem. Soc. 117, 16631664
166. Walker, M. C., and Edmondson, D. E. (1994)
Biochemistry 33, 70887098
166a. Pawelek, P. D., Cheah, J., Coulombe, R.,
Macheroux, P., Ghisla, S., and Vrielink, A.
(2000) EMBO J. 19, 42044215
167. Barber, M. J., Neame, P. J., Lim, L. W., White,
S., and Mathews, F. S. (1992) J. Biol. Chem. 267,
6611 6619
168. Trickey, P., Basran, J., Lian, L.-Y., Chen, Z.-w,
Barton, J. D., Sutcliffe, M. J., Scrutton, N. S.,
and Mathews, F. S. (2000) Biochemistry 39,
7678 7688
168a. Jang, M.-H., Basran, J., Scrutton, N. S., and
Hille, R. (1999) J. Biol. Chem. 274, 1314713154
169. Falzon, L., and Davidson, V. L. (1996)
Biochemistry 35, 12111 12118
170. Lindqvist, Y., Brndn, C.-I., Mathews, F. S.,
and Lederer, F. (1991) J. Biol. Chem. 266, 3198
3207
171. Tegoni, M., and Cambillau, C. (1994) Protein
Sci. 3, 303313
172. Gondry, M., and Lederer, F. (1996) Biochemis-
try 35, 85878594
173. Tegoni, M., Gervais, M., and Desbois, A.
(1997) Biochemistry 36, 89328946
173a. Lehoux, I. E., and Mitra, B. (2000) Biochemistry
39, 1005510065
174. Schaller, R. A., Mohsen, A.-W. A., Vockley, J.,
and Thorpe, C. (1997) Biochemistry 36, 7761
7768
175. Rudik, I., Ghisla, S., and Thorpe, C. (1998)
Biochemistry 37, 84378445
176. Mancini-Samuelson, G. J., Kieweg, V., Sabaj,
K. M., Ghisla, S., and Stankovich, M. T. (1998)
Biochemistry 37, 1460514612
176a. Srivastava, D. K., and Peterson, K. L. (1998)
Biochemistry 37, 84468456
177. Mohsen, A.-W. A., and Vockley, J. (1995)
Biochemistry 34, 1014610152
177a. Ackrell, B., McIntire, B., and Vessey, D. (2000)
Trends Biochem. Sci. 25, 910
178. Rtey, J., Seibl, J., Arigoni, D., Cornforth, J. W.,
Ryback, G., Zeylemaker, W. P., and Veeger, C.
(1970) Eur. J. Biochem. 14, 232242
179. Birch-Machin, M. A., Farnsworth, L., Ackrell,
B. A. C., Cochran, B., Jackson, S., Bindoff, L.
A., Aitken, A., Diamond, A. G., and Turnbull,
D. M. (1992) J. Biol. Chem. 267, 1155311558
180. Schmidt, D. M., Saghbini, M., and Scheffler, I.
E. (1992) Biochemistry 31, 84428448
181. Sucheta, A., Ackrell, B. A. C., Cochran, B., and
Armstrong, F. A. (1992) Nature (London) 356,
361362
182. Thieme, R., Pai, E. F., Schirmer, R. H., and
Schulz, G. E. (1981) J. Mol. Biol. 152, 763782
183. Sled, V. D., Rudnitzky, N. I., Hatefi, Y., and
Ohnishi, T. (1994) Biochemistry 33, 1006910075
184. Westenberg, D. J., Gunsalus, R. P., Ackrell, B.
A. C., Sices, H., and Cecchini, G. (1993) J. Biol.
Chem. 268, 815822
185. Hgerhll, C., Fridn, H., Aasa, R., and
Hederstedt, L. (1995) Biochemistry 34, 11080
11089
186. Van Hellemond, J. J., Klockiewicz, M.,
Gaasenbeek, C. P. H., Roos, M. H., and
Tielens, A. G. M. (1995) J. Biol. Chem. 270,
3106531070
187. Waksman, G., Krishna, T. S. R., Williams, J.,
CH, and Kuriyan, J. (1994) J. Mol. Biol. 236,
800816
188. Mulrooney, S. B., and Williams, C. H. (1994)
Biochemistry 33, 31483154
189. Arscott, L. D., Gromer, S., Schirmer, R. H.,
Becker, K., and Williams, C. H., Jr. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 36213626
190. Lennon, B. W., and Williams, C. H., Jr. (1995)
Biochemistry 34, 36703677
191. Pai, E. F., and Schulz, G. E. (1983) J. Biol.
Chem. 258, 17521757
192. Karplus, P. A., and Schulz, G. E. (1987) J. Mol.
Biol. 195, 701729
193. Mittl, P. R. E., and Schulz, G. E. (1994) Protein
Sci. 3, 799809
194. Zhang, Y., Bond, C. S., Bailey, S., Cunning-
ham, M. L., Fairlamb, A. H., and Hunter, W.
N. (1996) Protein Sci. 5, 5261
195. Walsh, C., Bradley, M., and Nadeau, K. (1991)
Trends Biochem. Sci. 16, 305309
196. Zheng, R., Cenas, N., and Blanchard, J. S.
(1995) Biochemistry 34, 1269712703
197. Krauth-Siegel, R. L., and Schneck, R. (1995)
FASEB J. 9, 11381146
198. Brown, N. L. (1985) Trends Biochem. Sci. 10,
400403
199. Schiering, N., Kabsch, W., Moore, M. J.,
Distefano, M. D., Walsh, C. T., and Pai, E. F.
(1991) Nature (London) 352, 168172
199a. Engst, S., and Miller, S. M. (1999) Biochemistry
38, 35193529
200. Serre, L., Vellieux, F. M. D., Gomez-Moreno, M.
M. C., Fontecilla-Camps, J. C., and Frey, M.
(1996) J. Mol. Biol. 263, 2039
201. Aliverti, A., Bruns, C. M., Pandini, V. E.,
Karplus, P. A., Vanoni, M. A., Curti, B., and
Zanetti, G. (1995) Biochemistry 34, 83718379
202. Ermler, U., Siddiqui, R. A., Cramm, R., and
Friedrich, B. (1995) EMBO J. 14, 60676077
203. Ingelman, M., Bianchi, V., and Eklund, H.
(1997) J. Mol. Biol. 268, 147157
204. Menz, R. I., and Day, D. A. (1996) J. Biol. Chem.
271, 2311723120
205. Takano, S., Yano, T., and Yagi, T. (1996)
Biochemistry 35, 91209127
205a. Morandi, P., Valzasina, B., Colombo, C., Curti,
B., and Vanoni, M. A. (2000) Biochemistry 39,
727735
206. Fieschi, F., Nivire, V., Frier, C., Dcout, J.-L.,
and Fontecave, M. (1995) J. Biol. Chem. 270,
3039230400
206a. Ingelman, M., Ramaswamy, S., Nivire, V.,
Fontecave, M., and Eklund, H. (1999)
Biochemistry 38, 70407049
207. Tanner, J. J., Lei, B., Tu, S.-C., and Krause, K.
L. (1996) Biochemistry 35, 1353113539
208. Porter, T. D. (1991) Trends Biochem. Sci. 16,
154158
209. Vang, M., Roberts, D. L., Paschke, R., Shea, T.
M., Masters, B. S. S., and Kin, J.-J. P. (1997)
Proc. Natl. Acad. Sci. U.S.A. 94, 8411 8416
209a. Uhlmann, H., and Bernhardt, R. (1995) J. Biol.
Chem. 270, 2995929966
209b. Ziegler, G. A., and Schulz, G. E. (2000)
Biochemistry 39, 1098610995
210. Kobayashi, K., Tagawa, S., Sano, S., and Asada,
K. (1995) J. Biol. Chem. 270, 2755127554
211. Sano, S., Miyake, C., Mikami, B., and Asada,
K. (1995) J. Biol. Chem. 270, 21354 21361
212. Tedeschi, G., Chen, S., and Massey, V. (1995) J.
Biol. Chem. 270, 11981204
213. Li, R., Bianchet, M. A., Talalay, P., and Amzel,
L. M. (1995) Proc. Natl. Acad. Sci. U.S.A. 92,
88468850
References
831
250. Ghisla, S., and Massey, V. (1980) J. Biol. Chem.
255, 56885696
251. Hamilton, G. H. (1971) Prog. Bioorg. Chem. 1,
83157
252. Walsh, C. T., Krodel, E., Massey, V., and
Abeles, R. H. (1973) J. Biol. Chem. 248, 1946
1951
253. Ghisla, S., and Massey, V. (1989) Eur. J.
Biochem. 181, 117
254. Curti, B., Ronchi, S., and Simonetta, M. P.
(1992) in Chemistry and Biochemistry of
Flavoenzymes (Mller, F., ed), pp. 6994, CRC
Press, Boca Raton, Florida
255. Lederer, F. (1992) Protein Sci. 1, 540548
256. Miura, R., and Miyake, Y. (1988) Bioorg. Chem.
16, 97110
257. Todone, F., Vanoni, M. A., Mozzarelli, A.,
Bolognesi, M., Coda, A., Curti, B., and
Mattevi, A. (1997) Biochemistry 36, 58535860
258. Rietveld, P., Arscott, L. D., Berry, A., Scrutton,
N. S., Deonarain, M. P., Perham, R. N., and
Williams, C. H. (1994) Biochemistry 33, 13888
13895
258a. Krauth-Siegel, R. L., Arscott, L. D.,
Schnleben-Janas, A., Schirmer, R. H., and
Williams, C. H., Jr. (1998) Biochemistry 37,
1396813977
259. Williams, C. H., Jr. (1995) FASEB J. 9, 12671276
260. Barman, B. G., and Tollin, G. (1972) Biochemis-
try 11, 47604765
261. Su, Y., and Tripathi, G. N. R. (1994) J. Am.
Chem. Soc. 116, 44054407
262. Zheng, Y.-J., and Ornstein, R. L. (1996) J. Am.
Chem. Soc. 118, 94029408
263. Mller, F., Hemmerich, P., and Ehrenberg, A.
(1971) in Flavins and Flavoproteins (Kamin, H.,
ed), pp. 107180, Univ. Park Press, Baltimore,
Maryland
264. Mller, F., Eriksson, L. E. G., and Ehrenberg,
A. (1970) Eur. J. Biochem. 12, 93103
265. Miura, R., Setoyama, C., Nishina, Y., Shiga, K.,
Mizutani, H., Miyahara, I., and Hirotsu, K.
(1997) FASEB J. 11, A1306
265a. Miura, R., Setoyama, C., Nishina, Y., Shiga, K.,
Mizutani, H., Miyahara, I., and Hirotsu, K.
(1977) J. Biochem. 122, 825833
266. Smith, W. W., Burnett, R. M., Darling, G. D.,
and Ludwig, M. L. (1977) J. Mol. Biol. 117,
195225
267. Chang, F.-C., and Swenson, R. P. (1999)
Biochemistry 38, 71687176
268. Zhou, Z., and Swenson, R. P. (1996) Biochemistry
35, 1598015988
269. Ludwig, M. L., Pattridge, K. A., Metzger, A.
L., and Dixon, M. M. (1997) Biochemistry 36,
12591280
269a. Lostao, A., Gmez-Moreno, C., Mayhew, S.
G., and Sancho, J. (1997) Biochemistry 36,
1433414344
270. Hoover, D. M., Drennan, C. L., Metzger, A. L.,
Osborne, C., Weber, C. H., Pattridge, K. A.,
and Ludwig, M. L. (1999) J. Mol. Biol. 294,
725743
271. Moonen, C. T. W., and Mller, F. (1982)
Biochemistry 21, 408414
272. Jorns, M. S., Sancar, G. B., and Sancar, A.
(1984) Biochemistry 23, 26732679
273. McKean, M. C., Beckman, J. D., and Frerman,
F. E. (1983) J. Biol. Chem. 258, 18661870
274. Roberts, D. L., Salazar, D., Fulmer, J. P.,
Frerman, F. E., and Kim, J.-J. P. (1999)
Biochemistry 38, 19771989
275. Huang, L., Rohlfs, R. J., and Hille, R. (1995) J.
Biol. Chem. 270, 2395823965
276. Balasubramanian, S., Coggins, J. R., and
Abell, C. (1995) Biochemistry 34, 341348
277. Macheroux, P., Petersen, J., Bornemann, S.,
Lowe, D. J., and Thorneley, R. N. F. (1996)
Biochemistry 35, 16431652
References
278. Yu, M. W., and Fritchie, C. J. J. (1975) J. Biol.
Chem. 250, 946951
279. Kenney, W. C., and Singer, T. P. (1977) J. Biol.
Chem. 252, 47674772
280. Ostrowski, J., Barber, M. J., Rueger, D. C.,
Miller, B. E., Siegel, L. M., and Kredich, N. M.
(1989) J. Biol. Chem. 264, 15796 15808
281. Covs, J., Zeghouf, M., Macherel, D., Guigliar-
elli, B., Asso, M., and Fontecave, M. (1997)
Biochemistry 36, 59215928
282. Campbell, W. H., and Kinghorn, J. R. (1990)
Trends Biochem. Sci. 15, 315319
283. Hyde, G. E., Crawford, N. M., and Campbell,
W. H. (1991) J. Biol. Chem. 266, 2354223547
283a. Terao, M., Kurosaki, M., Saltini, G., Demontis,
S., Marini, M., Salmona, M., and Garattini, E.
(2000) J. Biol. Chem. 275, 3069030700
284. Bruice, T. C. (1980) Acc. Chem. Res. 13, 256262
285. Entsch, B., Palfey, B. A., Ballou, D. P., and
Massey, V. (1991) J. Biol. Chem. 266, 1734117349
286. Gatti, D. L., Entsch, B., Ballou, D. P., and
Ludwig, M. L. (1996) Biochemistry 35, 567578
287. Schreuder, H. A., Hol, W. G. J., and Drenth, J.
(1990) Biochemistry 29, 31013108
288. Yamasaki, M., and Yamano, T. (1973) Biochem.
Biophys. Res. Commun. 51, 612619
289. Mernyi, G., and Lind, J. (1991) J. Am. Chem.
Soc. 113, 31463153
290. Entsch, B., and van Berkel, W. J. H. (1995)
FASEB J. 9, 476483
291. Snell, E. E., and Broquist, H. P. (1949) Arch.
Biochem. Biophys. 23, 326328
291a. Reed, L. J. (1998) Protein Sci. 7, 220224
292. Reed, L. J., DeBusk, B. G., Gunsalus, I. C., and
Hornberger, C. S. (1951) Science 114, 9394
293. Jukes, T. H. (1997) Protein Sci. 6, 254256
294. Schmidt, U., Grafen, P., Altland, K., and
Goedde, H. W. (1969) Adv. Enzymol. 32, 432469
295. Reed, K. E., Morris, T. W., and Cronan, J. E., Jr.
(1994) Proc. Natl. Acad. Sci. U.S.A. 91, 37203724
296. Pares, S., Cohen-Addad, C., Sieker, L.,
Neuburger, M., and Douce, R. (1994) Proc.
Natl. Acad. Sci. U.S.A. 91, 48504853
296a. Morris, T. W., Reed, K. E., and Cronan, J. E., Jr.
(1994) J. Biol. Chem. 269, 1609116100
297. Roche, T. E., and Patel, M. E., eds. (1990)
Alpha-Keto Acid Dehydrogenase Complexes
Organization, Regulation, and Biomedical
Ramifications, Vol. 573, New York Academy of
Sciences, New York
298. Patel, M. S., and Roche, T. E. (1990) FASEB J.
4, 32243233
299. Stoops, J. K., Cheng, R. H., Yazdi, M. A.,
Maeng, C.-Y., Schroeter, J. P., Klueppelberg, U.,
Kolodziej, S. J., Baker, T. S., and Reed, L. J.
(1997) J. Biol. Chem. 272, 57575764
300. Yi, J., Nemeria, N., McNally, A., Jordan, F.,
Machado, R. S., and Guest, J. R. (1996) J. Biol.
Chem. 271, 3319233200
301. Berg, A., Vervoort, J., and de Kok, A. (1996) J.
Mol. Biol. 261, 432442
302. Wynn, R. M., Davie, J. R., Zhi, W., Cox, R. P.,
and Chuang, D. T. (1994) Biochemistry 33,
89628968
302a. Wynn, R. M., Davie, J. R., Chuang, J. L., Cote,
C. D., and Chuang, D. T. (1998) J. Biol. Chem.
273, 1311013118
303. Mattevi, A., Obmolova, G., Kalk, K. H., van
Berkel, W. J. H., and Hol, W. G. J. (1993) J.
Mol. Biol. 230, 12001215
304. Liu, T.-C., Korotchkina, L. G., Hyatt, S. L.,
Vettakkorumakankav, N. N., and Patel, M. S.
(1995) J. Biol. Chem. 270, 1554515550
305. Guan, Y., Rawsthorne, S., Scofield, G., Shaw,
P., and Doonan, J. (1995) J. Biol. Chem. 270,
54125417
306. Mattevi, A., Obmolova, G., Schulze, E., Kalk,
K. H., Westphal, A. H., de Kok, A., and Hol, W.
G. J. (1992) Science 255, 15441550
306a. Mattevi, A., Obmolova, G., Kalk, K. H.,
Westphal, A. H., de Kok, A., and Hol, W. G. J.
(1993) J. Mol. Biol. 230, 11831199
306b. Izard, T., AEvarsson, A., Allen, M. D.,
Westphal, A. H., Perhan, R. N., de Kok, A.,
and Hol, W. G. J. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 12401245
307. Reed, L. J., and Hackert, M. L. (1990) J. Biol.
Chem. 265, 89718974
307a. Knapp, J. E., Mitchell, D. T., Yazdi, M. A.,
Ernst, S. R., Reed, L. J., and Hackert, M. L.
(1998) J. Mol. Biol. 280, 655668
308. Dardel, F., Davis, A. L., Laue, E. D., and Perham,
R. N. (1993) J. Mol. Biol. 229, 10371048
309. Green, J. D. F., Laue, E. D., Perham, R. N., Ali,
S. T., and Guest, J. R. (1995) J. Mol. Biol. 248,
328343
310. Mattevi, A., Obmolova, G., Kalk, K. H.,
Teplyakov, A., and Hol, W. G. J. (1993)
Biochemistry 32, 38873901
311. Hendle, J., Mattevi, A., Westphal, A. H., Spee,
J., de Kok, A., Teplyakov, A., and Hol, W. G. J.
(1995) Biochemistry 34, 42874298
312. Ricaud, P. M., Howard, M. J., Roberts, E. L.,
Broadhurst, R. W., and Perham, R. N. (1996) J.
Mol. Biol. 264, 179190
313. Maeng, C.-Y., Yazdi, M. A., Niu, X.-D., Lee, H.
Y., and Reed, L. J. (1994) Biochemistry 33,
1380113807
314. McCartney, R. G., Sanderson, S. J., and Lindsay,
J. G. (1997) Biochemistry 36, 68196826
315. Yang, Y.-S., and Frey, P. A. (1986) Biochemistry
25, 81738178
315a. Pan, K., and Jordan, F. (1998) Biochemistry 37,
13571364
315b. Hennig, J., Kern, G., Neef, H., Spinka, M.,
Bisswanger, H., and Hbner, G. (1997)
Biochemistry 36, 15772 15779
315c. Kee, K., Niu, L., and Henderson, E. (1998)
Biochemistry 37, 4224 4234
316. Han, D., Handelman, G., Marcocci, L., Sen, C.
K., Roy, S., Kobuchi, H., Tritschler, H. J.,
Floh, L., and Packer, L. (1997) BioFactors 6,
321338
317. Muller, Y. A., Schumacher, G., Rudolph, R., and
Schulz, G. E. (1994) J. Mol. Biol. 237, 315335
318. Kerscher, L., and Oesterhelt, D. (1982) Trends
Biochem. Sci. 7, 371374
319. Muller, Y. A., and Schulz, G. E. (1993) Science
259, 965967
319a. Tittmann, K., Golbik, R., Ghisla, S., and
Hbner, G. (2000) Biochemistry 39, 10747
10754
320. Menon, S., and Ragsdale, S. W. (1997) Biochem-
istry 36, 84848494
320a. Bouchev, V. F., Furdui, C. M., Menon, S.,
Muthukumaran, R. B., Ragsdale, S. W., and
McCracken, J. (1999) J. Am. Chem. Soc. 121,
37243729
321. Smith, E. T., Blamey, J. M., and Adams, M. W.
W. (1994) Biochemistry 33, 10081016
322. Mai, X., and Adams, M. W. W. (1994) J. Biol.
Chem. 269, 1672616732
323. Gehring, V., and Arnon, D. I. (1972) J. Biol.
Chem. 247, 69636969
324. Sun, W., Williams, C. H., Jr., and Massey, V.
(1996) J. Biol. Chem. 271, 1722617233
325. Mh, U., Williams, C. H., and Massey, V.
(1994) J. Biol. Chem. 269, 79948000
326. Unkrig, V., Neugebauer, F. A., and Knappe, J.
(1989) Eur. J. Biochem. 184, 723728
327. Maiorino, M., Chu, F. F., Ursini, F., Davies, K.
J. A., Doroshow, J. H., and Esworthy, R. S.
(1991) J. Biol. Chem. 266, 77287732
328. Wong, K. K., Murray, B. W., Lewisch, S. A.,
Baxter, M. K., Ridky, T. W., Ulissi-DeMario, L.,
and Kozarich, J. W. (1993) Biochemistry 32,
1410214110
832 Chapter 15. Coenzymes of OxidationReduction Reactions
387. Meiering, E. M., and Wagner, G. (1995) J. Mol.
Biol. 247, 294308
388. Cannon, W. R., Garrison, B. J., and Benkovic,
S. J. (1997) J. Am. Chem. Soc. 119, 23862395
389. Nakano, T., Spencer, H. T., Appleman, J. R.,
and Blakley, R. L. (1994) Biochemistry 33,
99459952
390. Howell, E. E., Villafranca, J. E., Warren, M. S.,
Oatley, S. J., and Kraut, J. (1986) Science 231,
11231128
391. Benkovic, S. J., Fierke, C. A., and Naylor, A.
M. (1988) Science 239, 11051110
392. Chen, Y.-Q., Kraut, J., Blakley, R. L., and
Callender, R. (1994) Biochemistry 33, 70237032
393. Basran, J., Casarotto, M. G., Barsukov, I. L.,
and Roberts, G. C. K. (1995) Biochemistry 34,
28722882
394. Jeong, S.-S., and Gready, J. E. (1995) Biochemis-
try 34, 37343741
395. Appling, D. R. (1991) FASEB J. 5, 26452651
396. Stover, P. J., Chen, L. H., Suh, J. R., Stover, D.
M., Keyomarsi, K., and Shane, B. (1997) J. Biol.
Chem. 272, 18421848
397. Rosenblatt, D. S. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. II (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 31113128, McGraw-
Hill, New York
398. Slieker, L. J., and Bencovic, S. J. (1984) J. Am.
Chem. Soc. 106, 18331838
399. Kallen, R. G., and Jencks, W. P. (1966) J. Biol.
Chem. 241, 5845 5850,58515863
400. Walker, J. L., and Oliver, D. J. (1986) J. Biol.
Chem. 261, 22142221
401. Kume, A., Koyata, H., Sakakibara, T.,
Ishiguro, Y., Kure, S., and Hiraga, K. (1991) J.
Biol. Chem. 266, 33233329
402. Pasternack, L. B., Laude, D. A., Jr., and
Appling, D. R. (1992) Biochemistry 31, 8713
8719
403. Kennard, O., and Salisbury, S. A. (1993) J. Biol.
Chem. 268, 1070110704
404. Zieske, L. R., and Davis, L. (1983) J. Biol. Chem.
258, 1035510359
404a. Barber, R. D., and Donohue, T. J. (1998) J. Mol.
Biol. 280, 775784
404b. Fernndez, M. R., Biosca, J. A., Torres, D.,
Crosas, B., and Pars, X. (1999) J. Biol. Chem.
274, 3786937875
405. Mejillano, M. R., Jahansouz, H., Matsunaga, T.
O., Kenyon, G. L., and Himes, R. H. (1989)
Biochemistry 28, 51365145
406. DAri, L., and Rabinowitz, J. C. (1991) J. Biol.
Chem. 266, 2395323958
406a. Shen, B. W., Dyer, D. H., Huang, J.-Y., DAri,
L., Rabinowitz, J., and Stoddard, B. L. (1999)
Protein Sci. 8, 13421349
407. Schmidt, A., Wu, H., MacKenzie, R. E., Chen,
V. J., Bewly, J. R., Ray, J. E., Toth, J. E., and
Cygler, M. (2000) Biochemistry 39, 6325 6335
407a. Pawelek, P. D., and MacKenzie, R. E. (1998)
Biochemistry 37, 11091115
408. Wahls, W. P., Song, J. M., and Smith, G. R.
(1993) J. Biol. Chem. 268, 2379223798
409. Klein, C., Chen, P., Arevalo, J. H., Stura, E. A.,
Marolewski, A., Warren, M. S., Benkovic, S. J.,
and Wilson, I. A. (1995) J. Mol. Biol. 249, 153
175
409a. Greasley, S. E., Yamashita, M. M., Cai, H.,
Benkovic, S. J., Boger, D. L., and Wilson, I. A.
(1999) Biochemistry 38, 1678316793
409b. Thoden, J. B., Firestine, S., Nixon, A.,
Benkovic, S. J., and Holden, H. M. (2000)
Biochemistry 39, 87918802
410. Caperelli, C. A., and Giroux, E. L. (1997) Arch.
Biochem. Biophys. 341, 98103
411. Schmitt, E., Blanquet, S., and Mechulam, Y.
(1996) EMBO J. 15, 47494758
329. Frey, M., Rothe, M., Wagner, A. F. V., and
Knappe, J. (1994) J. Biol. Chem. 269, 12432
12437
330. Kraus, R. J., Foster, S. J., and Ganther, H. E.
(1983) Biochemistry 22, 58535858
331. Broderick, J. B., Duderstadt, R. E., Fernandez,
D. C., Wojtuszewski, K., Henshaw, T. F., and
Johnson, M. K. (1997) J. Am. Chem. Soc. 119,
73907391
331a. Klzer, R., Pils, T., Kappl, R., Httermann, J.,
and Knappe, J. (1998) J. Biol. Chem. 273, 4897
4903
332. Wu, W., Lieder, K. W., Reed, G. H., and Frey,
P. A. (1995) Biochemistry 34, 1053210537
333. Wagner, A. F. V., Frey, M., Neugebauer, F. A.,
Schfer, W., and Knappe, J. (1992) Proc. Natl.
Acad. Sci. U.S.A. 89, 9961000
333a. Gauld, J. W., and Eriksson, L. A. (2000) J. Am.
Chem. Soc. 122, 20352040
334. Parast, C. V., Wong, K. K., Lewisch, S. A.,
Kozarich, J. W., Peisach, J., and Magliozzo, R. S.
(1995) Biochemistry 34, 23932399
335. Parast, C. V., Wong, K. K., Kozarich, J. W.,
Peisach, J., and Magliozzo, R. S. (1995)
Biochemistry 34, 57125717
335a. Reddy, S. G., Wong, K. K., Parast, C. V.,
Peisach, J., Magliozzo, R. S., and Kozarich, J.
W. (1998) Biochemistry 37, 558563
336. Mastropaolo, D., Camerman, A., and Camer-
man, N. (1980) Science 210, 334336
337. Bailey, L. B., ed. (1995) Folate in Health and
Disease, Dekker, New York
338. Curtius, H.-C., Matasovic, A., Schoedon, G.,
Kuster, T., Guibaud, P., Giudici, T., and Blau,
N. (1990) J. Biol. Chem. 265, 39233930
339. Blakley, R. L., and Benkovic, S. J., eds. (1984
1986) Folates and Pterins, Wiley, New York (3
Vols.)
340. Hadorn, E. (1962) Sci. Am. 206(April), 101110
341. Ghosh, D. K., and Stuehr, D. J. (1995) Biochem-
istry 34, 801807
342. Witteveen, C. F. B., Giovanelli, J., and Kaufman,
S. (1996) J. Biol. Chem. 271, 41434147
343. Rembold, H., and Gyure, W. L. (1972) Angew.
Chem. Int. Ed. Engl. 11, 10611072
344. Jacobson, K. B., Dorsett, D., Pfleiderer, W.,
McCloskey, J. A., Sethi, S. K., Buchanan, M. V.,
and Rubin, I. B. (1982) Biochemistry 21, 5700
5706
345. Wiederrecht, G. J., Paton, D. R., and Brown,
G. M. (1984) J. Biol. Chem. 259, 21952200
346. Cremer-Bartels, G., and Ebels, I. (1980) Proc.
Natl. Acad. Sci. U.S.A. 77, 24152418
347. Van Haastert, P. J. M., DeWitt, R. J. W.,
Grijpma, Y., and Konijn, T. M. (1982) Proc.
Natl. Acad. Sci. U.S.A. 79, 62706274
348. Irby, R. B., and Adair, J., WL. (1994) J. Biol.
Chem. 269, 2398123987
349. Stiefel, E. I. (1996) Science 272, 15991600
350. Escalante-Semerena, J. C., Rinehart, K. L., Jr.,
and Wolfe, R. S. (1984) J. Biol. Chem. 259,
94479455
351. Wolfe, R. S. (1985) Trends Biochem. Sci. 10,
396399
352. Rouvire, P. E., and Wolfe, R. S. (1988) J. Biol.
Chem. 263, 79137916
353. White, R. H. (1993) Biochemistry 32, 745753
354. Hyatt, D. C., Maley, F., and Montfort, W. R.
(1997) Biochemistry 36, 45854594
355. Blakeley, R. L. (1984) in Folates and Pterins,
Vol. I (Blakeley, P. L., and Bencovic, S. J., eds),
pp. 191253, Wiley, New York
356. Stover, P., and Schirch, V. (1993) Trends
Biochem. Sci. 18, 102106
357. Lin, B.-F., and Shane, B. (1994) J. Biol. Chem.
269, 97059713
357a. Cherest, H., Thomas, D., and Surdin-Kerjan,
Y. (2000) J. Biol. Chem. 275, 1405614063
358. Keshavjee, K., Pyne, C., and Bognar, A. L.
(1991) J. Biol. Chem. 266, 1992519929
359. Scott, J. M. (1976) Biochem. Soc. Trans. 4, 845
850
360. Brody, T., Watson, J. E., and Stokstad, E. L. R.
(1982) Biochemistry 21, 276282
361. Kim, D. W., Huang, T., Schirch, D., and
Schirch, V. (1996) Biochemistry 35, 1577215783
362. Lipman, R. S. A., Bailey, S. W., Jarrett, J. T.,
Matthews, R. G., and Jorns, M. S. (1995)
Biochemistry 34, 1121711220
363. Mathews, C. K. (1971) Bacteriophage Biochemis-
try, Van Nostrand-Reinhold, Princeton, New
Jersey
364. Roth, B. (1986) Fed. Proc. 45, 27652772
365. Schweitzer, B. I., Dicker, A. P., and Bertino, J.
R. (1990) FASEB J. 4, 24412452
366. Groom, C. R., Thillet, J., North, A. C. T., Pictet,
R., and Geddes, A. J. (1991) J. Biol. Chem. 266,
1989019893
367. Blaney, J. M., Hansch, C., Silipo, C., and
Vittonia, A. (1984) Chem. Rev. 84, 333407
368. Dale, G. E., Broger, C., DArcy, A., Hartman, P.
G., DeHoogt, R., Jolidon, S., Kompis, I.,
Labhardt, A. M., Langen, H., Locher, H., Page,
M. G. P., Stber, D., Then, R. L., Wipf, B., and
Oefner, C. (1997) J. Mol. Biol. 266, 2330
369. Sirawaraporn, W., Sathitkul, T., Sirawaraporn,
R., Yuthavong, Y., and Santi, D. V. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 11241129
370. Birdsall, B., Tendler, S. J. B., Arnold, J. R. P.,
Feeney, J., Griffin, R. J., Carr, M. D., Thomas, J.
A., Roberts, G. C. K., and Stevens, M. F. G.
(1990) Biochemistry 29, 96609667
371. Jolivet, J., Cowan, K. H., Curt, G. A., Clen-
deninn, N. J., and Chabner, B. A. (1983) N.
Engl. J. Med. 309, 10941104
372. Federspiel, N. A., Beverley, S. M., Schilling, J.
W., and Schimke, R. T. (1984) J. Biol. Chem.
259, 91279140
373. Chu, E., Takimoto, C. H., Voeller, D., Grem, J.
L., and Allegra, C. J. (1993) Biochemistry 32,
47564760
374. Friesheim, J. H., and Matthews, D. A. (1984)
in Folate Antogonists as Therapeutic Agents, Vol.
1 (Sirotnak, F. M., Burchall, J. J., Ensminger,
W. D., and Montgomery, J. A., eds), pp. 69
131, Academic Press, Orlando, Florida
375. Fan, J., Vitols, K. S., and Huennekens, F. M.
(1991) J. Biol. Chem. 266, 1486214865
376. Henderson, G. B., Hughes, T. R., and Saxena,
M. (1994) J. Biol. Chem. 269, 1338213389
377. Thompson, P. D., and Freisheim, J. H. (1991)
Biochemistry 30, 81248130
378. Chen, L., Qi, H., Korenberg, J., Garrow, T. A.,
Choi, Y.-J., and Shane, B. (1996) J. Biol. Chem.
271, 1307713087
379. Roy, K., Mitsugi, K., and Sirotnak, F. M. (1997)
J. Biol. Chem. 272, 55875593
379a. Maziarz, K. M., Monaco, H. L., Shen, F., and
Ratnam, M. (1999) J. Biol. Chem. 274, 11086
11091
380. Park, H., Zhuang, P., Nichols, R., and Howell,
E. E. (1997) J. Biol. Chem. 272, 22522258
381. Sawaya, M. R., and Kraut, J. (1997) Biochemis-
try 36, 586603
382. Reyes, V. M., Sawaya, M. R., Brown, K. A., and
Kraut, J. (1995) Biochemistry 34, 27102723
383. Lee, H., Reyes, V. M., and Kraut, J. (1996)
Biochemistry 35, 70127020
384. Verma, C. S., Caves, L. S. D., Hubbard, R. E.,
and Roberts, G. C. K. (1997) J. Mol. Biol. 266,
776796
385. Cheung, H. T. A., Birdsall, B., Frenkiel, T. A.,
Chau, D. D., and Feeney, J. (1993) Biochemistry
32, 68466854
386. Epstein, D. M., Benkovic, S. J., and Wright, P.
E. (1995) Biochemistry 34, 1103711048
References
833
412. Kruschwitz, H. L., McDonald, D., Cossins, E.
A., and Schirch, V. (1994) J. Biol. Chem. 269,
2875728763
413. Lutsenko, S., Daoud, S., and Kaplan, J. H.
(1997) J. Biol. Chem. 272, 52495255
414. Rossini, C., Attygalle, A. B., Gonzlez, A.,
Smedley, S. R., Eisner, M., Meinwald, J., and
Eisner, T. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 67926797
415. Curthoys, N. P., and Rabinowitz, J. C. (1972) J.
Biol. Chem. 247, 19651971
415a. Krupenko, S. A., and Wagner, C. (1999) J. Biol.
Chem. 274, 3577735784
416. Hardy, L. W., Graves, K. L., and Nalivaika, E.
(1995) Biochemistry 34, 84228432
417. Powers, S. G., and Snell, E. E. (1976) J. Biol.
Chem. 251, 37863793
418. Carreras, C. W., and Santi, D. V. (1995) Ann.
Rev. Biochem. 64, 721762
419. Schiffer, C. A., Clifton, I. J., Davisson, V. J.,
Santi, D. V., and Stroud, R. M. (1995) Biochem-
istry 34, 1627916287
420. Birdsall, D. L., Finer-Moore, J., and Stroud, R.
M. (1996) J. Mol. Biol. 255, 522535
421. Sage, C. R., Rutenber, E. E., Stout, T. J., and
Stroud, R. M. (1996) Biochemistry 35, 16270
16281
421a. Anderson, A. C., ONeil, R. H., DeLano, W. L.,
and Stroud, R. M. (1999) Biochemistry 38,
1382913836
422. Huang, W., and Santi, D. V. (1997) Biochemis-
try 36, 18691873
422a. Strop, P., Changchien, L., Maley, F., and
Montfort, W. R. (1997) Protein Sci. 6, 2504
2511
423. Knighton, E. R., Kan, C.-C., Howland, E.,
Janson, C. A., Hostomska, Z., Welsh, K. M.,
and Matthews, D. A. (1994) Nature Struct. Biol.
1, 186194
423a. Song, H. K., Sohn, S. H., and Suh, S. W. (1999)
EMBO J. 18, 11041113
424. Butler, M. M., Graves, K. L., and Hardy, L. W.
(1994) Biochemistry 33, 1052110526
425. Graves, K. L., and Hardy, L. W. (1994)
Biochemistry 33, 1304913056
426. Santi, D. V., and Hardy, L. W. (1987) Biochemis-
try 26, 85998606
426a. Roje, S., Wang, H., McNeil, S. D., Raymond,
R. K., Appling, D. R., Shachar-Hill, Y.,
Bohnert, H. J., and Hanson, A. D. (1999) J.
Biol. Chem. 274, 3608936096
427. Green, J. M., Ballou, D. P., and Matthews, R.
G. (1988) FASEB J. 2, 4247
428. Vanoni, M. A., Lee, S., Floss, H. G., and
Matthews, R. G. (1990) J. Am. Chem. Soc. 112,
39873992
429. Matthews, R. G. (1982) Fed. Proc. 41, 26002604
430. te Brmmelstroet, B. W., Hensgens, C. M. H.,
Keltjens, J. T., van der Drift, C., and Vogels, G.
D. (1990) J. Biol. Chem. 265, 18521857
431. Clark, J. E., and Ljungdahl, L. G. (1984) J. Biol.
Chem. 259, 1084510849
432. Zhao, S., Roberts, D. L., and Ragsdale, S. W.
(1995) Biochemistry 34, 1507515083
433. Mller, V., Blaut, M., and Gottschalk, G.
(1993) in Methano-genesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
360406, Chapman and Hall, New York
434. Ferry, J. G., ed. (1993) Methanogenesis: Ecology,
Physiology, Biochemistry and Genetics, Chap-
man & Hall, New York
435. Thauer, R. K., Hedderich, R., and Fischer, R.
(1993) in Methanogenesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
209252, Chapman and Hall, New York
436. Taylor, G. D., and Wolfe, R. S. (1974) J. Biol.
Chem. 249, 4879 4885
References
437. Keltjens, J. T., Raemakers-Franken, P. C., and
Vogels, G. D. (1993) in Microbial Growth on C1
Compounds (Murrell, J. C., and Kelly, D. P., eds),
pp. 135150, Intercept Ltd., Andover,UK
438. Leigh, J. A., Rinehart, K. L., Jr., and Wolfe, R.
S. (1985) Biochemistry 24, 995999
439. Leigh, J. A., Rinehart, K. L., Jr., and Wolfe, R.
S. (1984) J. Am. Chem. Soc. 106, 36363640
440. White, R. H. (1989) Biochemistry 28, 860865
440a. Ermler, U., Merckel, M. C., Thauer, R. K., and
Shima, S. (1997) Structure 5, 635646
440b. Shima, S., Warkentin, E., Grabarse, W., Sordel,
M., Wicke, M., Thauer, R. K., and Ermler, U.
(2000) J. Mol. Biol. 300, 935950
441. Mukhopadhyay, B., Purwantini, E., Pihl, T. D.,
Reeve, J. N., and Daniels, L. (1995) J. Biol.
Chem. 270, 28272832
441a. Allen, J. R., Clark, D. D., Krum, J. G., and
Ensign, S. A. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 84328437
442. Duine, J. A., and Frank, J. (1981) Trends
Biochem. Sci. 6, 278280
443. Ohta, S., Fujita, T., and Tobari, J. (1981) J.
Biochem. 90, 205213
444. Itoh, S., Ogino, M., Fukui, Y., Murao, H.,
Komatsu, M., Ohshiro, Y., Inoue, T., Kai, Y.,
and Kasai, N. (1993) J. Am. Chem. Soc. 115,
99609967
445. de Jong, G. A. H., Caldeira, J., Sun, J.,
Jongejan, J. A., de Vries, S., Loehr, T. M.,
Moura, I., Moura, J. J. G., and Duine, J. A.
(1995) Biochemistry 34, 94519458
445a. Keitel, T., Diehl, A., Knaute, T., Stezowski, J.
J., Hhne, W., and Grisch, H. (2000) J. Mol.
Biol. 297, 961974
446. Matsushita, K., Shinagawa, E., Adachi, O.,
and Ameyama, M. (1989) Biochemistry 28,
62766280
446a. Oubrie, A., Rozeboom, H. J., Kalk, K. H.,
Olsthoorn, A. J. J., Duine, J. A., and Dijkstra,
B. W. (1999) EMBO J. 18, 51875194
446b. Oubrie, A., Rozeboom, H. J., Kalk, K. H.,
Duine, J. A., and Dijkstra, B. W. (1999) J. Mol.
Biol. 289, 319333
446c. Elias, M. D., Tanaka, M., Izu, H., Matsushita,
K., Adachi, O., and Yamada, M. (2000) J. Biol.
Chem. 275, 73217326
446d. Dewanti, A. R., and Duine, J. A. (2000)
Biochemistry 39, 93849392
447. Xia, Z.-x, Dai, W.-w, Zhang, Y.-f, White, S. A.,
Boyd, G. D., and Mathews, F. S. (1996) J. Mol.
Biol. 259, 480501
448. White, S., Boyd, G., Mathews, F. S., Xia, Z.,
Dai, W., Zhang, Y., and Davidson, V. L. (1993)
Biochemistry 32, 1295512958
449. Ishida, T., Kawamoto, E., In, Y., Amano, T.,
Kanayama, J., Doi, M., Iwashita, T., and
Nomoto, K. (1995) J. Am. Chem. Soc. 117,
32783279
449a. Itoh, S., Kawakami, H., and Fukuzumi, S.
(1998) Biochemistry 37, 65626571
449b. Oubrie, A., Rozeboom, H. J., and Dijkstra, B.
W. (1999) Proc. Natl. Acad. Sci. U.S.A. 96,
1178711791
449c. Zheng, Y.-J., and Bruice, T. C. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 1188111886
450. McIntire, W. S. (1994) FASEB J. 8, 513521
451. Anthony, C. (1996) Biochem. J. 320, 697711
452. Klinman, J. P., and Mu, D. (1994) Ann. Rev.
Biochem. 63, 299344
453. Klinman, J. P. (1996) J. Biol. Chem. 271, 27189
27192
454. Davidson, V. L. (1992) Principles and Applica-
tions of Quinoproteins, Dekker, New York
455. Janes, S. M., Mu, D., Wemmer, D., Smith, A. J.,
Kaur, S., Maltby, D., Burlingame, A. L., and
Klinman, J. P. (1990) Science 248, 981987
456. Mu, D., Medzihradszky, K. F., Adams, G. W.,
Mayer, P., Hines, W. M., Burlingame, A. L.,
Smith, A. J., Cai, D., and Klinman, J. P. (1994)
J. Biol. Chem. 269, 99269932
457. Parsons, M. R., Convey, M. A., Wilmot, C. M.,
Yadav, K. D. S., Blakeley, V., Corner, A. S.,
Phillips, S. E. V., McPherson, M. J., and
Knowles, P. F. (1995) Structure 3, 11711184
458. Wilmot, C. M., Murray, J. M., Alton, G.,
Parsons, M. R., Convery, M. A., Blakeley, V.,
Corner, A. S., Palcic, M. M., Knowles, P. F.,
McPherson, M. J., and Phillips, S. E. V. (1997)
Biochemistry 36, 16081620
459. Monne-Loccoz, P., Nakamura, N., Steine-
bach, V., Duine, J. A., Mure, M., Klinman, J. P.,
and Sanders-Loehr, J. (1995) Biochemistry 34,
70207026
460. Parsons, M. R., Convery, M. A., Wilmot, C.
M., Yadav, K. D., Blakley, V., Corner, A. S.,
Phillips, S. E. V., McPherson, J. J., and
Knowles, P. F. (1995) Structure 3, 11711184
461. Warncke, K., Babcock, G. T., Dooley, D. M.,
McGuirl, M. A., and McCracken, J. (1994) J.
Am. Chem. Soc. 116, 40284037
462. Choi, Y.-H., Matsuzaki, R., Fukui, T., Shimizu,
E., Yorifuji, T., Sato, H., Ozaki, Y., and
Tanizawa, K. (1995) J. Biol. Chem. 270, 4712
4720
463. Choi, Y.-H., Matsuzaki, R., Suzuki, S., and
Tanizawa, K. (1996) J. Biol. Chem. 271, 22598
22603
463a. Schwartz, B., Green, E. L., Sanders-Loehr, J.,
and Klinman, J. P. (1998) Biochemistry 37,
1659116600
464. Medda, R., Padiglia, A., Pedersen, J. Z.,
Rotilio, G., Agr, A. F., and Floris, G. (1995)
Biochemistry 34, 1637516381
464a. Holt, A., Alton, G., Scaman, C. H., Loppnow,
G. R., Szpacenko, A., Svendsen, I., and Palcic,
M. M. (1998) Biochemistry 37, 4946 4957
465. Wang, S. X., Mure, M., Medzihradszky, K. F.,
Burlingame, A. L., Brown, D. E., Dooley, D.
M., Smith, A. J., Kagan, H. M., and Klinman, J.
P. (1996) Science 273, 10781084
465a. Wang, S. X., Nakamura, N., Mure, M.,
Klinman, J. P., and Sanders-Loehr, J. (1997)
J. Biol. Chem. 272, 2884128844
466. Backes, G., Davidson, V. L., Huitema, F.,
Duine, J. A., and Sanders-Loehr, J. (1991)
Biochemistry 30, 92019210
466a. Zhu, Z., and Davidson, V. L. (1998) J. Biol.
Chem. 273, 1425414260
467. McIntire, W. S., Wemmer, D. E., Chistoserdov,
A., and Lidstrom, M. E. (1991) Science 252,
817824
468. Huizinga, E. G., van Zanten, B. A. M., Duine,
J. A., Jongejan, J. A., Huitema, F., Wilson, K.
S., and Hol, W. G. J. (1992) Biochemistry 31,
97899795
469. Kuusk, V., and McIntire, W. S. (1994) J. Biol.
Chem. 269, 2613626143
470. Hyun, Y.-L., and Davidson, V. L. (1995)
Biochemistry 34, 816823
471. Chen, L., Durley, R., Poliks, B. J., Hamada, K.,
Chen, Z., Mathews, F. S., Davidson, V. L.,
Satow, Y., Huizinga, E., Vellieux, F. M. D., and
Hol, W. G. J. (1992) Biochemistry 31, 49594964
471a. Chen, L., Doi, M., Durley, R. C. E.,
Chistoserdov, A. Y., Lidstrom, M. E.,
Davidson, V. L., and Mathews, F. S. (1998) J.
Mol. Biol. 276, 131 149
472. Chen, L., Durley, R. C. E., Mathews, F. S., and
Davidson, V. L. (1994) Science 264, 8690
472a. Zhu, Z., and Davidson, V. L. (1999)
Biochemistry 38, 4862 4867
473. Mure, M., and Klinman, J. P. (1995) J. Am.
Chem. Soc. 117, 87078718
474. Lee, Y., and Sayre, L. M. (1995) J. Am. Chem.
Soc. 117, 1182311828
834 Chapter 15. Coenzymes of OxidationReduction Reactions
474a. Su, Q., and Klinman, J. P. (1998) Biochemistry
37, 1251312525
474b. Wilmot, C. M., Hajdu, J., McPherson, M. J.,
Knowles, P. F., and Phillips, S. E. V. (1999)
Science 286, 17241728
474c. Plastino, J., Green, E. L., Sanders-Loehr, J., and
Klinman, J. P. (1999) Biochemistry 38, 82048216
474d. Singh, V., Zhu, Z., Davidson, V. L., and
McCracken, J. (2000) J. Am. Chem. Soc. 122, 931
938
475. Itoh, S., Ogino, M., Haranou, S., Terasaka, T.,
Ando, T., Komatsu, M., Ohshiro, Y., Fuku-
zumi, S., Kano, K., Takagi, K., and Ikeda, T.
(1995) J. Am. Chem. Soc. 117, 1485 1493
476. Itoh, S., Kawakami, H., and Fukuzumi, S.
(1997) J. Am. Chem. Soc. 119, 439440
477. Gorren, A. C. F., Moenne-Loccoz, P., Backes,
G., de Vries, S., Sanders-Loehr, J., and Duine,
J. A. (1995) Biochemistry 34, 1292612931
478. Monne-Loccoz, P., Nakamura, N., Itoh, S.,
Fukuzumi, S., Gorren, A. C. F., Duine, J. A.,
and Sanders-Loehr, J. (1996) Biochemistry 35,
47134720
479. Pedersen, J. Z., EL-Sherbini, S., Finazzi-Agr,
A., and Rotilio, G. (1992) Biochemistry 31, 812
480. Warncke, K., Brooks, H. B., Lee, H.-i, Mc-
Cracken, J., Davidson, V. L., and Babcock, G.
T. (1995) J. Am. Chem. Soc. 117, 1006310075
481. Gorren, A. C. F., and Duine, J. A. (1994)
Biochemistry 33, 1220212209
482. Hartmann, C., Brzovic, P., and Klinman, J. P.
(1993) Biochemistry 32, 2234 2241
483. Steinebach, V., de Vries, S., and Duine, J. A.
(1996) J. Biol. Chem. 271, 55805588
484. Morton, R. A. (1971) Biol. Rev. Cambridge
Philos. Soc. 46, 4796
485. Morton, R. A. (1977) Biol. Rev. Cambridge
Philos. Soc. 46, 4796
486. Morton, R. A. (1972) Vitamins 5, 355391
487. Suzuki, H., and King, T. E. (1983) J. Biol. Chem.
258, 352358
488. Matsuma, K., Bowyer, J. R., Ohnishi, T., and
Dutton, L. P. (1983) J. Biol. Chem. 258, 15711579
489. He, D.-Y., Yu, L., and Yu, C.-A. (1994) J. Biol.
Chem. 269, 2788527888
490. He, D.-Y., Gu, L.-Q., Yu, L., and Yu, C.-A.
(1994) Biochemistry 33, 880884
491. Folkers, K., Yamamuro, Y., and Ito, K., eds.
(1980) Biomedical and Clinical Aspects of
Coenzyme Q, Elsevier, Amsterdam
492. Morton, R. A. (1972) Biochem. Soc. Symp. 35,
203217
493. Gibbs, M. (1971) Structure and Function of
Chloroplasts, Springer-Verlag, Berlin and New
York
494. Trumpower, B. L., ed. (1982) Function of
Quinones in Energy Conserving Systems,
Academic Press, New York
495. Suzuki, Y. J., Tsuchiya, M., Wassall, S. R.,
Choo, Y. M., Govil, G., Kagan, V. E., and
Packer, L. (1993) Biochemistry 32, 1069210699
496. Sun, I. L., Sun, E. E., Crane, F. L., Morr, D. J.,
Lindgren, A., and Lw, H. (1992) Proc. Natl.
Acad. Sci. U.S.A. 89, 1112611130
497. Hughes, P. E., and Tove, S. B. (1980) J. Biol.
Chem. 255, 70957097
498. Valgimigli, L., Banks, J. T., Ingold, K. U., and
Lusztyk, J. (1995) J. Am. Chem. Soc. 117, 9966
9971
499. Nagaoka, S.-i, and Ishihara, K. (1996) J. Am.
Chem. Soc. 118, 73617366
500. van Belzen, R., Kotlyar, A. B., Moon, N.,
Dunham, R. D., and Albracht, S. P. J. (1997)
Biochemistry 36, 886893
501. Salerno, J. C., Osgood, M., Liu, Y., Taylor, H.,
and Scholes, C. P. (1990) Biochemistry 29,
69876993
References
502. Swallow, A. J. (1982) in Function of Quinones in
Energy Conserving Systems (Trumpower, B. L.,
ed), pp. 5971, Academic Press, New York
503. McTigue, J. J., Dhaon, M. K., Rich, D. H., and
Suttie, J. W. (1984) J. Biol. Chem. 259, 42724278
504. Suttie, J. W. (1993) FASEB J. 7, 445452
505. Morris, D. P., Soute, B. A. M., Vermeer, C., and
Stafford, D. W. (1993) J. Biol. Chem. 268, 8735
8742
506. Lingenfelter, S. E., and Berkner, K. L. (1996)
Biochemistry 35, 82348243
507. Kuliopulos, A., Hubbard, B. R., Lam, Z.,
Koski, I. J., Furie, B., Furie, B. C., and Walsh,
C. T. (1992) Biochemistry 31, 77227728
507a. Wu, S.-M., Mutucumarana, V. P., Geromanos,
S., and Stafford, D. W. (1997) J. Biol. Chem. 272,
1171811722
508. Wood, G. M., and Suttie, J. W. (1988) J. Biol.
Chem. 263, 32343239
509. Metzler, D. E. (1977) Biochemistry. The Chemical
Reactions of Living Cells, Academic Press, New
York (p. 581)
510. Sadowski, J. A., Esmon, C. T., and Suttie, J. W.
(1976) J. Biol. Chem. 251, 27702776
511. Anton, D. L., and Friedman, P. A. (1983) J.
Biol. Chem. 258, 1408414087
512. Naganathan, S., Hershline, R., Ham, S. W.,
and Dowd, P. (1994) J. Am. Chem. Soc. 116,
98319839
513. Dowd, P., Hershline, R., Ham, S. W., and
Naganathan, S. (1995) Science 269, 16841691
514. Bouchard, B. A., Furie, B., and Furie, B. C.
(1999) Biochemistry 38, 9517 9523
514a. Zheng, Y.-J., and Bruice, T. C. (1998) J. Am.
Chem. Soc. 120, 16231624
515. Dubois, J., Gaudry, M., Bory, S., Azerad, R.,
and Marquet, A. (1983) J. Biol. Chem. 258,
78977899
516. Dubois, J., Dugave, C., Fours, C., Kaminsky,
M., Tabet, J.-C., Bory, S., Gaudry, M., and
Marquet, A. (1991) Biochemistry 30, 10506
10512
517. Decottignies-Le Marchal, P., Ducrocq, C.,
Marquet, A., and Azerad, R. (1984) J. Biol.
Chem. 259, 1501015012
518. Lee, J. J., Principe, L. M., and Fasco, M. J.
(1985) Biochemistry 24, 70637070
519. Hildebrandt, E. F., and Suttie, J. W. (1982)
Biochemistry 21, 24062411
520. Fasco, M. J., Preusch, P. C., Hildebrandt, E.,
and Suttie, J. W. (1983) J. Biol. Chem. 258,
43724380
521. Esmon, C. T. (1989) J. Biol. Chem. 264, 4743
4746
522. Stenflo, J., hlin, A.-K., Owen, W. G., and
Schneider, W. J. (1988) J. Biol. Chem. 263, 2124
523. Pryor, W. A. (1982) Ann. N.Y. Acad. Sci. 393,
122
524. Ham, A.-J. L., and Liebler, D. C. (1995)
Biochemistry 34, 5754 5761
525. Valgimigli, L., Ingold, K. U., and Lusztyk, J.
(1996) J. Am. Chem. Soc. 118, 35453549
526. Christen, S., Woodall, A. A., Shigenaga, M. K.,
Southwell-Keely, P. T., Duncan, M. W., and
Ames, B. N. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 32173222
527. Schwarz, K., and Foltz, C. M. (1957) J. Am.
Chem. Soc. 79, 32923293
528. Shamberger, R. J. (1983) Biochemistry of
Selenium, Plenum, New York
529. Keating, K. I., and Dagbusan, B. C. (1984)
Proc. Natl. Acad. Sci. U.S.A. 81, 34333437
530. Stadtman, T. C. (1991) J. Biol. Chem. 266,
1625716260
531. Stadtman, T. C. (1996) Ann. Rev. Biochem. 65,
83100
532. Burk, R. F. (1991) FASEB J. 5, 22742279
533. Low, S. C., and Berry, M. J. (1996) Trends
Biochem. Sci. 21, 203208
534. Ren, B., Huang, W., kesson, B., and Laden-
stein, R. (1997) J. Mol. Biol. 268, 869 885
535. Ladenstein, R., and Wendel, A. (1983) Eur. J.
Biochem. 133, 5169
536. Shen, Q., Chu, F.-F., and Newburger, P. E.
(1993) J. Biol. Chem. 268, 1146311469
537. Chu, F.-F., Doroshow, J. H., and Esworthy, R.
S. (1993) J. Biol. Chem. 268, 25712576
538. Duan, Y.-J., Komura, S., Fiszer-Szafarz, B.,
Szafarz, D., and Yagi, K. (1988) J. Biol. Chem.
263, 1900319008
539. Schnurr, K., Belkner, J., Ursini, F., Schewe, T.,
and Khn, H. (1996) J. Biol. Chem. 271, 4653
4658
540. Friedman, P. A. (1984) N. Engl. J. Med. 310,
14581460
540a. Shisler, J. L., Senkevich, T. G., Berry, M. J., and
Moss, B. (1998) Science 279, 102105
541. Visser, T. J. (1980) Trends Biochem. Sci. 5, 222
224
542. Toyoda, N., Harney, J. W., Berry, M. J., and
Larsen, P. R. (1994) J. Biol. Chem. 269, 20329
20334
543. DePalo, D., Kinlaw, W. B., Zhao, C., Engel-
berg-Kulka, H., and St. Germain, D. L. (1994)
J. Biol. Chem. 269, 1622316228
544. Davey, J. C., Becker, K. B., Schneider, M. J., St.
Germain, D. L., and Galton, V. A. (1995) J.
Biol. Chem. 270, 2678626789
545. Croteau, W., Whittemore, S. L., Schneider, M.
J., and St. Germain, D. L. (1995) J. Biol. Chem.
270, 1656916575
546. St. Germain, D. L., Schwartzman, R. A.,
Groteau, W., Kanamori, A., Wang, Z., Brown,
D. D., and Galton, V. A. (1994) Proc. Natl.
Acad. Sci. U.S.A. 91, 77677771
547. Tamura, T., and Stadtman, T. C. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 10061011
548. Gladyshev, V. N., Jeang, K.-T., and Stadtman,
T. C. (1996) Proc. Natl. Acad. Sci. U.S.A. 93,
61466151
548a. Sun, Q.-A., Wu, Y., Zappacosta, F., Jeang,
K.-T., Lee, B. J., Hatfield, D. L., and
Gladyshev, V. N. (1999) J. Biol. Chem. 274,
2452224530
549. Marcocci, L., Floh, L., and Packer, L. (1997)
BioFactors 6, 351358
550. Hill, K. E., Lloyd, R. S., Yang, J.-G., Read, R.,
and Burk, R. F. (1991) J. Biol. Chem. 266,
1005010053
551. Himeno, S., Chittum, H. S., and Burk, R. F.
(1996) J. Biol. Chem. 271, 1576915775
552. Steinert, P., Ahrens, M., Gross, G., and Floh, L.
(1997) BioFactors 6, 311319
552a. Saito, Y., Hayashi, T., Tanaka, A., Watanabe,
Y., Suzuki, M., Saito, E., and Takahashi, K.
(1999) J. Biol. Chem. 274, 28662871
553. Read, R., Bellew, T., Yang, J.-G., Hill, K. E.,
Palmer, I. S., and Burk, R. F. (1990) J. Biol.
Chem. 265, 1789917905
554. Vendeland, S. C., Beilstein, M. A., Chen, C. L.,
Jensen, O. N., Barofsky, E., and Whanger, P. D.
(1993) J. Biol. Chem. 268, 1710317107
555. Vendeland, S. C., Beilstein, M. A., Yeh, J.-Y.,
Ream, W., and Whanger, P. D. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 87498753
556. Yeh, J.-Y., Beilstein, M. A., Andrews, J. S., and
Whanger, P. D. (1995) FASEB J. 9, 392396
557. Calvin, H. I., Cooper, G. W., and Wallace, E.
(1981) Gamete Research 4, 139149
558. Shamberger, R. J. (1983) Biochemistry of
Selenium, Plenum, New York (pp.4749)
559. Yamazaki, S. (1982) J. Biol. Chem. 257, 7926
7929
560. Yamazaki, S., Tsai, L., Stadtman, T. C.,
Teshima, T., and Nakaji, A. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 13641366
561. Berg, B. L., Baron, C., and Stewart, V. (1991) J.
Biol. Chem. 266, 2238622391
835
Study Questions
1. S-adenosylmethionine is also a biological methyl
group donor. The product of its methyl transferase
reactions is S-adenosylhomocysteine. This product
is further degraded by S-adenosylhomocysteine
hydrolase, an enzyme that contains tightly bound
NAD
+
, to form homocysteine and adenosine.
Write a step-by-step mechanism for the action of
this hydrolase.
2. Compare the chemical mechanisms of enzyme-
catalyzed decarboxylation of the following:
a) a -oxo-acid such as acetoacetate or oxalo-
acetate
b) an -oxo-acid such as pyruvate
c) an amino acid such as L-glutamate
3. Describe the subunit structure of the enzyme
pyruvate dehydrogenase. Discuss the functioning
of each of the coenzymes that are associated with
these subunits and write detailed mechanisms for
each step in the pyruvate dehydrogenase reaction.
4. Free formate can be assimilated by cells via the
intermediate 10-formyl-tetrahydrofolate (10-
formyl-THF).
a) Describe the mechanism of synthesis of this
compound from formate and tetrahydrofolate.
b) Diagram a hypothetical transition state for the
first step of this reaction sequence.
c) Describe two or more uses that the human
body makes of 10-formyl-THF.
5. Using partial structural formulas, describe the
reactions by which serine and methionine react to
formN-formylmethionine needed for protein
synthesis.
6. Write the equations for each of the reactions shown
below. Using the E
0'
values below, calculate approxi-
mate Gibbs energies for each reaction, and show
by the relative length of the arrows on which side
of the reaction the equilibrium lies.
a) The oxidation of malate by NAD
+
b) The oxidation of succinate by NAD
+
c) The oxidation of succinate by enzyme-bound
FAD
d) What can you say about the cofactor required
for oxidation of succinate from your calcula-
tions?
The values of E
0'
for several half reactions are
given below. Everything has been rounded to one
significant figure so that a calculator is unnecessary.
Reaction E
0'
(volts)
NAD
+
+ H
+
+ 2 e

NADH 0.3
enzyme bound FAD + 2 H
+
+ 2 e

enzyme bound FADH


2
0.0
fumarate + 2 H
+
+ 2 e

succinate 0.0
oxalacetate + 2 H
+
+ 2 e

malate 0.2
7. Some acetogenic bacteria, which convert CO
2
to
acetic acid, form pyruvate for synthesis of carbo-
hydrates, etc., by formation of formaldehyde and
conversion of the latter to glycine by reversal of
the PLP and lipoic acid-dependent glycine decar-
boxylase, a 4-protein system. The glycine is then
converted to serine, pyruvate, oxaloacetate, etc.
Propose a detailed pathway for this sequence.
References
562. Gladyshev, V. N., Boyington, J. C., Khangulov,
S. V., Grahame, D. A., Stadtman, T. C., and
Sun, P. D. (1996) J. Biol. Chem. 271, 80958100
563. Boyington, J. C., Gladyshev, V. N., Khangulov,
S. V., Stadtman, T. C., and Sun, P. D. (1997)
Science 275, 13051306
564. Arkowitz, R. A., and Abeles, R. H. (1990) J.
Am. Chem. Soc. 112, 870872
565. Arkowitz, R. A., and Abeles, R. H. (1991)
Biochemistry 30, 40904097
566. Stadtman, T. C., and Davis, J. N. (1991) J. Biol.
Chem. 266, 2214722153
567. Arkowitz, R. A., and Abeles, R. H. (1989)
Biochemistry 28, 4639 4644
567a. Self, W. T., and Stadtman, T. C. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 72087213
568. Gladyshev, V. N., Khangulov, S. V., and
Stadtman, T. C. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 232236
569. Sliwkowski, M. X., and Stadtman, T. C. (1985)
J. Biol. Chem. 260, 31403144
570. Veres, Z., Tsai, L., Scholz, T. D., Politino, M.,
Balaban, R. S., and Stadtman, T. C. (1992)
Proc. Natl. Acad. Sci. U.S.A. 89, 29752979
571. Diamond, A. M., Choi, I. S., Crain, P. F.,
Hashizume, T., Pomerantz, S. C., Cruz, R.,
Steer, C. J., Hill, K. E., Burk, R. F., McCloskey,
J. A., and Hatfield, D. L. (1993) J. Biol. Chem.
268, 1421514223
572. Diplock, A. T., and Lucy, J. A. (1973) FEBS
Lett. 29, 205210
573. Sandholm, M., and Sipponen, P. (1973) Arch.
Biochem. Biophys. 155, 120124
574. Sarathchandra, S. V., and Watkinson, J. H.
(1981) Science 211, 600601
575. Reamer, D. C., and Zoller, W. H. (1980) Science
208, 500502
576. Mozier, N. M., McConnell, K. P., and Hoffman,
J. L. (1988) J. Biol. Chem. 263, 45274531
577. Harrison, D. J., Fluri, K., Seiler, K., Fan, Z.,
Effenhauser, C. S., and Manz, A. (1993) Science
261, 895897
578. Kromayer, M., Wilting, R., Tormay, P., and
Bck, A. (1996) J. Mol. Biol. 262, 413420
579. Glass, R. S., Singh, W. P., Jung, W., Veres, Z.,
Scholz, T. D., and Stadtman, T. C. (1993)
Biochemistry 32, 1255512559
580. Veres, Z., Kim, I. Y., Scholz, T. D., and Stadtman,
T. C. (1994) J. Biol. Chem. 269, 1059710603
581. Mullins, L. S., Hong, S.-B., Gibson, G. E.,
Walker, H., Stadtman, T. C., and Raushel, F.
M. (1997) J. Am. Chem. Soc. 119, 66846685
836 Chapter 16. Transition Metals in Catalysis and Electron Transport
Contents
A. Iron
1. Uptake by Living Cells
Siderophores
Uptake of iron by eukaryotic cells
Transferrins
2. Storage of Iron
3. Heme Proteins
Some names to remember
Hemes and heme proteins
4. The Cytochromes
The c-type cytochromes
Cytochromes b, a, and o
5. Mechanisms of Biological Electron Transfer
Electron-transfer pathways?
Docking
Coupling and gating of electron transfer
Effects of ionic equilibria on electron transfer
6. Reactions of Heme Proteins with Oxygen or
Hydrogen Peroxide
Oxygen-carrying proteins
Catalases and peroxidases
Mechanisms of catalase and peroxidase catalysis
Haloperoxidases
7. The IronSulfur Proteins
Ferredoxins, high-potential iron proteins,
and rubredoxins
Rubredoxins
Chloroplast-type ferredoxins
The 3Fe 4S clusters
Properties of ironsulfur clusters
Functions of ironsulfur enzymes
8. The (-oxo) Diiron Proteins
Hemerythrin
Purple acid phosphatases
Diiron oxygenases and desaturases
9. Ribonucleotide Reductases
10. Superoxide Dismutases
B. Cobalt and Vitamin B
12
1. Coenzyme Forms
2. Reduction of Cyanocobalamin and Synthesis
of Alkyl Cobalamins
3. Three Nonenzymatic Cleavage Reactions of
Vitamin B
12
Coenzymes
4. Enzymatic Functions of B
12
Coenzymes
Cobalamin-dependent ribonucleotide reductase
The isomerization reactions
Stereochemistry of the isomerization reactions
Aminomutases
Transfer reactions of methyl groups
Corrinoid-dependent synthesis of acetyl-CoA
837
837
838
839
840
841
843
843
844
845
846
847
848
848
849
849
850
850
850
852
853
855
857
857
858
859
859
859
860
862
862
862
863
863
863
866
867
869
870
870
871
872
874
874
875
876
877
877
878
879
881
881
882
883
884
884
885
885
887
887
888
889
890
890
892
893
893
895
903
842
868
871
Boxes
Box 16-A Magnetic Iron Oxide in Organisms
Box 16-B Cobalamin (Vitamin B
12
)
Tables
Table 16-1 Isomerization Reactions Involving
Hydrogen Transfer and Dependent
upon a Vitamin B
12
Coenzyme
C. Nickel
1. Urease
2. Hydrogenases
3. Cofactor F
430
and Methyl-Coenzyme M
Reductase
4. Tunichlorins
5. Carbon Monoxide Dehydrogenases and
Carbon Monoxide Dehydrogenase/Acetyl-CoA
Synthase
D. Copper
1. Electron-Transferring Copper Proteins
2. Copper, Zinc-Superoxide Dismutase
3. Nitrite and Nitrous Oxide Reductases
4. Hemocyanins
5. Copper Oxidases
6. Cytochrome c Oxidase
E. Manganese
F. Chromium
G. Vanadium
H. Molybdenum
1. Molybdenum Ions and Coenzyme Forms
2. Enzymatic Mechanisms
3. Nutritional Need for Mo
I. Tungsten
References
Study Questions
Transition metal ions function in many biological oxidation-reduction processes. (Left) A
heme ring in nitrite reductase chelates an iron ion in its center, holding the Fe
2+
or Fe
3+
with
bonds from four nitrogen atoms and a fifth bond from an imidazole ring below. Above the
iron is a nitrite ion, NO
2

, awaiting reduction to nitric oxide NO in a denitrifying bacterium.


From Ranghino et al. (2000) Biochemistry 39, 1095810966. (Right) The active site of a bacterial
dimethylsulfoxide reductase has an atom of molybdenum or tungsten at its center. The
metal is held by four sulfur atoms from two molybdopterin molecules and an oxygen atom
of a serine side chain. Two other oxygen atoms are bound as oxo groups and may participate
in catalysis. From Stewart et al. (2000) J. Mol. Biol. 299, 593 600.
Tyr114
Ser147
Trp116
O
N, C, P
S
Mo
Asp 346
His 345
His 200
837
(16-1a)
(16-1b)
Transition Metals in Catalysis and Electron Transport
16
bacteria, possess no oxygen-requiring enzymes and are
almost devoid of both iron and copper. All other
organisms appear to require iron for life.
1. Uptake by Living Cells
A major problem for cells is posed by the relative
insolubility of ferric hydroxide and other compounds
from which iron must be extracted by the organism. A
consequence is that iron is often taken up in a chelated
form and is transferred from one organic ligand, often
a protein, to another with little or no existence as free
Fe
3+
or Fe
2+
. As can be calculated from the estimated
solubility product of Fe(OH)
3
(Eq. 16-1),
7
the equilibrium
concentration of Fe
3+
at pH 7 is only 10
17
M.
K
sp
= [Fe
3+
][OH

]
3
< 10
38
M
4
or [Fe
3+
] / [H
+
]
3
< 10
4
M
4
at 25 C
For a 2 m
3
bacterial cell this amounts to just one free
Fe
3+
ion in almost 100 million cells at any single moment.
The importance of chelated forms of iron becomes
obvious. It is also evident from Eq. 16-1 that, in addi-
tion to chelation, a low external pH can also facilitate
uptake of Fe
3+
by organisms.
The values of the formation constants for chelates
of Fe
2+
typically lie between those of Mn
2+
and Co
2+
(Fig. 6-6, Table 6-9). For example, K
1
= 10
14.3
M
1
for
formation of the Fe
2+
chelate of EDTA. The smaller
and more highly charged Fe
3+
is bonded more strongly
(K
1
= 10
25
M
1
). These binding constants are indepen-
dent of pH. However, the binding of any metal ion is
affected by pH, as discussed in Chapter 6. A fact of
Tyr114
Ser147
Trp116
O
N, C, P
S
Mo
Asp 346
His 345
His 200
Although the amounts present within living cells
are very small, the ions of the transition metals Fe, Co,
Ni, Cu, and Mn are extremely active centers for catalysis,
especially of reactions that take advantage of the
ability of these metals to exist in more than one oxida-
tion state.
1 4
Iron, copper, and nickel are also compo-
nents of the electron carrier proteins that function as
oxidants or reductants in many biochemical processes.
These metals are all nutritionally essential, as are
chromium and vanadium. Among the heavier transi-
tion elements molybdenum is a constituent of an
important group of enzymes that includes the sulfite
oxidase of human liver and nitrogenase of nitrogen-
fixing bacteria. Tungsten occasionally substitutes for
molybdenum.
A. Iron
Iron is one of the most abundant elements in the
earths crust, being present to the extent of ~ 4% in a
typical soil. Its functions in living cells are numerous
and diverse.
2,5 8
The average overall iron content of
both bacteria and fungi is ~ 1 mmol / kg, but that of
animal tissues is usually less. Seventy percent of the
3 5 g of iron present in the human body is located in
the bloods erythrocytes, whose overall iron content is
~ 20 mM. In other tissues the total iron averages closer to
0.3 mM and consists principally of storage forms. The
total concentration of iron in all of the iron-containing
enzymes of tissues amounts to only about 0.01 mM.
Although these concentrations are low, the iron is
concentrated in oxidative enzymes of membranes and
may attain much higher concentrations locally. Only a
few parasitic or anaerobic bacteria, e.g. the lactic acid
838 Chapter 16. Transition Metals in Catalysis and Electron Transport
N N
N
OH
O
O
O N
R
OH
CH
3
H H
O
OH
OH
OH
H
H
considerable biochemical significance is the stronger
binding of Fe
3+
to oxygen-containing ligands than to
nitrogen atoms, while Fe
2+
tends to bind preferentially
to nitrogen. It is also significant that Fe
3+
bound to
oxygen ligands tends to exchange readily with other
ferric ions in the medium, whereas Fe
3+
bound to
nitrogen-containing ligands such as heme exchanges
slowly. This fact is important for both iron-transport
compounds and enzymes.
Siderophores. If a suitably high content of iron
(e.g., 50 M or more for E. coli) is maintained in the
external medium, bacteria and other microorganisms
have little problem with uptake of iron. However,
when the external iron concentration is low, special
compounds called siderophores are utilized to render
the iron more soluble.
7 11c
For example, at iron con-
centrations below 2 M, E. coli and other enterobacte-
ria secrete large amounts of enterobactin (Fig. 16-1).
The stable Fe
3+
enterobactin complex is taken up by a
transport system that involves receptors on the outer
bacterial membrane.
9,12,13
Siderophores from many
bacteria have in common with enterobactin the pres-
ence of catechol (ortho-dihydroxybenzene) groups
that chelate the iron.
The three catechol groups of enterobactin are carried
on a cyclic serine triester structure. A variety of both
cyclic and linear structures are found among other
catechol siderophores.
14 19
For example, parabactin
and agrobactin (Fig. 16-1) contain a backbone of
spermidine
20
(Chapter 24). After the Fe
3+
enterobactin
complex enters a bacterial cell the ester linkages of a
siderophore are cleaved by an esterase. Because of the
extremely high formation constant of ~ 10
52
M
1
for
the complex
11
the only way for a cell to release the
Fe
3+
is through this irreversible destruction of the iron
carrier.
11d
Reduction to Fe
2+
may be involved in release
of iron from some siderophores.
11e
The first known siderophore, isolated in 1952 by
Neilands,
22
is ferrichrome (Fig. 16-1), a cyclic hexa-
peptide containing hydroxamate groups at the iron-
binding centers. Oxygen atoms form the bonds to iron
C
H
C
N
H
3
C
C
C
N
C
N
N
C
C
N
H
O
O
H
H
O
H
H
3
C C
N
O O
O
O
C
H
C
O
N
H
H
C
H
C
O
N
H
C
H
H
C
O
H
H
H
N
C
CH
3
Fe
3+
O

Figure 16-1 Structures of several siderophores and of their metal


complexes. (A) Enterobactin of E. coli and other enteric bacteria;
12
(B) parabactin (R = H) from Paracoccus denitrificans and agrobactin (R = OH)
from Agrobacterium tumefaciens;
20
(C) ferrichrome;
9
(D) pyochelin from
Pseudomonas aeruginosa.
21
CO
HN
NH
CO
CO
O
O
O
O
O
O O
O
O
H
HN
O O
H
H
O
Fe
3
OH
N
S
N
S
CH
3
COO

O
C
R N
O
R'

Hydroxamate group
C D
A B
839 A. Iron
in this compound also. Ferrichrome binds Fe
3+
with a
formation constant of ~ 10
29
M
1
. The binding is not as
tight as with enterobactin and the iron can be released
by enzymatic reduction to Fe
2+
which is much less
tightly bound than is Fe
3+
. The released ferrichrome
can be secreted and used repeatedly to bring in more
iron. Ferrichrome is produced by various fungi and
bacilli and is only one of a series of known hydroxa-
mate siderophores.
16
Since iron is essential to virtually
all parasitic organisms the ability to obtain iron is often
the limiting factor in establishing an infection.
23,24
In E. coli there are seven different outer membrane
receptors for siderophores.
25
One of these, the gated
porin FepA, is specific for ferric enterochelin. With
the assistance of another protein, TonB, it allows the
ferric siderophore to penetrate the outer membrane.
26
A different receptor, FhuA, binds ferrichrome. Both
FepA and FhaA are large 22-strand porins resembling
the 16-strand porin shown in Fig. 8-20. However, they
are nearly 7 nm long with internal diameters three
times those of the 16-strand porins. In addition, loops
of polypeptide chain on the outer edges can close
while an N-terminal domain forms a cork that remains
in place until the Fe
3+
-siderophore complex enter the
channel and binds. Like the hatches in an air lock on
a spacecraft, the outer loops then close, after which the
inner cork unwinds to allow the siderophore complex
to enter the periplasmic space. The apparatus requires
an energy supply, which apparently is provided by an
additional complex consisting of proteins TonB, ExbA,
and ExbD. They evidently couple the electrochemical
gradient across the cell membrane (Chapter 8, B1 and
C5 and Chapter 18) with the operation of the channel
gates. Some bacteria use a different strategy for passage
across the outer membrane.
11c
The channel of a receptor
protein contains a molecule of an iron-free siderophore.
When a molecule of Fe
3+
-siderophore binds in the
outside part of the channel the Fe
3+
jumps to the inner
siderophore, which then dissociates from the receptor,
carrying the Fe
3+
-siderophore complex into the peri-
plasmic space. This mechanism also seems to be
available in E. coli. The ferric uptake regulation (Fur)
protein binds excess free Fe
2+
, the resulting complex
acting as a repressor of all of the iron uptake genes in
E. coli.
11e
Additional proteins are required for passage
through the membrane.
11b,23,25,27
These are ABC trans-
porters, which utilize hydrolysis of ATP as an energy
source (Chapter 8, Section C,4). For uptake of the Fe
3+
-
ferrichrome complex protein FhuD is the periplasmic
binding protein, FhuC is an integral membrane com-
ponent, and the cytosolic FhuC contains the ATPase
center.
11b
Another ABC transporter, found in many
bacteria, carries unchelated Fe
3+
across the inner mem-
brane. The binding protein component for Hemophilus
influenzae, designated Hit, resembles one lobe of mam-
malian transferrin (Fig. 16-2).
11b
The siderophore
receptors of bacteria have been parasitized by vari-
ous bacteriophages and toxic proteins. For example,
FepA is also a receptor for the toxic colicins B and D
(Box 8-D) and tonB is a receptor for bacteriophage
T1.
9,13
Some bacteria do not form siderophores but take
up Fe
2+
. Even E. coli, when grown anaerobically,
synthesizes an uptake system for Fe
2+
. It utilizes a
75-residue peptide encoded by gene feoA and a 773-
residue protein encoded by feoB.
28,11b
Uptake of iron by eukaryotic cells. The yeast
Saccharomyces cerevisiae utilizes two systems for uptake
of iron.
29,30
A low-affinity system transports Fe
2+
with
an apparent K
m
of ~ 30 M, while a high-affinity system
has a K
m
of ~ 0.15 M. Study of these systems has been
greatly assisted by the use of genetic methods devel-
oped for both bacteria and yeast (Chapter 26). The
low-affinity iron uptake depends upon a protein trans-
porter encoded by gene FET4 and on a reductase
encoded by genes FRE1 and FRE2, proteins that are
embedded in the cytoplasmic membrane.
29,31-33
It
might seem reasonable that the FET3 copper oxido-
reductase should keep Fe
2+
reduced while it is trans-
ported. However, it appears to oxidize Fe
2+
to Fe
3+
.
The high-affinity uptake system is more puzzling. It
requires a permease encoded by FTR I and an additional
protein encoded by FET3.
30,33 35
The Fet3 protein is a
copper oxidoreductase related to ceruloplasmin
(Section D). The protein Fre1p (encoded by FRE1) is a
metalloreductase that reduces Cu
2+
to Cu
+
, as well as
Fe
3+
to Fe
2+
. It is essential for copper uptake (Section
D).
33
It has long been known that ceruloplasmin is
required for mobilization of iron from mammalian
tissues.
30
Hereditary ceruloplasmin deficiency causes
accumulation of iron in tissues.
36
Yeast also contains
both mitochondrial and vacuolar iron transporters.
37,37a,b
The uptake of iron by animals is not as well under-
stood
38 40
but it resembles that of yeast.
41
A general
divalent cation transporter that is coupled to the mem-
brane proton gradient is involved in intestinal iron
uptake.
42,60
Ascorbic acid promotes the uptake of iron,
presumably by reducing it to Fe(II), which is more
readily absorbed than Fe(III), and also by promoting
ferritin synthesis.
43
Uptake is also promoted by meat
in the diet.
44
Within the body iron is probably trans-
ferred from one protein to another with only a transient
existence as free Fe
2+
. An average daily human diet
contains ~ 15 mg of iron, of which ~ 1 mg is absorbed.
This is usually enough to compensate for the small
losses of the metal from the body, principally through
the bile. Once it enters the body, iron is carefully con-
served. The 9 billion red blood cells destroyed daily
yield 20 25 mg of iron which is almost all reused or
stored. The body apparently has no mechanism for
excretion of large amounts of iron; a persons iron
content is regulated almost entirely by the rate of
840 Chapter 16. Transition Metals in Catalysis and Electron Transport
uptake. This rate is increased during pregnancy and,
in young women, to compensate for iron lost in men-
strual bleeding. Nevertheless, control of iron uptake is
imperfect and perhaps 500 million people around the
world suffer from iron deficiency.
44,45
For others, an
excessive intake of iron or a genetic defect lead to
accumulation of iron to toxic levels, a condition called
hemochromatosis.
44,46,46a,b
This condition may also
arise in any disease that leads to excessive destruction
of hemoglobin or accumulation of
damaged erythrocytes. Examples
are -thalassemia (Chapter 28) and
cerebral malaria.
47
Treatment with
chelating agents designed to remove
iron is often employed.
47,48
Transferrins. Within the body
iron is moved from one location to
another while bound as Fe
3+
to
transferrins, a family of related
680 - to 700-residue 80-kDa pro-
teins.
38,49 53
Each transferrin mole-
cule contains two Fe
+3
binding sites,
one located in each of two similar
domains of the folded peptide
chain. A dianion, usually CO
3
2
,
is bound together with each Fe
3+
.
Milk transferrin (lactoferrin
51a,b,c
also found in leukocytes), hen egg
transferrin (ovotransferrin),
52,52a
and rabbit and human serum
transferrin
54,54a
all have similar
structures. Each Fe
3+
is bonded to
oxygen anions from two tyrosine
side chains, an aspartate carboxy-
late, an imidazole group, and the
bound carbonate ion (Fig. 16-2B).
Transferrin of blood plasma is
encoded by a separate gene but has
a similar structure. Transferrin of
chickens appears to be identical to
conalbumin of egg whites. The
iron-binding proteins of body fluids
are sometimes given the group
name siderophilins. Transferrins
may function not only in transport
of iron throughout the body but
also as iron buffers that provide a
relatively constant iron concentra-
tion within tissues.
The entrance of iron into the
body through the intestinal mucosal
cells may involve the transferrin
present in those cells
44
and the
influx of iron may also be regulated
by blood plasma transferrin. There
is also a nontransferrin pathway.
42,55
Transferrins bind Fe
2+
weakly and it is likely that a
transferrin Fe
2+
HCO
3

complex formed initially


undergoes oxidation to the Fe
3+
CO
3
2
complex within
cells and within the bloodstream. A conformational
change closes the protein around the iron ions.
56
In
yeast the previously mentioned copper oxidoreductase
encoded by the FET3 gene appears to not only oxidize
Fe
2+
but also transfer the resulting Fe
3+
to transferrin.
Ceruloplasmin may play a similar role in mammals.
33
Figure 16-2 (A) Ribbon drawing of the polypeptide chain of a transferrin,
human lactoferrin. The N lobe is to the left and the C lobe to the right.
Each active site contains bound Fe
3+
and a molecule of oxalate dianion
which replaces the physiological CO
3
2
. From Baker et al.
51
Courtesy of
Edward Baker. (B) Schematic diagram showing part of the hydrogen-bond
network involved in binding the Fe
3+
in the N lobe of hen ovotransferrin.
Some side chain groups and water molecules have been omitted. The
positions of hydrogen atoms and the charge state of acidbase groups are
uncertain. Most of the hydrogen-bond distances (O - - O, O - - N, N - - N)
indicated by dashed lines are between 0.27 and 0.3 nm. Release of the
bound Fe
3+
may be accomplished in part by protonation of the bound
CO
3
2
to form HCO
3

. See Kurokawa et al.


52
80
O
O

82
O
H
H
H
O
H
N
N
Fe
3+
92
O

301
N
H H
H
+
H
H
N
209
191
N
H
H
N
H
H
H
N
+
121
O
117
124
O
O
N
H
H N
122
123 O

H
O

O
H
H
N
O O
62
61
N
H
H
N
60
250

A
B
841
Iron is transferred from the
plasma transferrin into cells of the
body following binding of the Fe
3+

transferrin complex to specific


receptors. The surface of an imma-
ture red blood cell (reticulocyte)
may contain 300,000 transferrin
receptors, each capable of catalyzing
the entry of ~ 36 iron ions per hour.
38
The receptor is a 180-kDa dimeric
glycoprotein. When the Fe
3+

transferrin complex is bound, the


receptors aggregate in coated pits
and are internalized. The mecha-
nism of release of the Fe
3+
may
occur by different mechanisms in
the two lobes.
57
The pH of the
endocytic vesicles containing the
receptor complex is probably low-
ered to ~ 5.6 as in lysosomes. This
may protonate the bound CO
3
2
in
the complex
51c,54a,58
and assists in
the release of the Fe
3+
, possibly after
reduction to Fe
2+
. Both the apo-
transferrin and its receptor are
returned to the cell surface for reuse,
the apotransferrin being released
into the blood. Chelating agents
such as pyrophosphate, ATP, and
citrate as well as simple anions
59
may also assist in removal of iron
from transferrin. The same trans-
membrane transporter that is
involved in intestinal iron uptake
42
is needed to remove iron from the
endosome after release.
60
2. Storage of Iron
Within tissues of animals,
plants, and fungi much of the iron is
packaged into the red-brown water-
soluble protein ferritin, which
stores Fe(III) in a soluble, nontoxic,
and readily available form.
61 64
Although bacteria store very little
iron,
65
some of them also contain a
type of ferritin.
6667a
On the other
hand, the yeast S. cerevisieae stores
iron in polyphosphate-rich granules,
even though a ferritin is also
present.
65
Ferritin contains 17 23%
iron as a dense core of hydrated
ferric oxide ~ 7 nm in diameter
surrounded by a protein coat made
up of twenty-four subunits of mo-
Figure 16-3 Structure of the protein shell of ferritin (apoferritin). (A) Ribbon
drawing of the 163-residue monomer. From Crichton.
62
(B) Stereo drawing of
a hexamer composed of three dimers. (C) A tetrad of four subunits drawn as a
space-filling diagram and viewed down the four-fold axis from the exterior of
the molecule. (D) A half molecule composed of 12 subunits inscribed within a
truncated rhombic dodecahedron. BD from Bourne et al.
74
A. Iron
C
D
A
B
842 Chapter 16. Transition Metals in Catalysis and Electron Transport
storage form of iron, hemosiderin, seems to consist
of ferritin partially degraded by lysosomes and con-
taining a higher iron content than does ferritin. Depo-
sitions of hemosiderin in the liver can rise to toxic
levels if excessive amounts of iron are absorbed.
Iron can be deposited in ferritin by allowing apo-
ferritin to stand with an Fe(II) salt and a suitable oxidant,
which may be O
2
. Physiological transfer of Fe(III)
from transferrin to ferritin is thought to require prior
reduction to Fe(II). The reoxidation by O
2
to Fe(III) for
deposition in the ferritin core (Eq. 16-2) is catalyzed by
ferrooxidase sites located in the centers of the helical
bundles of the H-chains.
70 73
2 Fe
2+
+ O
2
+ 4 H
2
O
2 Fe (O) OH
core
+ H
2
O
2
+ 4 H
+
BOX 16-A MAGNETIC IRON OXIDE IN ORGANISMS
An unusual form of stored iron is the magnetic
iron oxide magnetite (Fe
3
O
4
). Honeybees,
a,b
monarch butterflies,
bc
homing pigeons,
c e
migrating
birds, and even magnetotactic bacteria
f
contain
deposits of Fe
3
O
4
that are suspected of being used
in nagivation.
c,g
Some bacteria have magnetic iron
sulfide particles.
h-j
Human beings have magnetic
bones in their sinuses
k
and in their brains
l
and may
be able to sense direction magnetically. A set of
possible magnetoreceptor cells, as well as associated
nerve pathways, have been identified in trout.
j
In
the magnetotactic bacteria found in the Northern
Hemisphere the magnetic domains are oriented
parallel with the axis of motility of the bacteria
which tend to swim toward the geomagnetic North
and downward into sediments. Similar bacteria from
the Southern Hemisphere prefer to swim south and
downward. The magnetic polarity of the bacterial
magnetite crystals can be reversed by strong magnetic
pulses, after which the bacteria swim in the direction
opposite to their natural one.
m
Magnetic ferritin
can be produced artificially in the laboratory.
n
The
resulting particles may have practical uses, for
a
Hsu, C.-Y., and Li, C.-W. (1994) Science 265, 9597
b
Nichol, H., and Locke, M. (1995) Science 269, 18881889
bc
Etheredge, J. A., Perez, S. M., Taylor, O. R., and Jander, R. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 1384513846
c
Gould, J. L. (1982) Nature (London) 296, 205211
d
Guilford, T. (1993) Nature (London) 363, 112113
e
Moore, B. R. (1980) Nature (London) 285, 6970
f
Blakemore, R. P., and Frankel, R. B. (1981) Sci. Am. 245 (Dec),
5865
g
Maugh, T. H., II,(1982) Science 215, 1492 1493
h
Dunin-Borkowski, R. E., McCartney, M. R., Frankel, R. B.,
Bazylinski, D. A., Psfai, M., and Buseck, P. R. (1998) Science
282, 18681870
Magnetotactic soil bacterium containing 36 magnetite-
containing magnetosomes. Courtesy of Dennis Bazylinski.
lecular mass 17 - to 21-kDa. Each subunit is folded as
a four-helix bundle (Fig. 16-3). Mammalian ferritins
consist of combinations of subunits of two or more
types. For example, human ferritins contain similarly
folded 19-kDa L (light) and 21-kDa H (heavy) sub-
units.
68
The twenty-four subunits are arranged in a
cubic array (Fig. 7-13, Fig. 16-3). The outer diameter
of the 444-kDa apoferritin is ~ 12 nm. The completely
filled ferritin molecule contains 23% Fe and over 2000
atoms of iron in a crystalline lattice. Larger ferritins
may contain as many as 4500 atoms of iron with the
approximate composition [Fe(O)OH]
8
FeOPO
3
H
2
.
69
Phosphate ions are sometimes bound into surface
layers of the ferritin cores.
69a
Ferritin cores are readily
visible in the electron microscope, and ferritin is often
used as a labeling reagent in microscopy. Another (16-2)
example, in medical magnetic imaging. Magnetic
materials in the human body are of interest not only
in terms of a possible sensory function but also
because of possible effects of electromagnetic fields
on human health and behavior.
o
i
Psfai, M., Buseck, P. R., Bazylinski, D. A., and Frankel, R. B.
(1998) Science 280, 880883
j
Walker, M. M., Diebel, C. E., Haugh, C. V., Pankhurst, P. M.,
Montgomery, J. C., and Green, C. R. (1997) Nature (London) 390,
371376
k
Baker, R. R., Mather, J. G., and Kennaugh, J. H. (1983) Nature
(London) 301, 7880
l
Kirschvink, J. L., Kobayashi-Kirschvink, A., and Woodford, B. J.
(1992) Proc. Natl. Acad. Sci. U.S.A. 89, 76837687
m
Blakemore, R. P., Frankel, R. B., and Kalmijn, A. J. (1980) Nature
(London) 286, 384385
n
Meldrum, F. C., Heywood, B. R., and Mann, S. (1992) Science
257, 522523
o
Barinaga, M. (1992) Science 256, 967
843
The ferrooxidase site is a dinuclear iron center (see
Section 8) in which two iron ions (probably Fe
2+
) are
bound as in Fig. 16-4. They are then converted to Fe
3+
ions by O
2
, which may bind initially to the Fe
2+
, form-
ing a transient blue intermediate that is thought to
have a peroxodiferric structure, perhaps of the follow-
ing type.
7173
Reaction of this intermediate with H
2
O
may yield H
2
O
2
plus a biomineral precursor Fe
2+
O
Fe
2+
, which is incorporated into the core.
72a
Ferritin H
subunits predominate in tissues with high oxygen levels,
e.g., heart and blood cells, while the L subunits pre-
dominate in tissues with slower turnover of iron, e.g.,
liver.
72
The L subunits lack ferrooxidase activity but,
in the centers of their helical bundles, contain polar
side chains that may help to initiate growth of the
mineral core.
64
Removal of Fe(III) from storage in ferritin cores
may require reduction to Fe(II) again, possibly by
ascorbic acid
75
or glutathione. Some bacterial ferritins
contain a bound cytochrome b which may assist in
reduction.
67,67a
Released iron in the Fe
2+
state can be
incorporated into iron-containing proteins or into heme.
The enzyme ferrochelatase
7676b
catalyzes the transfer
of free Fe
2+
into protoporphyrin IX (Section 3) to form
protoheme (Fig. 16-5). Iron in the Fe(II) state may
also be oxidized to Fe
3+
through action of the copper-
containing ceruloplasmin (Section D) and be incorpo-
rated into heme by direct transfer from ferritin.
53
A. Iron
O O
Fe
3
Fe
3+ +

3. Heme Proteins
In 1879, German physiological chemist Hoppe-
Seyler showed that two of the most striking pigments
of nature are related. The red iron-containing heme
from blood and the green magnesium complex
chlorophyll a of leaves have similar ring structures.
Later, H. Fischer proved their structures and provided
them with the names and numbering systems that are
used today. This information is summarized in the
following section.
Some names to remember. Porphins are planar
molecules which contain large rings made by joining
four pyrrole rings with methine bridges. In the
chlorins, found in the chlorophylls, one of the rings
(ring D in chlorophyll, Fig. 23-20) is reduced. The
specific class of porphins known as porphyrins have
eight substituents around the periphery of the large
ring. Like the chlorins and the corrins of vitamin B
12
(Section B), the porphyrins are all formed biosyntheti-
cally from porphobilinogen. This compound is
polymerized in two ways (see Fig. 24-21) to give por-
phyrins of types I and III (Fig. 16-5). In type I porphy-
rins, polymerization of porphobilinogen has taken
place in a regular way so that the sequences of the
carboxymethyl and carboxyethyl side chains (often
referred to as acetic acid and propionic acid side chains,
respectively) are the same all the way around the
outside of the molecule. However, most biologically
important porphyrins belong to type III, in which the
first three rings A, B, and C have the same sequence of
carboxymethyl and carboxyethyl side chains, but in
which ring D has been incorporated in a reverse fashion.
Thus, the carboxyethyl side chains of rings C and D
are adjacent to each other (Fig. 16-5). Porphyrins
containing all four carboxymethyl and four carboxy-
ethyl side chains intact are known as uroporphyrins.
Uroporphyrins I and III are both excreted in small
amounts in the urine. Another excretion product is
coproporphyrin III, in which all of the carboxymethyl
side chains have been decarboxylated to methyl groups.
The feathers of the tropical touraco are colored with
copper(II) complex of coproporphyrin III and this
Figure 16-4 The dinuclear iron center or ferroxidase center
of human ferritin based on the structure of a terbium(III)
derivative.
73
Courtesy of Pauline Harrison.
O
O O
Q141
E107
N
H
H
H H
O

O
O
CH
3
A144
O O
O
E62
E61
H65
N
H
N
E27
O
Fe
A
Fe
B

Porphobilinogen
N
CH
2
CH
2
H
HOOC CH
2
COOH
CH
2
H
2
N
844 Chapter 16. Transition Metals in Catalysis and Electron Transport
porphyrin as well as others are commonly found in
birds eggs. The heme proteins are all derived from
protoporphyrin IX, which is formed by decarboxylation
and dehydrogenation of two of the carboxyethyl side
chains of uroporphyrin III to vinyl groups (Fig. 16-5).
Hemes and heme proteins. Protoporphyrin IX
contains a completely conjugated system of double
bonds. In the center two hydrogen atoms are attached,
one each to two of the nitrogens; they are free to move
to other nitrogens in the center with rearrangement of
the double bonds. Thus, there is tautomerism as well
as resonance within the heme ring.
7778a
The two
central hydrogens can be replaced by many metal ions
to form stable chelates. The complexes with Fe
2+
are
known as hemes and the Fe
2+
complex with protopor-
phyrin IX as protoheme. Heme complexes of Fe
2+
may be designated as ferrohemes and the Fe
3+
com-
pounds as ferrihemes. The Fe
3+
protoporphyrin IX
is also called hemin, and may be crystallized as a
chloride salt.
78b
Iron tends to have a coordination
number of six, and other ligands can attach to the iron
from the two axial positions on opposite sides of the
planar heme. If these are nitrogen ligands, such as
pyridine or imidazole, the resulting compounds,
called hemochromes, have characteristic absorption
spectra. An example is cytochrome b
5
, which contains
two axial imidazole groups.
Several modifications of protoheme are indicated
in Fig. 16-5. To determine which type of heme exists
in a particular protein, it is customary to split off the
heme by treatment with acetone and hydrochloric acid
and to convert it by addition of pyridine to the pyri-
dine hemochrome for spectral analysis. By this means,
protoheme was shown to occur in hemoglobin, myo-
globin, cytochromes of the b and P450 types, and
catalases and many peroxidases. Cytochromes a and
a
3
contain heme a, while one of the terminal oxidase
Uroporphyrin I Coproporphyrin III
Protoheme and variants
Figure 16-5 Structures of some biologically
important porphyrins. (A) Uroporphyrin I.
(B) Coproporphyrin III. Note that a different
tautomeric form is pictured in B than in A.
Tautomerism of this kind occurs within all
of the porphyrins. (C) Protoheme, the Fe
2+
complex of protoprophyrin IX, present in
hemoglobin, cytochromes b, and other proteins.
A B
C D
COOH
COOH
COOH
HOOC
COOH
COOH
COOH
N
N N
N
H
H
HOOC
COOH
COOH
N
N N
N
Fe
H
3
C
H
3
C
CHO in heme a
CH
3
CH
2
C
HC
CH
2
CH O
CH
2
CH
in chlorocruoroheme
CH
2
OH
CH C
CH
3
CH
2
H in heme a and o
CH
3
in heme d
CH
3
OH
OH
COOH
Both vinyl groups of protoheme are
converted to CH
S
CH
3
in heme c
protein
A B
C D
COOH
CH
3
COOH
H
3
C
COOH
COOH
CH
3
N
N N
N
H H
H
3
C
3
CH
3
in heme o
C
A B
845
Figure 16-6 Structures of
isobacteriochlorin prosthetic
groups. (A) Siroheme from
nitrite and sulfite reductases;
(B) acrylochlorin heme from
dissimilatory nitrite reductases
of Pseudomonas and Paracoccus.
N
N N
N
Fe
COOH
HOOC
COOH
COOH
H
HOOC
H
3
C
COOH
H
H
3
C
COOH
COOH
in Siroamide heme
H
2
N C
O
in both rings
of heme d
1
C
O
N
N N
N
Fe
COOH
H
3
C
COOH
CH
3
HOOC
H
3
C
COOH
H
3
C
C
C
HO
O
OH
O
Siroheme
Acrylochlorin heme
A. Iron
systems of enteric bacteria contains the closely related
heme o (Fig. 16-5).
79,80
A second terminal oxidase of
those same bacteria contains heme d (formerly a
2
).
Heme c (present in cytochromes c and f) is a variation
in which two SH groups of the protein have added to
the vinyl groups of protoheme to form two thioether
linkages (Fig. 16-5). A few cytochromes c have only
one such linkage. In myeloperoxidase (Section 6) three
covalent linkages, different than those in cytochrome c,
join the heme to the protein.
81,82
There is a possibility
that heme a may sometimes form a Schiff base with a
lysyl amino group through its formyl group.
83,84
Heme d is a chlorin,
85
as is acrylochlorin heme
from certain bacterial nitrite reductases (Fig. 16-6).
86,87
Siroheme (Fig. 16-6), which is found in both nitrite
and sulfite reductases of bacteria (Chapter 24),
88,89
is an isobacteriochlorin in which both the A and B
rings are reduced. It apparently occurs as an amide
siroamide (Fig. 16-6) in Desulfovibrio.
90
Heme d
1
of
nitrite reductases of denitrifying bacteria is a dioxo-
bacteriochlorin derivative (Fig. 16-6).
91,92
As in myoglobin, hemoglobin (Fig. 7-23), and
cytochrome c (see Fig 16-8), one axial coordination
position on the iron of most heme proteins (customarily
called the proximal position) is occupied by an imida-
zole group of a histidine side chain. However, in
cytochrome P450 and chloroperoxidase a thiolate ( S

)
group from a cysteinyl side chain, and in catalase a
phenolate anion from a tyrosyl side chain, occupies
the proximal position. The sixth or distal coordination
position is occupied by the sulfur atom of methionine
in cytochrome c and most other cytochromes with
low-spin iron but cytochromes b
5
and c
3
have histidine.
The high-spin heme proteins, such as cytochromes c,
A
B
globins, peroxidase, and catalase, usually have no
ligand other than weakly bound H
2
O in the distal
position.
93
Hemes are found in all organisms except the
anaerobic clostridia and lactic acid bacteria. Heme
proteins of blood carry oxygen reversibly, whereas
those of terminal oxidase systems, hydroxylases,
and oxygenases activate oxygen, catalyzing reac-
tions with hydrogen ions and electrons or with carbon
compounds. The heme-containing peroxidases and
catalases catalyze reactions not with O
2
but with H
2
O
2
.
Another group of heme proteins includes most of the
cytochromes, which are purely electron-transferring
compounds.
4. The Cytochromes
The iron in the small proteins known as cyto-
chromes acts as an electron carrier, undergoing alter-
nate reduction to the +2 state and oxidation to the
+3 state. The cytochromes, discovered in 1884 by
McMunn,
94
were first studied systematically in the
1920s by Keilin (Chapter 18) and have been isolated
from many sources.
95 97
The classification into groups
a, b, and c according to the position of the longest
wavelength light absorption band (the band; Fig.
16-7) follows a practice introduced by Keilin. How-
ever, it is now customary to designate a new cyto-
chrome by giving the heme type (a, b, c or d) together
with the wavelength of the band, e.g., cytochrome
c
552
or cyt b
557.5
.
Cytochromes of the b type including bacterial
cytochrome o contain protoheme. Because the sixth
846 Chapter 16. Transition Metals in Catalysis and Electron Transport
position is ligated, most cytochromes b do not react
with O
2
. However, cytochromes o and d serve as
terminal electron acceptors (cytochrome oxidases) and
are oxidizable by O
2
. Another protoheme-containing
cytochrome, involved in hydroxylation (Chapter 18), is
cytochrome P450. Here the 450 refers to the position
of the intense Soret band (also called the band) of
the spectrum (Fig. 16-7) in a difference spectrum run in
the presence and absence of CO. Other properties are
also used in arriving at designations for cytochromes.
For example, cytochrome a
3
has a spectrum similar to
that of cytochrome a but it reacts readily with both CO
and O
2
.
Another property that distinguishes various cyto-
chromes is the redox potential E (Table 6-8), which in
this discussion is given for pH 7.0. Cytochromes carry
electrons between other oxidoreductase proteins of
widely varying values of E
0'
. Because of the various
heme environments cytochromes have greatly differing
values of E
0'
, allowing them to function in many differ-
ent biochemical systems.
97a,97b
For mitochondrial cyto-
chrome c the value of E is ~ + 0.265 V but for the closely
related cytochrome f of chloroplasts it is ~ +0.365 V
and for cytochrome c
3
of Desulfovibrio about 0.330 V.
There is more than an 0.6-volt difference between E
of cytochromes f and c
3
. Cytochromes b tend to have
lower E values, close to zero, than most cytochromes
c, while cytochrome a
3
has E ~ +0.385 V.
The c-type cytochromes. Mitochondrial cyto-
chrome c is one of the few intracellular heme pigments
that is soluble in water and that can be removed easily
from membranes. A small 13-kDa protein typically
containing about 104 amino acid residues, cytochrome
c has been isolated from plants, animals, and eukary-
otic microorganisms.
95 97,99,100
Complete amino acid
sequences have been determined for over 100 species.
Within the peptide chain 28 positions are invariant
and a number of other positions contain only conser-
vative substitutions. Cytochrome c was one of the first
proteins to be used in attempting to trace evolutionary
relationships between species by observing differences
in sequence. Humans and chimpanzees have identical
cytochrome c, but 12 differences in amino acid sequence
occur between humans and the horses and 44 between
human and Neurospora.
96
The related cytochrome c
2
of
the photosynthetic bacterium Rhodospillum rubrum is
thought to have diverged in evolution 2 x 10
9
years
ago from the precursor of mammalian cytochrome c.
Even so, 15 residues remain invariant.
101
Structural studies
95 97,101 103
on cytochromes of the
c and c
2
types show that the heme group provides a
core around which the peptide chain is wound. The
104 residues of mitochondrial cytochrome c are enough
to do little more than envelope the heme. In both the
oxidized and reduced forms of the protein, methionine
80 (to the left in Fig. 16-8A) and histidine 18 (to the
right) fill the axial coordination positions of the iron.
The heme is nearly buried and inaccessible to the
surrounding solvent.
The shorter chains of the 82- to 86-residue cyto-
chromes c
550
(from Pseudomonas
102
), c
553
,
102a
and c
555
(from Chlorobium
105
) as well as the longer 112-residue
polypeptide of cytochrome c
2
from Rhodospirillum
rubrum
93
have nearly the same folding pattern as that in
mitochondrial cytochrome c. However, the 128-residue
chain of the dimeric cytochrome c from Rhodospirillum
molischianum forms an antiparallel four-helix bundle
(Fig. 16-8).
106109
This is the same folding pattern present
in the ferritin monomer (Fig. 16-3), hemerythrin (Fig.
2-22), and many other proteins including cytochrome
b
562
of E. coli.
110
Cytochrome f, which functions in
photosynthetic electron transport, is also a c-type
cytochrome but with a unique protein fold.
111,112
Most cytochromes have only one heme group per
polypeptide chain,
112
but the 115-residue cytochrome
c
3
from the sulfate-reducing bacterium Desulfovibrio
binds four hemes (Fig. 16-8C).
104,113 115
Each one seems
to have a different redox potential in the 0.20 to
0.38 V range.
114
Another c-type cytochrome, also
from Desulfovibrio, contains six hemes in a much larger
66-kDa protein and functions as a nitrite reductase.
116
Figure 16-7 Absorption spectra of oxidized and reduced
horse heart cytochrome c at pH 6.8. From data of Margoliash
and Frohwirt.
98
Wave number, cm
1
10
-3
16 20 24 28 32 36 40 44
12
10
8
6
4
2
0
14
16
M
o
l
a
r

e
x
t
i
n
c
t
i
o
n

c
o
e
f
f
i
c
i
e
n
t
,


1
0
-
4
Wavelength, nm
500 400 350 300 250 220 600

(soret)
Reduced
Oxidized
847
Figure 16-8 Structures of three
cytochromes of the c type. (A)
Horse mitochondrial cytochrome c;
(B) a subunit of the dimeric
cytochrome c from Rhodospirillum
molischianum; (C) cytochrome c
3
from Desulfovibrio desulfuricans.
(A) and (B) courtesy of Salemme;
101
(C) from Simes et al.
104
Courtesy
of Maria A. Carrondo.
A. Iron
Triheme and octaheme proteins are also known.
117
Many cytochromes c are soluble but others are
bound to membranes or to other proteins. A well-
studied tetraheme protein binds to the reaction centers
of many purple and green bacteria and transfers elec-
trons to those photosynthetic centers.
118 120
Cytochrome
c
2
plays a similar role in Rhodobacter, forming a complex
of known three-dimensional structure.
121
Additional
cytochromes participate in both cyclic and noncyclic
electron transport in photosynthetic bacteria and
algae (see Chapter 23).
120,122 124
Some bacterial mem-
branes as well as those of mitochondria contain a
cytochrome bc
1
complex whose structure is shown
in Fig. 18-8.
125,126
Cytochromes b, a, and o. Protoheme-containing
cytochromes b are widely distributed.
127,128
There are
at least five of them in E. coli. Whether in bacteria,
mitochondria, or chloroplasts, the cytochromes b function
within electron transport chains, often gathering elec-
trons from dehydrogenases and passing them on to
c-type cytochromes or to ironsulfur proteins. Most
cytochromes b are bound to or embedded within
membranes of bacteria, mitochondria, chloroplasts,
or endoplasmic reticulum (ER). For example, cyto-
chrome b
5
129,129a
delivers electrons to a fatty acid desat-
urating system located in the ER of liver cells and to
many other reductive biosynthetic enzymes.
130 132
The
protein contains 132 amino acid residues plus another
85 largely hydrophobic N-terminal residues that pro-
vide a nonpolar tail which is thought to be buried in
the ER membranes.
130
Solubilization of the protein
causes loss of this N-terminal sequence. The heme in
cytochrome b
5
is not covalently bonded to the protein
but is held tightly between two histidine side chains.
The polypeptide chain is folded differently than in
either cytochrome c or myoglobin.
The folding pattern of cytochrome b
5
is also found
in the complex heme protein flavocytochrome b
2
from yeast (Chapter 15)
133
and probably also in liver
sulfite oxidase
134,135
Both are 58-kDa peptides which
can be cleaved by trypsin to 11-kDa fragments that
have spectroscopic similarities and sequence homology
with cytochrome b
5
. Sulfite oxidase also has a molyb-
denum center (Section H). The 100-residue N-terminal
portion of flavocytochrome b
2
has the cytochrome b
5
folding pattern but the next 386 residues form an
eight-stranded ( /)
8
barrel that binds a molecule
of FMN.
133,136
All of these proteins pass electrons to
cytochrome c. In contrast, the folding of cytochrome
C
A
B
848 Chapter 16. Transition Metals in Catalysis and Electron Transport
b
562
of E. coli resembles that of cytochrome c (Fig.
16-8).
110,137
However, it has methionine side chains
as both the fifth and sixth iron ligands.
Cytochromes b of mitochondrial membranes are
involved in passing electrons from succinate to ubiqui-
none in complex II
138
and also from reduced ubiqui-
none to cytochrome c
1
in the 248-kDa complex III (Fig.
18-8). A similar complex is present in photosynthetic
purple bacteria.
123,139
Cytochrome b
560
functions in the
transport of electrons from succinate dehydrogenase
to ubiquinone,
138
and cytochrome b
561
of secretory
vesicle membranes has a specific role in reducing
ascorbic acid radicals.
140
In bacteria some cytochromes b and d
1
serve as
terminal electron carriers able to react with O
2
, nitrite,
or nitrate, while others act as carriers between redox
systems.
141 143a
The aldehyde heme a is utilized by
animals and by some bacteria in cytochrome c oxidase,
a complex enzyme whose three-dimensional structure
is known (see Fig. 18-10) and which is discussed further
in Chapter 18.
5. Mechanisms of Biological Electron Transfer
The heme groups of the cytochromes as well as
many other transition metal centers act as carriers of
electrons. For example, cytochrome c may accept an
electron from reduced cytochrome c
1
and pass it to
cytochrome oxidase or cytochrome c peroxidase. The
electron moves from one heme group to another over
distances as great as 2 nm. Similar electron-transfer
reactions between defined redox sites are met in photo-
synthetic reaction centers (Fig. 23-31), in metalloflavo-
proteins (Fig. 15-9), and in mitochondrial membranes.
What are the factors that determine the probability
of an electron transfer reaction and the rate at which it
may occur? They include: (1) The distance from the
electron donor to the acceptor. (2) The thermodynamic
driving force G for the reaction. This can be approx-
imated using the difference in standard electrode
potentials (as in Table 6-1) between donor and acceptor.
G = 96.5 E kJ/ mol at 25 C. (3) The chemical
makeup of the material through which the electron
transfer takes place. (4) Any changes in the geometry
or charge state of the donor or acceptor that accompany
the transfer. (5) The orientation of the acceptor and
donor groups.
144 146
It is usually assumed that the
FranckCondon principle is obeyed, i.e., that the elec-
tron jump occurs so rapidly (< 10
12
s) that there is no
change in the positions of atomic nuclei (see also
Chapter 23). Subsequent rearrangement of nuclear
positions may occur at rates that allow a rapid overall
reaction.
The various factors that affect the rate of electron
transfer were incorporated by Marcus into a quantita-
tive theory. Electron transfer is often discussed in
terms of this classic Marcus theory together with
effects of quantum mechanical tunneling.
144,147 150
According to Marcus the electron transfer rate from
a donor to an acceptor at a fixed separation depends
upon G, a nuclear reorganization parameter (), and
the electronic coupling strength H
AB
between reac-
tant and product in the transition state (Eq. 16-3):
k
ET
= (4
3
/h
2
k
B
T) | H
AB
|
2
exp [ (G + )
2
/ 4 k
B
T]
Here H
AB
is a quantum mechanical matrix whose
strength decreases exponentially with the distance of
separation R as e
R
where is a coefficient of the order
of 9 14 nm
1
. At the closest contact (R = 0) the rate k
ET
,
by extrapolation from experimental data on small syn-
thetic compounds, is close to the molecular vibration
frequency of 10
13
s
1
.
151,152
At distances greater than 2 nm
the rate would be negligible were it not for other factors.
Using mutant proteins as well as a variety of redox
pairs and electron-transfer distances the validity of the
Marcus equation with respect to the thermodynamic
driving force and distance dependence has been veri-
fied.
153
This is even true for cytochrome c mutants
functioning in living yeast cells.
146
A huge amount of experimental work with proteins
has been done to test and refine the theories of electron
transport. For example, electron donor groups with
various reduction potentials have been attached to
various sites on the surface of a protein containing a
heme or other electron accepting group. Ruthenium
complexes such as Ru(III) (NH
3
)
5
3+
form tight covalent
linkages to imidazole nitrogens
154,155
such as that of
His 33 of horse heart cytochrome c. This metal can
be reduced rapidly to Ru(II) by an external reagent,
after which the transfer of an electron from the Ru(II)
across a distance of 1.2 nm to the heme Fe(III) can be
followed spectroscopically. The reduction potentials
E for the Ru(III) / Ru(II) and Fe(III) / Fe(II) couples at
pH 7 in these compounds are 0.16 and 0.27 V, respec-
tively. Thus, an electron will jump spontaneously
from the Ru(II) to the Fe(III) with G = 15.4 kJ/ mol.
A rate constant of ~5 s
1
, which was nearly independent
of temperature, was observed. Since the structures
of Fe(II) and Fe(III) forms of cytochrome c differ only
slightly,
156
the electron transfer apparently occurs with
only a small amount of geometric rearrangement. The
distribution of charges and dipoles within the protein
may be such that the Fe
2+
and Fe
3+
complexes have
almost equal thermodynamic stability.
Electron-transfer pathways? In spite of the
success of the Marcus theory, rates of electron-transfer
from the iron of cytochrome c have been found to vary
for different pathways.
150,153,155
For example, transfer
of an electron from Fe(II) in reduced cytochrome c to
an Ru(III) complex on His 33 was fast (~ 440 s
1
)
157
but
(16-3)
849
the rate of transfer to an equidistant Fe(III) ion on Met
65 was at most 0.6 s
1.
These results suggested that
distinct electron-transfer pathways exist. One sugges-
tion was that the sulfur atom of Met 80 donates an
electron to Fe
3+
leaving an electron-deficient radical.
The hole so created could be filled by an electron
jumping in from the OH group of the adjacent Tyr 67,
which might then accept an electron from an external
acceptor via Tyr 74 at the protein surface. Do elec-
trons flow singly or as pairs from the surface through
hydrogen-bonded paths? Use of both semisynthesis
155
and directed mutation
153
of cytochromes c is permitting
a detailed study of these effects. A striking result is
that substitution of the conserved residue phenylalanine
82 in a yeast cytochrome c with leucine or isoleucine
retards electron transfer by a factor of ~ 10
4
.
Docking. It is now recognized that there are
distinct docking sites on the surface of electron-
transport proteins. For rapid electron transfer to occur
the two electron carriers must be properly oriented
and docked by formation of correct polar and nonpolar
interactions. Early indications of the importance of
docking came from study of modified cytochromes c.
Each one of the 19 lysine side chains was individually
altered by acylation or alkylation to remove the positive
charge or to replace it with a negative charge. The rate
of electron transfer into cytochrome c from hexacyano-
ferrate was decreased by a factor of 1.3 2.0 when any
one lysine at positions 8, 13, 27, 72, or 79, which are
clustered around the heme edge, was modified.
158
Modification of Lys 22, 55, 99, or 100, distant from this
edge, had no effect. Electron transfer into cytochrome
c from its natural electron donor ubiquinol:cytochrome
c reductase was also strongly inhibited by modification
of lysines that surround the heme edge.
159
Modification
of these lysines also inhibited electron transfer out of
Figure 16-9 Stereoscopic
-carbon plot of yeast cyto-
chrome c peroxidase (top)
and yeast cytochrome c
(below) as determined from
a cocrystal by Pelletier and
Kraut.
164
The heme rings of
the two proteins appear in
bold lines, as does the ring
of tryptophan 191 and the
backbone of residues 191
193 of the cytochrome c
peroxidase. Drawing from
Miller et al.
165
cytochrome c into its natural acceptors, cytochrome c
oxidase and cytochrome c peroxidase (Fig. 16-9).
160
A major factor in these effects is probably the large
dipole moment in the cytochrome c that arises from
the unequal distribution of surface charges. This
charge distribution must assist in the proper docking
of the cytochrome with its natural electron donors and
acceptors. The positive surface charges presumably
also facilitate the reaction with hexacyanoferrate (II)
or ascorbate, both of which are negatively charged
reductants that react rapidly with cytochrome c.
Measurements of many kinds have been made
between natural donoracceptor pairs such as cyto-
chrome ccytochrome b
5
,
161,162
cytochrome ccytochrome
c peroxidase (Fig. 16-9),
153,163166
trimethylamine dehy-
drogenaseFMN to Fe
4
S
4
center (Fig. 15-9),
167
and
methylamine dehydrogenase (TTQ radical)amicya-
nin (Cu
2+
).
168
Designed metalloproteins are being
studied as well.
169
Femtosecond laser spectroscopy is
providing a new approach.
169a
Coupling and gating of electron transfer.
Electrons are thought to be transferred into or out of
cytochrome c through the exposed edge of the heme.
The rate depends upon effective coupling, which in
turn may depend upon orientation as well as the
structure and dynamics of the protein.
170
Proteins
with much structure appear to provide stronger
coupling than those that are largely composed of
helices.
144,171
The high nuclear reorganization energy
of helices may block electron transfer along some
pathways.
153
A conformational change,
167,169
transfer
of a proton, or binding of some other specific ion
172
before electron transfer occurs can be the gating process
that determines the rate of electron transfer.
162,167,173
Electron transfer can also be coupled to an unfavor-
able, but fast, equilibrium.
A. Iron
850 Chapter 16. Transition Metals in Catalysis and Electron Transport
rapid reactions. Two oxygen atoms, each with six
valence electrons, might reasonably be expected to
form dioxygen, O
2
, as a double-bonded structure with
one and one bond as follows (left):
However, O
2
is paramagnetic and contains two un-
paired electrons.
175
From this evidence O
2
might be
assigned the structure on the right.
175
The oxygen
molecule is very stable, and it is relatively difficult
to add an electron to form the reactive superoxide
anion radical O
2

.
For this reason, oxidative attack by O
2
tends to be
slow. However, once an electron has been acquired,
it is easy for additional electrons to be added to the
structure and further reduction occurs more easily.
The biochemical question suggested is, How can
some heme proteins carry O
2
reversibly without any
oxidation of the iron contained in them while others
activate oxygen toward reaction with substrates?
Among this latter group, cytochrome c oxidase transfers
electrons to both oxygen atoms so that only H
2
O is a
product, whereas the hydroxylases and oxygenases,
which are discussed in Chapter 18, incorporate either
one or two of the atoms of O
2
, respectively, into an
organic substrate. Before examining these reactions
let us reconsider the heme oxygen carriers.
Oxygen-carrying proteins. In Chapter 7, we
examined the behavior of hemoglobin in the coopera-
tive binding of four molecules of O
2
and studied its
structural relationship to the monomeric muscle pro-
tein myoglobin.
176
The iron in functional hemoglobin
and myoglobin is always Fe(II) and is only very slowly
converted by O
2
into the Fe(III) forms methemoglobin
or metmyoglobin.
177,178
Erythrocytes contain an enzyme
system for immediately reducing methemoglobin back
to the Fe(II) state (see Box 15-H).
Binding of O
2
to the iron in the heme is usually
considered not to cause a change in the oxidation state
of the metal. However, oxygenated heme has some of
the electronic characteristics of an Fe
3+
OO

peroxide
anion. Bonding of the heme iron to oxygen is thought
to occur by donation of a pair of electrons by the oxy-
gen to the metal. In deoxyhemoglobin the Fe(II) ion is
in the high-spin state; four of the five 3d orbitals in
the valence shell of the iron contain one unpaired
electron and the fifth orbital contains two paired elec-
trons. The binding of oxygen causes the iron to revert
Effects of ionic equilibria on electron transfer.
The charge on an ion of Fe
2+
in a heme is exactly bal-
anced by two negative charges on the porphyrin ring.
However, when the Fe
2+
loses an electron to become
Fe
3+
an extra positive charge is suddenly present in the
center of the protein. This change in charge will have
a powerful electrostatic effect on charged groups in the
immediate vicinity of the iron and even at the outer
surface of the molecule. For example, an anion from
the medium or from a neighboring protein molecule
might become bound to the heme protein (Eq. 16-4).
X

+ [ Fe (II) (heme) ] [ Fe (III) (heme) ]


+
X

+ e

In this case the presence of a high concentration of X

in the medium would favor the oxidation of Fe(II) to


Fe(III). The reduced heme would be a better reducing
agent and the oxidized form a weaker oxidant than in
the absence of X

. If YH were a group in the protein


the loss of an electron could cause YH to dissociate so
that Y

and the Fe(III) would interact more tightly (Eq.


16-5).
HY [ Fe (II) (heme) ]
Y

[ Fe (III) (heme) ]
+
+ H
+
+ e

We see that electron transfer can be accompanied by


loss of a proton and that E may become pH depen-
dent. (See also Eq. 16-18.) Even with cytochrome c,
although there is little structural change upon electron
transfer, there is an increased structural mobility in the
oxidized form.
156
This may be important for coupling
and could also facilitate associated proton-transfer
reactions. For example, it is possible that in some
cytochromes the imidazole ring in the fifth coordina-
tion position may become deprotonated upon oxida-
tion. This possibility is of special interest because
cytochromes are components of proton pumps in
mitochondrial membranes (Chapter 18).
6. Reactions of Heme Proteins with Oxygen
or Hydrogen Peroxide
As Ingraham remarked,
174
Living in a bath of
20% oxygen, we tend to forget how reactive it is.
From a thermodynamic viewpoint, all living matter
is extremely unstable with respect to combustion by
oxygen. Ordinarily, a high temperature is required
and if we are careful with fire, we can expect to escape
a catastrophe. However, one mole of properly chelated
copper could catalyze consumption of all of the air in
an average room within one second.
174
Biochemists
are interested in both the fact that O
2
is kinetically
stable and unreactive and also that oxidative enzymes
such as cytochrome c oxidase are able to promote
O O
Dioxygen
O O
O O
Superoxide anion radical

(16-4)
(16-5)
851
The coordination of the heme iron to histidine also
appears to provide the basis for the cooperativity in
binding of oxygen by hemoglobin.
186
The radius of
high-spin iron, whether Fe(II) or Fe(III), is so large that
the iron cannot fit into the center of the porphyrin ring
but is displaced toward the coordinated imidazole
group by a distance of ~ 0.04 nm for Fe(II).
187
Thus,
in deoxyhemoglobin both iron and the imidazole
group lie further from the center of the ring than they
do in oxyhemoglobin. In the latter, the iron lies in the
center of the porphyrin ring because the change to the
low-spin state is accompanied by a decrease in ionic
radius.
186,188
The change in protein conformation
induced by this small shift in the position of the iron
ion was described in Chapter 7. However, the exact
nature of the linkage between the Fe position and the
conformational changes is not clear.
The mechanical response to the movement of the
iron and proximal histidine, described in Chapter 7,
may explain this linkage. However, oxygenation may
also induce a change in the charge distribution within
the hydrogen-bond network of the protein. The carbo-
nyl group shown in the foregoing structure is attached
to the F helix (see Fig. 7-23) and is
also hydrogen bonded to other amide
groups. Electron withdrawal into
the hemeoxygen complex would
tend to strengthen the hydrogen
bond as indicated by the resonance
forms shown and also to weaken
to the low-spin state in which all of the electrons are
paired and the paramagnetism of hemoglobin is lost.
The stability of hemeoxygen complexes is thought to
be enhanced by back-bonding, i.e., the donation of
an electron pair from one of the filled d orbitals of the
iron atom to form a bond with the adjacent oxygen.
179
This can be indicated symbolically as follows:
These structures, which have been formulated by
assuming that one of the unshared electron pairs on
O
2
forms the initial bond to the metal, are expected to
lead to an angular geometry which has been observed
in X-ray structures of model compounds,
180
in oxy-
myoglobin (Fig. 16-10),
181,182
and in oxyhemoglobin.
183
Neutron diffraction studies have shown that the outer-
most oxygen atom of the bound O
2
is hydrogen bonded
to the H atom on the N

atom of the distal imidazole


ring of His E7 (Fig. 16-10). Carbon monoxide binds
with the C O axis perpendicular to the heme plane
and unable to form a corresponding hydrogen bond.
184
This decreases the affinity for CO and helps to protect
us from carbon monoxide poisoning.
All oxygen-carrying heme proteins have another
imidazole group that binds to iron on the side oppo-
site the oxygen site. Without this proximal imidazole
group, heme does not combine with oxygen. Coordi-
nation with heterocyclic nitrogen compounds favors
formation of low-spin iron complexes and simple
synthetic compounds that closely mimic the behavior
of myoglobin have been prepared by attaching an
imidazole group by a chain of appropriate length to
the edge of a heme ring.
179,185
Similar compounds
bearing a pyridine ring in the fifth coordination position
have a low affinity for oxygen. Thus, the polarizable
imidazole ring itself seems to play a role in promoting
oxygen binding. The electrons of the imidazole ring
may also participate in bonding to the iron as is indi-
cated in the following structures.
179
The bonding
to the iron would allow the iron to back-bond more
strongly to an O
2
atom entering the sixth coordination
position. These diagrams illustrate another feature
found frequently in heme proteins: The N H group
of the imidazole is hydrogen bonded to a peptide
backbone carbonyl group.
H N
O N N Fe H
H N
O N N Fe
+
+ +
H
+
A. Iron
O
Fe
O
+
O
Fe
O

Figure 16-10 Geometry of bonding of O


2
to myoglobin and
position of hydrogen bond to N

of the distal histidine E7


side chain. After Perutz.
182
N
H
His E7
N
N
H
N
His F8
Fe
Val E11
Phe CD1
O
O
852 Chapter 16. Transition Metals in Catalysis and Electron Transport
competing hydrogen bonds.
189,190
This could affect the
charge distribution in the upper end of the F helix and
could conceivably induce a momentary conformational
change that could facilitate the rearrangement of
structure that was discussed in Chapter 7 (Fig. 7-25).
The
1

2
contact in which a change of hydrogen bond-
ing takes place is located nearby behind the F and G
helices. In any event, it is remarkable that nature has
so effectively made use of the subtle differences in the
properties of iron induced by changes in the electron
distribution within the d orbitals of this transition
metal.
A few groups of invertebrates, e.g., the sipun-
culid worms, use a nonheme iron-containing protein,
hemerythrin, as an oxygen carrier.
191,192
Its 113-
residue subunits are often associated as octamers of
C
4
symmetry, each peptide chain having a four-helix
bundle structure (Fig. 2-22). Instead of a heme group,
each monomer contains two atoms of high-spin Fe(II)
held by a cluster of histidine and carboxylate side chains
(see Fig. 16-20).
193,194
Hemerythrin is a member of a
group of such diiron oxoproteins which are considered
further in Section 8. The copper oxygen carrier
hemocyanin is discussed in Section D.
Catalases and peroxidases. Many iron and
copper proteins do not bind O
2
reversibly but activate
it for further reaction. We will look at such metallo-
protein oxidases in Chapter 18. Here we will consider
heme enzymes that react not with O
2
but with peroxides.
The peroxidases,
194a
which occur in plants, animals, and
fungi, catalyze the following reactions (Eq. 16-6, 16-7):
H
2
O
2
+ AH
2
2 H
2
O + A
ROOH + AH
2
R OH + H
2
O + A
Here AH
2
is an oxidizable organic compound such as
an alcohol or a pair of one-electron donor molecules.
Catalases, which are found in almost all aerobic cells,
194b
may sometimes account for as much as 1% of the dry
weight of bacteria. The enzyme catalyzes the break-
down of H
2
O
2
to water and oxygen by a mechanism
similar to that employed by peroxidases. If Eq. 16-7 is
rewritten with H
2
O
2
for AH
2
and O
2
for A, we have
the following equation:
The action of catalase is very fast, almost 10
4
times
faster than that of peroxidases. The molecular activity
per catalytic center is about 2 x 10
5
s
1
.
Catalase exerts a protective function by preventing
the accumulation of H
2
O
2
which might be harmful to
cell constituents. The complete intolerance of obligate
anaerobes to oxygen may result from their lack of this
enzyme. Support for this protective function comes
from the existence of the human hereditary condition
acatalasemia.
195,196
Persons with extremely low
catalase activity are found worldwide but are especially
numerous in Korea. In Japan it is estimated that there
are 1800 persons lacking catalase. Because about half
of them have no symptoms, catalase might be judged
unessential. However, many of the individuals affected
develop ulcers around their teeth. Apparently, hydro-
gen peroxide produced by bacteria accumulates and
oxidizes hemoglobin to methemoglobin (Box 15-H)
depriving the tissues of oxygen.
Catalase from most eukaryotic species is tetra-
meric.
197
The protein from beef liver consists of 506-
residue subunits.
198
Human catalase is similar.
198a
The proximal ligand to the heme Fe
3+
is a tyrosinate
anion (Tyr 358), while side chains of His 75 and Asn
148 lie close to the heme on the distal H
2
O
2
-binding
side (Fig. 16-11). Larger ~ 650-residue fungal and
bacterial catalases have a similar folding pattern but
an extra C-terminal domain with a flavodoxin-like
structure.
197,199
Catalase is gradually inactivated by its
very reactive substrate. As isolated, beef liver catalase
usually contains about two subunits in which the heme
ring has been oxidatively cleaved to biliverdin
200
(Fig. 24-24) and various other alterations have been
found.
197
Each subunit of mammalian catalases nor-
mally contains a bound molecule of NADPH which
helps to protect against inactivation by H
2
O
2
.
201,202
Catalases from Neurospora and from E. coli contain
heme d rather than protoporphyrin.
197,203
Some lacto-
bacilli, lacking heme altogether, form a manganese-
containing pseudocatalase.
204,205
Of the plant peroxidases, which are found in
abundance in the peroxisomes, the 40-kDa mono-
meric horseradish peroxidase has been studied the
most.
206 208a
It occurs in over 30 isoforms and has an
extracellular role in generating free radical intermedi-
ates for polymerization and crosslinking of plant cell
wall components.
209
Secreted fungal peroxidases, e.g.,
such as those from Coprinus
210
and Arthromyces,
211
form a second class of peroxidases with related struc-
tues.
212
A third class is represented by ascorbate
peroxidase from the cytosol of the pea
212 214
and by
the small 34-kDa cytochrome c peroxidase from
yeast mitochondria
215
(Fig. 16-11). The latter has a
strong preference for reduced cytochrome c as a sub-
strate (Eq. 16-9).
216,217
(16-6)
(16-7)
(16-8) 2 H
2
O
2
2 H
2
O + O
2
Catalase
Cytochrome c
peroxidase
2 Cytochrome c (Fe
2+
)
2 Cytochrome c (Fe
3+
)
2H
+
H
2
O
ROOH
ROH
(16-9)
853
Figure 16-11 (A) Stereo drawing showing folding pattern
for beef liver catalase and the positions of the NADPH (upper
left) and heme (center). From Fita and Rossmann.
198
(B)
Diagram of proposed structure of an Fe(III) OOH ferric
peroxide complex of human catalase (see also Fig. 16-14).
A possible mechanism by which the peroxide is cleaved
(step b in Fig. 16-14) is indicated by the arrows. His 75 and
Asn 148 are directly involved, and a charge relay system
below the ring may also participate. From Putnam et al.
198a
A
B
A. Iron
Because the three-dimensional structures of the peroxi-
dase, its reductant cytochrome c, and the complex of
the two (Fig. 16-9) are known, cytochrome c peroxidase
is the subject of much experimental study. Other
fungal peroxidases, some of which contain manganese
rather than iron, act to degrade lignin (Chapter 25).
218
A lignin peroxidase from the white wood-rot fungus
Phanerochaete chrysosporium has a surface tryptophan
with a specifically hydroxylated C carbon atom
which may have a functional role in catalysis.
218a,b
The human body contains lactoperoxidase, a
product of exocrine secretion into milk, saliva, tears,
etc., and peroxidases with specialized functions in
saliva, the thyroid, eosinophils,
219
and neutrophils.
220
The functions are largely protective but the enzymes
also participate in biosynthesis. Mammalian peroxi-
dases have heme covalently linked to the proteins, as
indicated in Fig. 16-12.
220 222a
The active site structure of peroxidases (Fig. 16-13)
is quite highly conserved. As in myoglobin, an imida-
zole group is the proximal heme ligand, but it is usually
hydrogen bonded to an aspartate carboxylate as a
catalytic diad (Fig. 16-13).
223
In cytochrome c peroxidase
there is also a buried tryptophan, which has already
been highlighted in Fig. 16-9. A conserved and essen-
tial feature on the distal side is another histidine,
which is hydrogen bonded to an asparagine
224
and
which can also hydrogen bond to the substrate H
2
O
2
.
Fungal peroxidases also have a conserved arginine on
the distal side. However, even an octapeptide with a
bound heme cut from cytochrome c acts as a micro-
peroxidase with properties similar to those of natural
peroxidases.
225
Peroxidases and catalases contain high-spin Fe(III)
and resemble metmyoglobin in properties. The en-
zymes are reducible to the Fe(II) state in which form
they are able to combine (irreversibly) with O
2
. We
see that the same active center found in myoglobin
and hemoglobin is present but its chemistry has been
modified by the proteins. The affinity for O
2
has been
altered drastically and a new group of catalytic activities
for ferriheme-containing proteins has emerged.
Mechanisms of catalase and peroxidase
catalysis. Attention has been focused on a series
of strikingly colored intermediates formed in the
presence of substrates. When a slight excess of H
2
O
2
is added to a solution of horseradish peroxidase,
the dark brown enzyme first turns olive green as
compound I is formed, and then pale red as it turns
into compound II. The latter reacts slowly with sub-
strate AH
2
or with another H
2
O
2
molecule to regenerate
the original enzyme. This sequence of reactions is
indicated by the colored arrows in Fig. 16-14, steps ad.
854 Chapter 16. Transition Metals in Catalysis and Electron Transport
Titrations with such reducing agents as ferrocyanide
or K
2
IrBr
6
have established that compound I is con-
verted into compound II by a one-electron reduction
and compound II to free peroxidase by another one-
electron reduction. Thus, the iron in compound I may
formally be designated Fe(V) and that in II as Fe(IV).
However, this does not tell us whether or not the oxy-
gen atoms of H
2
O
2
are present in compounds I and II.
The enzyme in the Fe
3+
form can be reduced to Fe
2+
(Fig. 16-14, step e), as previously mentioned, and when
the Fe
2+
enzyme reacts with H
2
O
2
it is apparently con-
verted into compound II (Fig. 16-14, step f). This suggests
that the latter is an Fe
2+
complex of the peroxide anion.
Here, P
2
represents the porphyrin ring:
P
2
Fe
+
(II) OOH
However, spectroscopic evidence suggests that com-
pound II is a ferryl iron complex which could be
derived from the preceding structure by addition of
a proton and loss of water.
226,227
P
2
Fe
2+
(IV) = O
Compound II
High concentrations of H
2
O
2
convert II into com-
pound III, which is thought to be the same as the
oxyperoxidase that is formed upon addition of O
2
to
the Fe(II) form of the free enzyme (Fig. 16-14, step g)
and corresponds in structure to oxyhemoglobin.
228
Compound I was at one time thought to be a com-
plex of H
2
O
2
or its anion with Fe(III), but its magnetic
and spectral properties are inconsistent with this struc-
ture. Rather, it too appears to contain ferryl iron
HC
C
COOH
CH
3
N
N N
N
Fe (IV)
COOH
H
3
C
H
3
C
CH
3
CH
2
CH
2
O
+
Compound I. The unpaired electron and the
positive charge are delocalized over the
porphyrin ring and perhaps into the
proximal histidine ring
Figure 16-12 Linkage of heme to mammalian peroxidases.
There are two ester linkages to carboxylate side chains from
the protein.
220,221
Myeloperoxidase contains a third
linkage.
222,222a
C
C
COOH
CH
2
N
N N
N
Fe
COOH
H
2
C
H
2
C
CH
3
H
CH
2
O
C
O
Protein
O
C
O
Protein
Protein
H
H
3
C S
+
CH
3
Third linkage
in myeloperoxidase
Figure 16-13 The active site of yeast cytochrome c peroxi-
dase. Access for substrates is through a channel above the
front edge of the heme ring as viewed by the reader. A
pathway for entrance of electrons may be via Trp 191 and
His 175. From Holzbaur et al.
206
Based on coordinates of
Finzel et al.
215
bound to an electron-deficient porphyrin -cation
radical.
229
The reaction with peroxide probably involves
initial formation of a peroxide anion complex (Fig.
16-14, step a) which is cleaved with release of water
(step b).
215,230
The resulting Fe(V)
+
= O compound is
converted to compound I by transfer of a single elec-
Asn 82
Arg 48
His 52
His 175
Trp 191
Asp 235
Trp 51
855
with substrates in a two-electron process
232 235
with
transfer of an oxygen atom to the substrate, a charac-
teristic also of reactions catalyzed by cytochromes
P450 (see Eq. 18-57).
Haloperoxidases. Many specialized peroxidases
are active in halogenation reactions. Chloroperoxi-
dases from fungi
236,237
catalyze chlorination reactions
like that of Eq. 16-11 using H
2
O
2
and Cl

as well as the
usual peroxidase reaction.
Chloroperoxidase is isolated in a low-spin Fe(III)
state. The reduced Fe(II) enzyme is a high-spin form
(16-10)
Figure 16-14 The catalytic cycles and other reactions of peroxidases and
catalases. The principal cycle for peroxidases is given by the colored arrows.
That of catalases is smaller, making use of step a, b, and c, which is marked by
a light green line.
a
b
c
d
e
f
g
Ferric peroxide
e

Oxyperoxidase
(Compound III)
Compound I
+
or
H
2
O
Fast
AH
2
O
2
O
2
H
2
O
2
H
2
O
H
2
O
2
O
2
c
Catalase
AH
2
H
2
O
H
2
O
2
i
h
H
2
O
2

Compound II
Ferryl
Fe (IV)
O
Fe (V)
O
+
Fe (IV)
O
Fe (II)
H
2
O
Fe (II)
O
O
Fe (III)
O
OH
AH + H
+
AH + H
+
H
+
Fe (III)
+
H
+
tron from the porphyrin to the iron.
In cytochrome c peroxidase com-
pound I contains a free radical on
the nearby Trp 191 ring instead of
on the porphyrin radical.
216
Con-
sistent with this is the fact that
horseradish peroxidase contains
phenylalanine in place of Trp 191.
If we consider the fate of sub-
strate AH
2
during the action of a
peroxidase, we see that donation of
an electron to compound I to con-
vert it into II (Fig. 16-14, step c) will
generate a free radical AH as well
as a proton. The radical may then
donate a second electron to II to
form the free enzyme. Alternatively,
a second molecule of AH
2
may react
(Fig. 16-14, step c) to form a second
radical AH. The two AH radicals
may then disproportionate to form
A and AH
2
or they may leave the
enzyme and react with other mole-
cules in their environment. Com-
pound II of horse radish peroxidase
is able to exchange the oxygen atom
of its Fe(IV) = O center with water
rapidly at pH 7, presumably by
donation of a proton from the near-
by histidine side chain (correspond-
ing to His 52 of Fig. 16-13).
227,230a,b
This histidine presumably also
functions in proton transfer during
reactions with substrates (see Fig.
16-11B).
224
The catalase compound I
appears to be converted in a two-
electron reduction by H
2
O
2
directly
to free ferricatalase without inter-
vention of compound II (Fig. 16-14,
step c). The catalytic histidine
probably donates a proton to help
form water from one of the oxygen

P
+
Fe (IV) = O + H
2
O
2
H
2
O + O
2
+ P Fe (III)
+
Compound I
atoms of the H
2
O
2
. Nevertheless, compound II does
form slowly, especially if a slow substrate such as
ethanol is present. The previously mentioned bound
NADPH apparently reduces compound II formed in
this way, converting the inactivated enzyme back to
active catalase.
201,231
This may involve unusual one-
electron oxidation steps for the NADPH.
231
Under
some circumstances compound II of peroxidases reacts
A. Iron
856 Chapter 16. Transition Metals in Catalysis and Electron Transport
with spectroscopic properties similar to those of the
oxygenases of the cytochrome P450 family, which are
discussed in Chapter 18.
238,239
Like the cytochromes
P450 chloroperoxidase contains a thiolate group of a
cysteine side chain as the fifth iron ligand.
235,239,240
Chloroperoxidase forms compounds I and II, as do
other peroxidases. A chloride ion may combine with
compound I to form a complex of hypochlorite with
the Fe(III) heme.

P
+
Fe (IV) = O + Cl

P Fe (III)
+
OCl
This intermediate could then halogenate substrate
AH (Eq. 16-13, step a), lose HOCl (Eq. 16-13, step b),
or generate Cl
2
by reaction with Cl

(Eq. 16-13, step c).


These are all well-established reactions for the enzyme.
In each case the chlorine in the peroxidase complex can
be viewed as an electrophile which is transferred to an
attacking nucleophile. A fourth reaction that can go
through the same intermediate is conversion of alkenes
to ,-halohydrins (Eq. 16-14).
241
Lactoperoxidase of
milk, reacting with I

+ H
2
O
2
, promotes an analogous
iodination of tyrosine and histidine residues of proteins.
With radioactive
125
I

or
131
I

it provides a convenient
and much used method for labeling of proteins in
a
b
c
d
AH ACl
H
2
O HOCl
H
+
, Cl

Cl
2
P Fe(III) OCl P Fe(III) OH
P Fe(III)
H
+
H
2
O
O O
+ Cl

+ H
2
O
2
O O
+ H
2
O + OH

Cl
(16-11)
(16-12)
(16-13)
exposed surfaces of membranes (Eq. 16-15).
242
Iodi-
nated tyrosine derivatives are formed in the thyroid
gland by a similar reaction catalyzed by thyroid
peroxidase.
232
Even horseradish peroxidase can
oxidize iodide ions but neither it nor lactoperoxidase
will carry out chlorination or bromination reactions.
Myeloperoxidase, present in specialized lyso-
somes of polymorphonuclear leukocytes (neutro-
phils),
243
utilizes H
2
O
2
and a halide ion to kill ingested
bacteria.
244,245
Phagocytosis induces increased respira-
tion by the leukocyte and generation of H
2
O
2
, partly
by the membrane-bound NADPH oxidase described
in Chapter 18. Some of the H
2
O
2
is used by myeloper-
oxidase to attack the bacteria, apparently through
generation of HOCl by peroxidation of Cl

. Human
myeloperoxidase is a tetramer of two 466-residue
chains and two 108-residue chains, which carry the
covalently linked heme.
222a,246
Another oxygen-
dependent killing mechanism that may also be used
by neutrophils is the generation of the reactive singlet
oxygen.
247
This can occur by reaction of hypochlorite
with H
2
O
2
(Eq. 16-16) or from an enzyme-bound hypo-
chlorite intermediate such as that shown in Eq. 16-13.
Hereditary deficiency of myeloperoxidase is relatively
common.
245
OCl

+ H
2
O
2
O
2
(
1
g) + H
2
O + Cl

singlet O
2
Lactoperoxidase
248
and chloroperoxidase
249
also
generate singlet oxygen. The possible biological
significance is discussed in Chapter 18. Eosinophil
peroxidase appears to promote formation of hydroxyl
radicals.
250
Bromoperoxidases are found in many
red and green marine algae.
251
Many of them contain
vanadium and function by a mechanism different
than that used by heme peroxidases (see Section G).
252,253
Another related nonheme enzyme is the seleno-
protein glutathione peroxidase. It reacts by a mecha-
nism very different (Eq. 15-59) from those discussed
C C C C
X HO
H
2
O
2
H
2
O
X

+ H
+
(16-14)
HN
H C O
OH
HN
H C O
OH
I
I

, H
2
O
2
peroxidase
(16-15)
(16-16)
857
here, as does NADPH peroxidase, a flavoprotein
with a cysteine sulfinate side chain in the active
site.
254255a
A lignin-degrading peroxidase from the
white wood rot fungus Phanerochaete chrysosporium is
a simple heme protein,
256
while other peroxidases
secreted by this organism contain Mn.
257,258
7. The IronSulfur Proteins
Not all of the iron within cells is chelated by por-
phyrin groups. Hemerythrin (see Fig. 16-20) has been
known for many years, but the general significance
of nonheme iron proteins was not appreciated until
large-scale preparation of mitochondria was devel-
oped by Crane in about 1945. The iron content of
mitochondria was found to far exceed that of the
heme proteins present. In 1960, Beinert, who was
studying the mitochondrial dehydrogenase systems
for succinate and for NADH, observed that when the
electron transport chain was partially reduced by
these substrates and the solutions were frozen at low
temperature and examined, a strong EPR signal was
observed at g = 1.94. The signal was obtained only
upon reduction by substrate, and fractionation pointed
to the nonheme iron proteins. Six or more proteins of
this type are involved in the mitochondrial electron
transport chain (Eq. 18-1), and numerous others have
become recognized as members of the same large
family of ironsulfur proteins (Fe S proteins).
259,260
Ferredoxins, high-potential iron proteins, and
rubredoxins. The presence of nonheme iron proteins
is most evident in the anaerobic clostridia, which
contain no heme. It was from these bacteria that the
first Fe S protein was isolated and named ferredoxin.
This protein has a very low reduction potential of
E(pH 7) = 0.41 V. It participates in the pyruvate
ferredoxin oxidoreductase reaction (Eq. 15-35), in
nitrogen fixation in some species, and in formation
of H
2
. A small green-brown protein, the ferredoxin of
Peptococcus aerogenes contains only 54 amino acids but
complexes eight atoms of iron. If the pH is lowered
to ~ 1, eight molecules of H
2
S are released. Thus, the
protein contains eight labile sulfur atoms in an iron
sulfide linkage. There are also eight iron atoms.
Another group of related electron carriers, the
high-potential iron proteins (HIPIP) contain four
labile sulfur and four iron atoms per peptide chain.
261 266
X-ray studies showed that the 86-residue polypeptide
chain of the HIPIP of Chromatium is wrapped around
a single ironsulfur cluster which contains the side
chains of four cysteine residues plus the four iron and
four sulfur atoms (Fig. 16-15D).
261
This kind of cluster
is referred to as [4Fe 4S], or as Fe
4
S
4
. Each cysteine
sulfur is attached to one atom of Fe, with the four iron
atoms forming an irregular tetrahedron with an FeFe
distance of ~ 0.28 nm. The four labile sulfur atoms
(S
2
) form an interpenetrating tetrahedron 0.35 nm on
a side with each of the sulfur atoms bonded to three
iron atoms. The cluster is ordinarily able to accept
only a single electron. The ironsulfur cluster struc-
ture was a surprise, but after its discovery it was
found that ions such as [ Fe
4
S
4
(S CH
2
CH
2
COO

)
4
]
6
assemble spontaneously from their components and
have a similar cluster structure.
259,266a
Thus, living
things have simply improved upon a natural bonding
arrangement.
The bacterial ferredoxins from Peptococcus,
Clostridium (Fig. 16-16B),
267,268
Desulfovibrio, and other
anaerobes each contain two Fe
4
S
4
clusters with essen-
tially the same structure as that of the Chromatium
HIPIP.
267,269
Each cluster can accept one electron.
Much of the amino acid sequence in the first half of the
ferredoxin chain is repeated in the second half, sug-
gesting that the chain may have originated as a result
of gene duplication. Many invariant positions are
present in the sequence, including those of the cysteine
residues forming the Fe S cluster. Ferredoxins with
single Fe
4
S
4
clusters are also known.
270
The ferredoxins have reduction potentials E
(pH 7) from about 0.4 V to as low as 0.6 V. However,
the corresponding values for HIPIP proteins range
from + 0.05 to + 0.50 V at pH 7.
271
This wide range of
potentials initially seemed strange because the structures
of the active centers of both the clostridial ferredoxins
and the Chromatium HIPIP appear virtually identical.
272
Part of the explanation lies in the fact that Fe
4
S
4
clusters
can exist in three oxidation states (Eq. 16-17) that
differ, one from another, by a single electron.
273
A. Iron
S- Cys
S- Cys
Cys- S
Cys- S
Fe
Fe
Fe
S
S
S
S
Fe
Fe
4
S
4
or [4Fe4S]
Cys- S
Fe
S- Cys
S
Cys- S
S
S
S
Fe
Fe
Fe
3
S
4
or [3Fe4S]
S- Cys
S- Cys
Cys- S
Cys- S
Fe
Rubredoxin
FeS cluster
S- Cys Cys- S
Cys- S S- Cys
Fe Fe
S
S
Fe
2
S
2
or [2Fe2S]
Figure 16-15 Four different ironsulfur clusters of a type
found in many proteins. From Beinert
259
with permission.
C D
A B
858 Chapter 16. Transition Metals in Catalysis and Electron Transport
S6
S42
Me
S39
S9
S6
S42
Me
S39
S9
Here the charges shown are those on the cluster. The
cysteine ligands from the protein each add an addi-
tional negative charge. The Chromatium HIPIP and the
clostridial ferredoxins have the middle oxidation state
in common. The cluster is a little smaller in the more
oxidized states; in the Chromatium HIPIP the Fe Fe
distance changes from 0.281 to 0.272 nm upon oxidation.
Rubredoxins. The simplest of the Fe S proteins
are the rubredoxins. These proteins contain iron but
no labile sulfur. The rubredoxin of Clostridium pasteur-
ianum is a 54-residue peptide containing four cysteines
whose side chains form a distorted tetrahedron about
a single iron atom (Fig. 16-16A).
274
Not shown for any
(16-17)
[Fe
4
S
4
]
+
[Fe
4
S
4
]
2+
[Fe
4
S
4
]
3+
e e
Oxidized Fd;
reduced high
potential iron
protein
Reduced Fd Oxidized high
potential iron
protein; Super-
oxidized Fd
Figure 16-16 (A) Superimposed
stereoscopic -carbon traces of the
peptide chain of rubredoxin from
Clostridium pasteurianum with either
Fe
3+
(solid circles) or Zn
2+
(open
circles) bound by four cysteine side
chains. From Dauter et al.
274
(B)
Alpha-carbon trace for ferredoxin
from Clostridium acidurici. The two
Fe
4
S
4
clusters attached to eight cys-
teine side chains are also shown. The
open circles are water molecules.
Based on a high-resolution X-ray
structure by Due et al.
267
Courtesy of
E. D. Due. (C) Polypeptide chain of
a chloroplast-type ferredoxin from the
cyanobacteriumSpirulina platensis.
The Fe
2
S
2
cluster is visible at the top
of the molecule. From Fukuyama et
al.
276
Courtesy of K. Fukuyama.
C
A
B
859
(16-18)
of the structures in Fig. 16-16 are NHS hydrogen
bonds that connect backbone NH groups of the peptide
chain to the sulfur atoms of the cysteine groups, form-
ing the clusters.
275
These bonds may have important
effects on properties of the cluster. Rubredoxins also
participate in electron transport and can substitute for
ferredoxins in some reactions. Larger 14-kDa and 18-kDa
rubredoxins able to bind two iron ions participate in
electron transport in a hydroxylase system of Pseudomonas
(Chapter 18).
277
A smaller 7.9-kDa 2-Fe desulforedoxin
functions in sulfate-reducing bacteria.
278
Chloroplast-type ferredoxins. Members of a
large class of [2Fe 2S] or Fe
2
S
2
ferredoxins each con-
tain two iron atoms and two labile sulfur atoms with
the linear structure of Fig. 16-15B.
279,279a
Best known
are the chloroplast ferredoxins, which transfer electrons
from photosynthetic centers of chloroplasts to the flavo-
protein reductase that reduces NADP
+
to NADPH.
280,280a
The structure of a cyanobacterial protein of this type
is shown in Fig. 16-16C.
276
A second group of Fe
2
S
2
ferredoxins are found in bacteria including E. coli
281
and in human mitochondria.
282
For example, in
steroid hormone-forming tissues the ferredoxin
adrenodoxin
282a
carries electrons to cytochromes
P450. Its Fe
2
S
2
center receives electrons from adreno-
doxin reductase (Chapter 15 banner).
282b
The nitrogen-
fixing Clostridium pasteurianum also contains a
ferredoxin of this class.
283
In Pseudomonas putida
(Chapter 18) the related 106-residue putidaredoxin
transfers electrons to cytochromes P450.
284
The 3Fe 4S clusters. A 106-residue ferredoxin
from Azotobacter vinelandii contains seven iron atoms
in two Fe S clusters that operate at very different
redox potentials of 0.42 and + 0.32 V.
285
Other similar
seven-iron proteins are known.
286
From EPR measure-
ments it appeared that both clusters function between
the 2
+
and 3
+
(oxidized and superoxidized) states of
Eq. 16-17, despite the widely differing potentials. A
super-reduced all-Fe(II) form with E

(pH 7) = 0.70 V
can also be formed.
286
X-ray crystallographic studies
have revealed that the protein contains one Fe
4
S
4
cluster and one Fe
3
S
4
cluster (Fig. 16-15C).
286 288
The
structures and environments of the Fe
3
S
4
clusters are
similar to those of Fe
4
S
4
clusters but they lack one iron
and one cysteine side chain. An Fe
4
S
4
cluster may
sometimes lose S
2
to form an Fe
3
S
4
cluster such as the
one in the A. vinelandii ferredoxin. A less likely possi-
bility is isomerization to a linear Fe
3
S
4
structure.
289
Aconitase (Eq. 13-17) isolated under aerobic conditions
contains an Fe
3
S
4
cluster and is catalytically inactive.
Incubation with Fe
2+
activates the enzyme and recon-
verts the Fe
3
S
4
to an Fe
4
S
4
cluster (Fig. 13-4).
260,289,290
Properties of ironsulfur clusters. These
clusters were viewed for many years as unstable and
unable to exist outside of a protein. However, if pro-
tected from oxygen and manipulated in the presence
of soluble organic thiols they are stable and cofactor-
like.
260
Intact clusters can be extruded from proteins
by treatment with thiols in nonaqueous media. Both
Fe
4
S
4
and Fe
2
S
2
clusters as well as more complex forms
have been synthesized
291
and nonenzymatic cluster
interconversions have been demonstrated. Binding
to proteins stabilizes the clusters further, but some
(Fe S) proteins are labile and difficult to study. This
is evidently because of partial exposure of the cluster
to the surrounding solvent. Not only can O
2
cause
oxidation of exposed clusters
291a
but also superoxide,
292
nitric oxide, and peroxynitrite can react with the iron.
Aconitase has only three cysteine side chains available
for coordination with Fe and the protein is unstable.
Apparently, a superoxidized [Fe
4
S
4
]
3+
cluster is formed
in the presence of O
2
but loses Fe
2+
to give an [Fe
3
S
4
]
+
cluster.
293
Another interesting cluster conversion is the join-
ing of two Fe
2
S
2
clusters in a protein to form a single
Fe
4
S
4
cluster at the interface between a dimeric protein.
Such a cluster is present in the nitrogenase iron protein
(Fig. 24-2) and probably also in biotin synthase.
294
The
clusters in such proteins can also be split to release the
monomers.
Synthetic ironsulfur clusters have weakly basic
properties
273
and accept protons with a pK
a
of from
3.9 to 7.4. Similarly, one clostridial ferredoxin, in the
oxidized form, has a pK
a
of 7.4; it is shifted to 8.9 in the
reduced form.
295
If we designate the low-pH oxidized
form of such a protein as HOx
+
and the reduced form
as HRed, we can depict the reduction of each Fe
4
S
4
cluster as follows.
HOx
+
+ e

HRed
Comparing with Eq. 6-64 and using the Michaelis pH
functions (first two terms of Eq. 7-13) for HOx
+
and
HRed, it is easy to show that the value of E (E
1/ 2
) at
which equal amounts of oxidized protein (HOx
+
+ Ox)
and reduced protein (HRed + Red

) are present is
given by Eq. 16-19, in which K
ox
and K
red
are the K
a
values for dissociation of the protonated oxidized and
reduced forms, respectively.
E E
K
K
1/2
0 red
+
ox
+
= ( lowpH) + log
(1 + / [H ])
(1 + / [H ])
V 0 0592 .

(16-19)
Cys S
Fe
S Cys
S
S
Fe
S
S
Fe
S
S
Cys
Cys
A. Iron
860 Chapter 16. Transition Metals in Catalysis and Electron Transport
At the high pH limit this becomes E
1/ 2
= E (low pH) +
0.0592 (pK
ox
pK
red
) and V = 0.371 + 0.0592 (7.4 8.9)
V = 0.431 V. Thus, the value of E
1/ 2
changes from
0.371 to 0.460 V as the pH is increased. In the pH
range between the pK
a
values of 7.4 and 8.9 reduction
of the protein will lead to binding of a proton from the
medium and oxidation to loss of a proton. Human
and other vertebrate ferredoxins also show pH-depen-
dent redox potentials.
282
This suggests, as with the
cytochromes, a possible role of Fe S centers in the
operation of proton pumps in membranes. Neverthe-
less, many ferredoxins, such as that of C. pasteurianum,
show a constant value of E from pH 6.3 to 10
296
and
appear to be purely electron carriers.
Both the iron and labile sulfur can be removed
from Fe S proteins and the active proteins can often
be reconstituted by adding sulfide and Fe
2+
ions.
Using this approach, the natural isotope
56
Fe (nuclear
spin zero) has been exchanged with
57
Fe, which has a
magnetic nucleus
297
and
32
S has been replaced by
77
Se.
The resulting proteins appear to function naturally
and give EPR spectra containing hyperfine lines that
result from interaction of these nuclei with unpaired
electrons in the Fe
4
S
4
clusters (Fig. 16-17). These obser-
vations suggest that electrons accepted by Fe
4
S
4
clusters
are not localized on a single type of atom but interact
with nuclei of both Fe and S. The native proteins as
well as many mutant forms are being studied by
NMR
299,300
and other spectroscopic techniques (Fig.
16-18),
260
by theoretical computations,
301 304
and by
protein engineering and rational design.
305,306
Functions of ironsulfur enzymes. Numerous
ironsulfur clusters are present within the membrane-
bound electron transport chains discussed in Chapter
18. Of special interest is the Fe
2
S
2
cluster present in
a protein isolated from the cytochrome bc complex
(complex III) of mitochondria. First purified by Rieske
et al.,
307
this protein is often called the Rieske iron
sulfur protein.
308
Similar proteins are found in cyto-
chrome bc complexes of chloroplasts.
125,300,309,310
In
32
S
80
S
77
Se
Figure 16-17 Electron paramagnetic resonance spectrum
of the Fe S protein putidaredoxin in the natural form (
32
S)
and with labile sulfur replaced by selenium isotopes. Well-
developed shoulders are seen in the low-field end of the
spectrum of the
77
Se (spin = 1/ 2)-containing protein. From
Orme-Johnson et al.
298
Courtesy of W. H. Orme-Johnson.
Figure 16-18 Mssbauer X-ray absorption spectra of iron
sulfur clusters. (See Chapter 23 for a brief description of the
method.) Quadrupole doublets are indicated by brackets
and isomer shifts are marked by triangles. (A) [Fe
2
S
2
]
1+
cluster of the Rieske protein from Pseudomonas mendocina, at
temperature T = 200 K. (B) [Fe
3
S
4
]
1+
state of D. gigas ferre-
doxin II, T = 90 K. (C) [Fe
3
S
4
]
0
state of D. gigas ferredoxin II,
T = 15 K. (D) [Fe
4
S
4
]
2+
cluster of E. coli FNR protein, T = 4.2 K.
(E) [Fe
4
S
4
]
1+
cluster of E. coli sulfite reductase, T = 110 K.
From Beinert et al.
260
A
B
C
D
E
A
b
s
o
r
p
t
i
o
n
-2 0 2
Velocity (mm/s)
861
some bacteria Rieske-type proteins deliver electrons to
oxygenases.
300
The 196-residue mitochondrial protein
has an unusually high midpoint potential, E
m
of ~ 0.30 V.
The Fe
2
S
2
cluster, which is visible in the atomic struc-
ture of complex III shown in Fig. 18-8, is coordinated
by two cysteine thiolates and two histidine side chains.
In a Rhodobacter protein they occur in the following
conserved sequences:
311
C133-T-H-L-G-C138 and
C153-P-C-H-G-S158. One iron is bound by C133 and
C153 and the other by H135 and H156 as follows:
These proteins may also have an ionizable group with
a pK
a
of ~ 8.0, perhaps from one of the histidines that
is linked to the oxidationreduction reaction.
Ironsulfur clusters are found in flavoproteins
such as NADH dehydrogenase (Chapter 18) and
trimethylamine dehydrogenase (Fig. 15-9) and in the
siroheme-containing sulfite reductases and nitrite
reductases.
312
These two reductases are found both
in bacteria and in green plants.
Spinach nitrite reductase,
313
which
is considered further in Chapter 24,
utilizes reduced ferredoxin to carry
out a six-electron reduction of NO
2

to NH
3
or of SO
3
2
to S
2
. The 61-kDa
monomeric enzyme contains one
siroheme and one Fe
4
S
4
cluster. A
sulfite reductase from E. coli utilizes
NADPH as the reductant. It is a
large
8

4
oligomer.
312
The 66-kDa
chains contain bound flavin
Figure 16-19 Stereoscopic view
of E. coli assimilatory sulfite reduc-
tase. The siroheme (Fig. 16-6) is in
the center with one edge toward
the viewer and the Fe
4
S
4
cluster is
visible on its right side. A single
S atom from a cysteine side chain
bridges between the Fe of the
siroheme and the Fe
4
S
4
cluster.
A phosphate ion is visible in the
sulfite-binding pocket at the left
center of the siroheme. From Crane
et al.
312
Courtesy of E. D. Getzoff.
C133 S
Fe (III)
S C153
S
Fe (II)
N
N
S
N
N
H
H
H135
H156
(4 FAD + 4 FMN),
312,314,315
while the 64-kDa subunits
contain both siroheme and a neighboring Fe
4
S
4
cluster
(Fig. 16-19).
89,304,312,316
The iron of the siroheme and
the closest iron atom of the cluster are bridged by a
single sulfur atom of a cysteine side chain.
A somewhat similar double cluster is present in
all-Fe hydrogenases from Clostridium pasteurianum.
316a,b,c
These enzymes also contain two or three Fe
4
S
4
clusters
and, in one case, an Fe
2
S
2
cluster.
316a
At the presumed
active site a special H cluster consists of an Fe
3
S
4
cluster with one cysteine sulfur atom shared by an
adjoining Fe
2
S
2
cluster: Because many hydrogenases
contain nickel, their chemistry and functions are dis-
cussed in Section C,2.
The role of the ironsulfur clusters in many of the
proteins that we have just considered is primarily one
of single-electron transfer. The Fe S cluster is a place
for an electron to rest while waiting for a chance to
react. There may sometimes be an associated proton
pumping action. In a second group of enzymes,
exemplified by aconitase (Fig. 13-4), an iron atom of a
cluster functions as a Lewis acid in facilitating removal
of an OH group in an , dehydration of a carboxylic
acid (Chapter 13). A substantial number of other
bacterial dehydratases as well as an important plant
dihydroxyacid dehydratase also apparently use
Fe S clusters in a catalytic fashion.
317
Fumarases A
and B from E. coli,
317
L-serine dehydratase of a Pepto-
streptococcus species,
317 319
and the dihydroxyacid
A. Iron
The hydrogenase FeS H cluster
862 Chapter 16. Transition Metals in Catalysis and Electron Transport
dehydratase
320,321
may all use their Fe
4
S
4
clusters in a
manner similar to that of aconitase (Eq. 13-17). How-
ever, the Fe S enzymes that dehydrate R-lactyl-CoA to
crotonyl-CoA,
322
4-hydroxybutyryl-CoA to crotonyl-
CoA,
323,324
and R-2-hydroxyglutaryl-CoA to E-glutaco-
nyl-CoA
325
must act by quite different mechanisms,
perhaps similar to those utilized in vitamin B
12
-depen-
dent reactions. In these enzymes, as in pyruvate formate
lyase (Eq. 15-38), the Fe S center may act as a radical
generator.
The molybdenum-containing enzymes considered
in Section F also contain Fe S clusters. Nitrogenases
(Chapter 24) contain a more complex FeSMo cluster.
Carbon monoxide dehydrogenase (Section C) contains
2 Ni, ~ 11 Fe, and 14 S
2
as well as Zn in a dimeric
structure. In these enzymes the Fe S clusters appear
to participate in catalysis by undergoing alternate
reduction and oxidation.
For a few enzymes such as aconitase and
amido-transferases,
326
an FeS cluster plays a
regulatory role in addition to or instead of a catalytic
function.
327
Cytosolic aconitase is identical to iron
regulatory proteins 1 (IRP1), which binds to iron
responsive elements in RNA to inhibit translation of
genes asso-ciated with iron uptake. A high iron con-
centration promotes assembly of the Fe
4
S
4
cluster (see
Chapter 28, Section C,6). Another example is provided
by the E. coli transcription factor SOXR. This protein,
which controls a cellular defense system against
oxygen-derived superoxide radicals, contains two Fe
2
S
2
centers. Oxidation of these centers by superoxide
radicals appears to induce the transcription of genes
encoding superoxide dismutase and other proteins
involved in protecting cells against oxidative injury
(Chapter 18).
328 330
8. The (-oxo) Diiron Proteins
Both the ferroxidase center of ferritin (Fig. 16-4)
and the oxygen-carrying hemerythrin are members
of a family of diiron proteins with similar active site
structures.
331,332
The pair of iron atoms, with the
bridging () ligands such as O
2
, HO

, HOO

, O
2
2
, is
often held between four helices, as can be seen in
Fig. 2-22 and in Fig. 16-20C. In most cases each iron
is ligated by at least one histidine, one glutamate side
chain, and frequently a tyrosinate side chain. As many
as three side chains may bind to each iron. In addition,
one or two carboxylate groups from glutamate or
aspartate side chains bridge to both irons, as does the
-oxo group, which is typically H
2
O or

OH. Examples
of this structure are illustrated in Figs. 16-4 and 16-20.
Although the active sites of all of the diiron proteins
appear similar, the chemistry of the catalyzed reactions
is varied.
Hemerythrin. When both iron atoms are in the
Fe(II) state, hemerythrin, like hemoglobin, functions as
a carrier of O
2
. In the oxidized Fe(III) methemerythrin
form the iron atoms are only 0.32 nm apart. Three
bridging () groups lie between them: two carboxylate
groups and a single oxygen atom which may be either
O
2
or OH

. One coordination position on one of the


hexacoordinate iron atoms is open and appears to be
the site of binding of oxygen. The O
2
is thought to
accept two electrons, oxidizing the two iron atoms to
Fe(III) and itself becoming a peroxide dianion O
2
2
.
The process is completely reversible. The conversion
of the oxygen to a bound peroxide ion is supported by
studies of resonance Raman spectra (see Chapter 23)
which also suggest that the peroxide group is proto-
nated. In the diferrous protein the -oxo bridge is
thought to be an

OH group. Upon oxygenation the


proton could be shared with or donated to the peroxo
group (Eq. 16-20).
334
A similar binding of O
2
as a
peroxide dianion appears to occur in the copper-
containing hemocyanins (Section C,3).
Purple acid phosphatases. Diiron-tyrosinate
proteins with acid phosphatase activity occur in mam-
mals, plants, and bacteria. Most are basic glycoproteins
with an intense 510 - to 550-nm light absorption band.
Well-studied members come from beef spleen, from
the uterine fluid of pregnant sows (uteroferrin),
335
and from human macrophages and osteoclasts.
336336b
One of the two iron atoms is usually in the Fe(III)
oxidation state, but the second can be reduced to
Fe(II) by mild reductants such as ascorbate. This half-
reduced form is enzymatically active and has a pink
color and a characteristic EPR signal. Treatment with
oxidants such as H
2
O
2
or hexacyannoferrate (III)
Fe
II
His
His
O
O
Fe
II
His
His
O
O
O
H
His
Fe
III
His
His
O
O
Fe
III
His
His
O
O
O
His
O
O
H
Deoxyhemerythrin
O
2
Oxyhemerythrin (16-20)
863
generates purple inactive forms which lack a detectable
EPR signal.
337,338
The Fe
3+
of the active enzyme can
be replaced
339
with Ga
3+
and the Fe
2+
with Zn
2+
with
retention of activity; also, some plants contain phos-
phatases with Fe Zn centers.
340,341
The catalytic
mechanism resembles those of other metallophos-
phatases (Chapter 12) and the change of oxidation
state of the Fe may play a regulatory role. On the
other hand, a principal function of uteroferrin may
be in transplacental transport of iron to the fetus.
342
Diiron oxygenases and desaturases. In the
(-oxo) diiron oxygenases O
2
is initially bound in a
manner similar to that in hemerythrin but one atom
of the bound O
2
is reduced to H
2
O using electrons
supplied by a cosubstrate such as NADPH. The other
oxygen atom enters the substrate. This is illustrated
by Eq. 16-21 for methane monooxygenase.
332,343 345
A toluene monooxygenase has similar properties.
346
CH
4
+ NADH + H
+
+ O
2
CH
3
OH + NAD
+
+ H
2
O
In green plants a soluble
9
stearoyl-acyl carrier
protein desaturase uses O
2
and NADH or NADPH to
introduce a double bond into fatty acids. The structure
of this protein (Fig. 16-20B,C) is related to those of
methane oxygenase and ribonucleotide reductase.
333,347
The desaturase mechanism is discussed in Chapter 21.
9. Ribonucleotide Reductases
Ribonucleotides are reduced to the 2'-deoxyribo-
nucleotides (Eq. 16-21) that are needed for DNA
synthesis by enzymes that act on either the di- or tri-
phosphates of the purine and pyrimidine nucleo-
sides
348351
(Chapter 25). These ribonucleotide reductases
utilize either thioredoxin or glutaredoxin (Box 15-C)
as the immediate hydrogen donors (Eq. 16-22). The
pair of closely spaced SH groups in the reduced
thioredoxin or glutaredoxin are converted into a
disulfide bridge at the same time that the 2'-OH of
the ribonucleotide di- (or tri-) phosphate is converted
to H
2
O. While some organisms employ a vitamin B
12
-
(16-21)
A. Iron
E229
H232
H146
E196
E105
E143
E229
H232
E196
H146 E105
E143
His 101
His 73
His 77
Fe (1)
Asp 106
Glu 58
Fe (2)
His 54
His 25
O
Methemerythrin
(Fe
3+
/Fe
3+
)
Figure 16-20 (A) The active site of
hemerythrin showing the two iron atoms
(green) and their ligands which include
the oxo bridge and two bridging car-
boxylate groups. From Lukat et al.
193
The
active site is between four parallel helices
as shown in Fig. 2-22. (B) Stereoscopic
view of the backbone structure of a
9
stearoyl-acyl carrier protein desaturase
which also contains a diiron center.
Notice the nine-antiparallel-helix bundle.
The diiron center is between four helices
as in hemerythrin. (C) Stereoscopic view
of the diiron center of the desaturase.
333
(B) and (C) courtesy of Ylva Lindqvist.
A B
C
864 Chapter 16. Transition Metals in Catalysis and Electron Transport
dependent enzyme for this purpose, most utilize iron
tyrosinate enzymes (Class I ribonucleotide reductases).
These are two-protein complexes of composition
2

2
.
The enzyme from E. coli contains 761-residue chains
and 375-residue chains. That from Salmonella typh-
imurium
351a
is similar, as are corresponding mammalian
enzymes and a virus-encoded ribonucleotide reductase
formed in E. coli following infection by T4 bacterio-
phage.
352
In every case the larger
2
dimer, which is
usually called the R1 protein, contains the substrate
binding sites, allosteric effector sites, and redox-active
SH groups. Each chain is folded into an unusual
( /)
10
barrel.
353,354
As in the more familiar ( /)
8
barrels, the active site is at the N termini of the
strands.
Each polypeptide chain of the
2
dimer or R2
protein contains a diiron center which serves as a
free radical generator.
354a,b,c
A few bacteria utilize a
dimanganese center.
355
Oxygenation of this center is
linked to the uptake of both a proton and an electron
and to the removal of a hydrogen atom from the ring
of tyrosine 166 to form H
2
O and an organic radical
(Eq. 16-23):
356 360
2 Fe(II)
2+
+ O
2
+ H
+
+ e

+ Y122-OH
Fe(III)
3+
O
2
Fe(III)
3+
Y122 O + H
2
O
The tyrosyl radical is used to initiate the ribonucleotide
reduction at the active site in the R1 protein ~ 3.5 nm
away. The tyrosyl radical is very stable and was dis-
covered by a characteristic EPR spectrum of isolated
enzyme. Alteration of this spectrum when bacteria
were grown in deuterated tyrosine indicated that the
radical is located on a tyrosyl side chain and that the
spin density is delocalized over the tyrosyl ring.
361,362
Using protein engineering techniques the ring was
located as Tyr 122 of the E. coli enzyme. A few of the
resonance structures that can be used to depict the
radical are the following:
OH OH
OH
+
+
OH
+
+
, etc.
A chain of hydrogen-bonded side chains apparently
provides a pathway for transfer of an unpaired electron
from the active site to the Tyr 122 radical and from
there to the radical generating center.
363
The tyrosyl
radical can be destroyed by removal of the iron by
exposure to O
2
or by treatment of ribonucleotide
reductases with hydroxyurea, which reduces the
radical and also destroys catalytic activity:
A second group of ribonucleotide reductases
(Class II), found in many bacteria, depend upon the
cobalt-containing vitamin B
12
coenzyme which is
discussed in Section B. These enzymes are monomeric
or homodimeric proteins of about the size of the larger
subunits of the Class I enzymes. The radical gener-
ating center is the 5'-deoxyadenosyl coenzyme.
350,364,365
Class III or anaerobic nucleotide reductases are
used by various anaerobic bacteria including E. coli
when grown anaerobically
350,366 369a
and also by some
bacteriophages.
370
Like the Class I reductases, they
have an
2

2
structure but each subunit contains an
Fe
4
S
4
cluster which serves as the free radical genera-
tor,
369
that forms a stable glycyl radical at G580.
369a
In this respect the enzyme resembles pyruvate formate-
lyase (Eq. 15-40). As with other enzymes using Fe
4
S
4
clusters as radical generators, S-adenosylmethionine
is also required. All ribonucleotide reductases may
operate by similar radical mechanisms.
350,351
When a 2'-Cl or -F analog of UDP was used in
place of the substrate an irreversible side reaction
occurred by which Cl

or F

, inorganic pyrophos-
phate, and uracil were released.
349
When one of these
enzyme-activated inhibitors containing
3
H in the 3'
position was tested, the tritium was shifted to the 2'
position with loss of Cl

and formation of a reactive


3'-carbonyl compound (Eq. 16-24) that can undergo
elimination at each end to give an unsaturated ketone
which inactivates the enzyme. This suggested that the
Fe-tyrosyl radical abstracts an electron (through a
(16-23)
R(SH)
2
Thioredoxin
or glutaredoxin
RS
2
H
2
O
O
Base
OH OH
H
O
Base
OH H
H
PPOCH
2
PPOCH
2
(16-22)
O C
NH
2
N
H
OH
Hydroxyurea
865
chain of intermediate groups) from an SH group,
now identified as C439 in the E. coli enzyme
371372
(Fig. 16-21). The resulting thiyl radical is thought to
abstract a hydrogen atom from C-3 of a true substrate
to form a substrate radical (Eq. 16-25, step a) which,
with the help of the C462 SH group,
facilitates the loss of

OH from C-2 in
step b. The resulting C2 radical would
be reduced by the nearby redox-active
thiol pair C462 and C225 (Fig. 16-21
and Eq. 16-25, step c). In Eq. 16-25 the
reaction is shown as a hydride transfer
with an associated one-electron shift
but the mechanism is uncertain. In
step d of Eq. 16-25 the thiyl radical is
regenerated and continues to function
in subsequent rounds of catalysis. In
the final step (step e) the redox active pair is reduced
by reduced thioredoxin or glutaredoxin. The active
site must open to release the product and to permit
this reduction, which may involve participation of still
other SH groups in the protein.
O
CH
2
Base
HO OH
O P P
H H
S
Cys 439
O
CH
2
Base
HO HO
O P P
H
SH
Cys 439
H
S
Cys 462
S
Cys 225
O
CH
2
Base
O
O P P
SH
Cys 439
S
Cys 462
S
Cys 225
H
O
CH
2
Base
HO H
O P P
H
S
Cys 439
S
Cys 462
S
Cys 225
H
O
CH
2
Base
HO H
O P P
H H
S
Cys 439
S
Cys 462
S
Cys 225
S
Cys 225
H
S
Cys 462
H
S
S
TR
H

TR(SH)
2
a
b
c
d
e
O
CH
2
Uracil
HO Cl
O P P
O
CH
2
Uracil
O H
O P P
H
3
H
Cl

H
2'
Elimination
Enzyme inactivation (16-24)
A. Iron
S224
C462
C225
HS
H
O
B
HS
O
HO
NH
H
O
E441
C439
O
S
O
H
N437
PPO
O

3
2
4
Figure 16-21 (A) Scheme showing the diiron center of the R2 subunit of E. coli ribonucleotide reductase. Included are the side
chains of tyrosine 122, which loses an electron to form a radical, and of histidine 118, aspartate 237, and tryptophan 48. These
side chains provide a pathway for radical transfer to the R1 subunit where the chain continues to tyrosines 738 and 737 and
cysteine 429.
354a c
From Andersson et al.
354c
(B) Schematic drawing of the active site region of the E. coli class III ribonucleotide
reductase with a plausible position for a model-built substrate molecule. Redrawn from Lenz and Giese
373
with permission.
A B
(16-25)
866 Chapter 16. Transition Metals in Catalysis and Electron Transport
open for binding of

O
2

. As indicated in Eq. 16-27,


uptake of one proton is associated with each reaction
step. As illustrated in Fig 16-22 for step a of Eq. 16-27,
the first proton may be taken up to convert the bound

OH to H
2
O. The enzyme in the Fe
2+
form has a pK
a
of
8.5 that has been associated with tyrosine 34. Perhaps
this residue is involved in the proton uptake process.
378
A similarly located Tyr 41 from Sulfolobus is covalently
modified, perhaps by phosphorylation.
378a
B. Cobalt and Vitamin B
12
The human body contains only about 1.5 mg of
cobalt, almost all of it is in the form of cobalamin,
vitamin B
12
. Ruminant animals, such as cattle and
sheep, have a relatively high nutritional need for
cobalt and in regions with a low soil cobalt content,
such as Australia, cobalt deficiency in these animals is
a serious problem. This need for cobalt largely reflects
the high requirement of the microorganisms of the
rumen (paunch) for vitamin B
12
. All bacteria require
vitamin B
12
but not all are able to synthesize it. For
example, E. coli lacks one enzyme in the biosynthetic
Q69
O
N
H
O
Y34
N
N
H73
D156
H26
H160
N
H
N
N
N
H
O
O
2

M
O

H
H
H
+
H
+ 3
H
Figure 16-22 (A) Structure of the active site of iron superoxide dismutase from E. coli. From Carlioz et al.
379
Courtesy of
M. Ludwig. (B) Interpretive drawing illustrating the single-electron transfer from a superoxide molecule to the Fe
3+
of super-
oxide dismutase and associated proton uptake. Based on Lah et al.
376
A B
a
b
O
2

O
2
O
2

H
2
O
2
M
2+
M
3+
H
+
H
+
(16-27)
10. Superoxide Dismutases
Metalloenzymes of at least three different types
catalyze the destruction of superoxide radicals that
arise from reactions of oxygen with heme proteins,
reduced flavoproteins, and other metalloenzymes.
These superoxide dismutases (SODs) convert super-
oxide anion radicals

O
2

into H
2
O
2
and O
2
(Eq. 16-26).
The H
2
O
2
can then be destroyed by catalase (Eq. 16-8).
2 O
2

+ 2 H
+
H
2
O
2
+ O
2
The much studied Cu / Zn superoxide dismutase of
eukaryotic cytoplasm is described in Section D. How-
ever, eukaryotic mitochondria contain manganese
SOD and some eukaryotes also synthesize an iron-
containing SOD. For example, the protozoan Leishma-
nia tropica, which takes up residence in the phagolyso-
somes of a victims macrophages, synthesizes an iron-
containing SOD
374
to protect itself against superoxide
generated by the macrophages. Mycobacterium tuber-
culosis secretes an iron SOD which assists its survival
in living tissues and is also a target for the immune
response of human hosts.
375
Iron and manganese SODs have ~ 20-kDa subunits
in each of which a single ion of Fe or Mn is bound by
three imidazole groups and a carboxylate group.
376,377
The metal ion undergoes a cyclic change in oxidation
state as illustrated by Eq. 16-27. Notice that two protons
must be taken up for formation of H
2
O
2
. In Cu / Zn
SOD the copper cycles between Cu
2+
and Cu
+
. The
structure of the active site of an Fe SOD is shown in
Fig. 16-22. That of Mn SOD is almost identical.
376
In
addition to the histidine and carboxylate ligands, the
metal binds a hydroxyl ion

OH or H
2
O and has a site
(16-26)
867 B. Cobalt and Vitamin B
12
H
2
C
Bz
Co(III)
+
5-Deoxyadenosylcobalamin
OH
O
N
NH
2
N
N
N
HO
H
130 CN
Bz
Cyanocobalamin
Co(III)
+
pathway and must depend upon other bacteria to
complete the synthesis.
1. Coenzyme Forms
For several years after the discovery of cobalamin
its biochemical function remained a mystery, a major
reason being the extreme sensitivity of the coenzymes
to decomposition by light. Progress came after Barker
and associates discovered that the initial step in the
anaerobic fermentation of glutamate by Clostridium
tetanomorphum is rearrangement to -methylaspar-
tate
380,381
(Eq. 16-28).
The latter compound can be catabolized by reactions
that cannot be used on glutamate itself. Thus, the
initial rearrangement is an indispensable step in the
energy metabolism of the bacterium. A new coen-
zyme required for this reaction was isolated in 1958
after it was found that protection from light during
the preparation was necessary. The coenzyme was
characterized in 1961 by X-ray diffraction
382
as
5'-deoxyadenosylcobalamin. It is related to cyano-
cobalamin (Box 16-B) by replacement of the CN group
by a 5'-deoxyadenosyl group as indicated in the fol-
lowing abbreviated formulas.
383 385
Here the planes
represent the corrin ring system and Bz the dimethyl-
benzimidazole that is coordinated with the cobalt from
below the ring.
OOC
C
CH
2
CH
2
COO
H H
3
N
OOC
C
C
H H
3
N
+
l-Glutamate

COO
H
3
C H

threo--Methylaspartate (16-28)
The most surprising structural feature is the Co C
single bond of length 0.205 nm. Thus, the coenzyme
is an alkyl cobalt, the first such compound found in
nature. In fact, alkyl cobalts were previously thought
to be unstable. Vitamin B
12
contains Co(III), and cyano-
cobalamin can be imagined as arising by replacement
of the single hydrogen on the inside of the corrin ring
by Co
3+
plus CN

. However, bear in mind that three


other nitrogens of the corrin ring and a nitrogen of
dimethylbenzimidazole also bind to the cobalt. Each
nitrogen atom donates an electron pair to form coordi-
nate covalent linkages. Because of resonance in the
conjugated double-bond system of the corrin, all four
of the Co N bonds in the ring are nearly equivalent
and the positive charge is distributed over the nitrogen
atoms surrounding the cobalt.
The strength of the axial Co C bond is directly
influenced by the strength of bonding of the dimethyl-
imidazole whose conjugate base has a microscopic
pK
a
of 5.5.
386
Protonation of this base breaks its bond
to cobalt and may thereby strengthen the CoC bond.
387
Steric factors are also important in determining the
strength of this bond. NMR techniques are now
playing an important role in investigation of these
factors.
386 388
In both bacteria and liver, the 5'-deoxyadenosyl
coenzyme is the most abundant form of vitamin B
12
,
while lesser amounts of methylcobalamin are
present. Other naturally occurring analogs of the
coenzymes include pseudo vitamin B
12
which con-
tains adenine in place of the dimethylbenzimidazole.
Like dimethylbenzimidazole, it is combined with
ribose in the unusual linkage. A compound called
factor A is the vitamin B
12
analog with 2-methyladenine.
Related compounds have been isolated from such
sources as sewage sludge which
abounds in anaerobic bacteria. It
has been suggested that plants may
contain vitamin B
12
-like materials
which do not support growth of
bacteria. Thus, we may not have
discovered all of the alkyl cobalt
coenzymes.
CH
3
Bz
Methylcobalamin
Co(III)
+
868 Chapter 16. Transition Metals in Catalysis and Electron Transport
The story of vitamin B
12
began with pernicious
anemia, a disease that usually affects only persons
of age 60 or more but which occasionally strikes
children.
a
Before 1926 the disease was incurable
and usually fatal. Abnormally large, immature, and
fragile red blood cells are produced but the total
number of erythrocytes is much reduced from 4 6 x
10
6
mm
3
to 1 3 x 10
6
mm
3
. Within the bone marrow
mitosis appears to be blocked and DNA synthesis is
suppressed. The disease also affects other rapidly
growing tissues such as the gastric mucous mem-
branes (which stop secreting HCl) and nervous
tissues. Demyelination of the central nervous
system with loss of muscular coordination (ataxia)
and psychotic symptoms is often observed.
with enthusiasm. Twenty-two years later vitamin
B
12
was isolated (as the crystalline derivative cyano-
cobalamin) and was shown to be the curative agent.
It is present in liver to the extent of 1 mg kg
1
or
~10
6
M. Although much effort was expended in
preparation of concentrated liver extracts for the
treatment of pernicious anemia, the lack of an assay
other than treatment of human patients made
progress slow.
In the early 1940s nutritional studies of young
animals raised on diets lacking animal proteins and
maintained out of contact with their own excreta
(which contained vitamin B
12
) demonstrated the
need for animal protein factor which was soon
shown to be the same as vitamin B
12
. The animal
feeding experiments also demonstrated that waste
liquors from streptomyces fermentations used in
production of antibiotics were extremely rich in
vitamin B
12
. Later this vitamin was recognized as a
growth factor for a strain of Lactobacillus lactis which
responded with half-maximum growth to as little as
0.013 g / l (10
11
M).
In 1948, red cobalt-containing crystals of vitamin
B
12
were obtained almost simultaneously by two
pharmaceutical firms. Charcoal adsorption from
liver extracts was followed by elution with alcohol
and numerous other separation steps. Later fermen-
tation broths provided a richer source. Chemical
studies revealed that the new vitamin had an enor-
mous molecular weight, that it contained one atom
of phosphorus which could be released as P
i
, a
molecule of aminopropanol, and a ribofuranoside
of dimethyl benzimidazole with the unusual
configuration.
Note the relationship of the dimethylbenzimida-
zole to the ring system of riboflavin (Box 15-B).
Several molecules of ammonia could be released
from amide linkages by hydrolysis, but all attempts
to remove the cobalt reversibly from the ring system
were unsuccessful. The structure was determined
in 1956 by Dorothy C. Hodgkin and coworkers
using X-ray diffraction.
b
At that time, it was the
largest organic structure determined by X-ray dif-
fraction. The complete laboratory synthesis was
accomplished in 1972.
c
The ring system of vitamin B
12
, like that of
porphyrins (Fig. 16-5), is made up of four pyrrole
rings whose biosynthetic relationship to the corre-
sponding rings in porphyrins is obvious from the
structures. In addition, a number of extra methyl
groups are present. A less extensive conjugated
system of double bonds is present in the corrin ring
of vitamin B
12
than in porphyrins, and as a result,
many chiral centers are found around the periphery
BOX 16-B COBALAMIN (VITAMIN B
12
)
NH
2
O
H
B
8 H
3
C
5
Cyanocobalamin
M
r
= 1355
(C
63
H
58
N
14
PO
14
Co)
O
HO
N
CH
3
CH
3
N
H
O
O
CH
2
OH
P
H
3
C
H
HN
O
O

O
O
H
O
H
H
2
N
H
N
D
CH
3
CH
3
H
3
C
H
2
N
H
2
N
H
3
C
H
3
C
H
O
O
A
C
CN
Co
+
H
O
CH
3
CH
3
NH
2
N
N 1
2
3
19
18 17
15
N

13
10
NH
2
In 1926, Minot and Murphy discovered that
pernicious anemia could be controlled by eating
one-half pound of raw or lightly cooked liver per
day, a treatment which not all patients accepted
H
2
N
CH
2
C
CH
3
H
HO
O
HO
HO HO
N
N
CH
3
CH
3

-Ribofuranoside of dimethylimidazole R-1-Amino-2-propanol


869
BOX 16-B (continued)
of the somewhat nonplanar rings.
Cyanocobalamin, the form of vitamin B
12
isolated
initially, contains cyanide attached to cobalt. It occurs
only in minor amounts, if at all, in nature but is
generated through the addition of cyanide during
the isolation. Hydroxocobalamin (vitamin B
12a
)
containing OH

in place of CN

does occur in nature.


However, the predominant forms of the vitamin are
the coenzymes in which an alkyl group replaces the
CN

of cyanocobalamin.
Intramuscular injection of as little as 3 6 g of
crystalline vitamin B
12
is sufficient to bring about
a remission of pernicious anemia and 1 g daily
provides a suitable maintenance dose (often admin-
istered as hydroxocobalamin injected once every
2 weeks). For a normal person a dietary intake of
2 5 g / day is adequate. There is rarely any diffi-
culty in meeting this requirement from ordinary
diets. Vitamin B
12
has the distinction of being syn-
thesized only by bacteria, and plants apparently
contain none. Consequently, strict vegetarians some-
times have symptoms of vitamin B
12
deficiency.
Pernicious anemia is usually caused by poor
absorption of the vitamin. Absorption depends
upon the intrinsic factor, a mucoprotein (or muco-
proteins) synthesized by the stomach lining.
a,d f
Pernicious anemia patients often have a genetic
predilection toward decreased synthesis of the
intrinsic factor. Gastrectomy, which decreases syn-
thesis of the intrinsic factor, or infection with fish
tapeworms, which compete for available vitamin B
12
and interfere with absorption, can also induce the
disease. Also essential are a plasma membrane
receptor
g,h
and two blood transport proteins
transcobalamin
d,i
and cobalophilin. The latter
is a glycoprotein found in virtually every human
biological fluid and which may protect the vitamin
from photodegradation by light that penetrates
tissues.
j
A variety of genetic defects involving
uptake, transport, and conversion to vitamin B
12
coenzyme forms are known.
f,k
Normal blood levels of vitamin B
12
are
~ 2 x 10
10
M or a little more, but in vegetarians
the level may drop to less than one-half this value. A
deficiency of folic acid can also cause megaloblastic
anemia, and a large excess of folic acid can, to some
extent, reverse the anemia of pernicious anemia and
mask the disease.
a
Karlson, P. (1979) Trends Biochem. Sci. 4, 286
b
Hodgkin, D. C. (1965) Science 150, 979 988
c
Maugh, T. H., II (1973) Science 179, 266 267
d
Grsbeck, R., and Kouvonen, I. (1983) Trends Biochem. Sci. 8,
203 205
e
Allen, R. H., Stabler, S. P., Savage, D. G., and Lindenbaum, J.
(1993) FASEB J. 7, 1344 1353
f
Fenton, W. A., and Rosenberg, L. E. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 2 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 3129 3149,
McGraw-Hill, New York
g
Seetharam, S., Ramanujam, K. S., and Seetharam, B. (1992)
J. Biol. Chem. 267, 74217427
h
Birn, H., Verroust, P. J., Nexo, E., Hager, H., Jacobsen, C.,
Christensen, E. I., and Moestrup, S. K. (1997) J. Biol. Chem. 272,
26497 26504
i
Fedosov, S. N., Berglund, L., Nexo, E., and Petersen, T. E. (1999)
J. Biol. Chem. 274, 2601526020
j
Frisbie, S. M., and Chance, M. R. (1993) Biochemistry 32, 13886
13892
k
Rosenblatt, D. S., Hosack, A., Matiaszuk, N. V., Cooper, B. A.,
and Laframboise, R. (1985) Science 228, 1319 1321
2. Reduction of Cyanocobalamin and
Synthesis of Alkyl Cobalamins
Cyanocobalamin can be reduced in two one-
electron steps (Eq. 16-29).
385,388
The cyanide ion is lost
B. Cobalt and Vitamin B
12
in the first step (Eq. 16-29, step a), which may be accom-
plished with chromous acetate at pH 5 or by catalytic
hydrogenation. The product is the brown paramag-
netic compound B
12r
, a tetragonal low-spin cobalt(II)
complex. In the second step (Eq. 16-29, step b), an
additional electron is added, e.g., from sodium boro-
hydride or from chromous acetate at pH 9.5, to give
the gray-green exceedingly reactive B
12s
. The latter is
thought to be in equilibrium with cobalt(III) hydride,
as shown in Eq. 16-30, step a. The hydride is unstable
and breaks down slowly to H
2
and B
12r
(Eq. 16-30,
step b).
389
CN
Cyanocobalamin
Co(III)
+
a
b
CN

B
12r
or Cob(II)alamin
Co(II)
+
B
12s
or Cob(I)alamin
Co(I)
e

(16-29)
H
Co(III)
+
a
b
B
12s
H
+
B
12r
Slow
H
2
1/2
(16-30)
870 Chapter 16. Transition Metals in Catalysis and Electron Transport
O
H
2
C
Adenine
HO OH
H
+
OH
Co(III)
+
Hydroxocobalamin
C
C
C
C
H OH
H OH
H
H O
CH
2
Adenine
Co(III)
+
HO

(16-33)
C C
X H
C C
H X
(16-34)
Vitamin B
12s
reacts rapidly with alkyl iodides (e.g.,
methyl iodide or a 5'-chloro derivative of adenosine)
via nucleophilic displacement to form the alkyl cobalt
forms of vitamin B
12
(Eq. 16-31). These reactions
provide a convenient way of preparing isotopically
labeled alkyl cobalamins, including those selectively
enriched in
13
C for use in NMR studies.
390
The bio-
synthesis of 5'-deoxyadenosylcobalamin utilizes the
same type of reaction with ATP as a substrate.
391
AB
12s
adenosyltransferase catalyzes nucleophilic
displacement on the 5' carbon of ATP with formation
of the coenzyme and displacement of inorganic tri-
polyphosphate PPP
i
.
3. Three Nonenzymatic Cleavage Reactions
of Vitamin B
12
Coenzymes
The 5'-deoxyadenosyl coenzyme is easily decom-
posed by a variety of agents. Anaerobic irradiation
with visible light yields principally vitamin B
12r
and a
cyclic 5',8-deoxyadenosine which is probably formed
through an intermediate radical
391a
(Eq. 16-32):
Irradiation in the presence of air gives a variety of
products.
Hydrolysis of deoxyadenosylcobalamin by acid
(1 M HCl, 100, 90 min) yields hydroxycobalamin,
adenine, and an unsaturated sugar (Eq. 16-33). The
initial reaction step is thought to be protonation of the
oxygen of the ribose ring.
A related cleavage by alkaline cyanide can be
viewed as a nucleophilic displacement of the deoxy-
adenosyl anion by cyanide. The end product is
dicyanocobalamin, in which the loosely bound
nucleotide containing dimethyl benzimidazole is
replaced by a second cyanide ion. Methyl and other
simple alkyl cobalamins are stable to alkaline cyanide.
A number of other cleavage reactions of alkyl cobalamins
are known.
392,393
4. Enzymatic Functions of B
12
Coenzymes
Three types of enzymatic reactions depend upon
alkyl corrin coenzymes. The first is the reduction of
ribonucleotide triphosphates by cobalamin-dependent
ribonucleotide reductase, a process involving intermo-
lecular hydrogen transfer (Eq. 16-21). The second type
of reaction encompasses the series of isomerizations
shown in Table 16-1. These can all be depicted as in
Eq. 16-34. Some group X, which may be attached by a
C C, C O, or C N bond, is transferred to an adjacent
carbon atom bearing a hydrogen. At the same time,
CH
2
Co(III)
+
Co(I)
I

R
CH
2
R I
(16-31)
O
H
2
C
Co
Adenine
OH OH
O
H
2
C
Co
OH OH
N
N
N
N
NH
2
O
H
2
C
OH OH
N
N
N
N
NH
2
H
hv
(16-32)
871
Methylmalonyl-CoA mutase
Isobutyryl-CoA mutase catalyzes
the same type of reaction
TABLE 16-1
Isomerization Reactions Involving Hydrogen Transfer and Dependent upon a Vitamin B
12
Coenzyme
C C
X
H
H
3
C C
OH
OH
H
CH
3
C
O
H
H
2
C C
OH
H
H
H
2
N
H
3
C C
NH
2
OH
H
H
2
N COOH
H NH
2
H
3
C
COOH
H
2
N H H NH
2
OOC
C
C
H
3
N H
*
+

COO
H
3
C H
S
C
C

COO
H
3
C H
CoA
O
OOC
C
C

COO
H
3
C H
CH
2

CH
3
C
O
H
NH
3
l-Glutamate
threo-l--Methylaspartate
(R) Methylmalonyl-CoA
Transferred via
B
12
coenzyme
C C
H
X
H
2
C C
OH
H
H
HO
OOC
C
CH
2
C
COO
H
3
N H H H
*
+

S
C
CH
2
C
COO
H H

O
CoA
OOC
C
CH
2
CH
2
COO
CH
2

General reaction
Migrating group is enclosed in a box
Dioldehydratase
Glycerol dehydratase catalyzes the same
type of reaction
Ethanolamine ammonia-lyase
l--Lysine mutase
d--Lysine mutase and ornithine mutase
catalyze the same type of reaction
Glutamate mutase
-Methyleneglutarate mutase
the hydrogen is transferred to the carbon to which X
was originally attached. The third type of reaction is
the transfer of methyl groups via methylcobalamin
and some related bacterial metabolic reactions.
Cobalamin-dependent ribonucleotide reductase.
Lactic acid bacteria such as Lactobacillus leichmanni and
many other bacteria utilize a 5'-deoxyadenosylcobal-
amin-containing enzyme to reduce nucleoside triphos-
phates according to Eq. 16-21. Thioredoxin or
dihydrolipoic acid can serve as the hydrogen donor.
Early experiments showed that protons from water
are reversibly incorporated at C-2' of the reduced
B. Cobalt and Vitamin B
12
nucleotide with retention of configuration. A more
important finding was a large kinetic isotope effect of
1.8 when 3'-
3
H-containing UTP was reduced by the
enzyme.
394
Reaction of the reductase with dihydrolipoic acid
in the presence of deoxy-GTP, which apparently serves
as an allosteric activator, leads to formation, within a
few milliseconds, of a radical with a characteristic EPR
spectrum that can be studied when the reaction mix-
ture is rapidly cooled to 130K. When GTP (a true
substrate) is used instead of dGTP, the radical signal
reaches a maximum in about 20 ms and then decays.
Of the various oxidation states of cobalt (3+, 2+, and 1+)
872 Chapter 16. Transition Metals in Catalysis and Electron Transport
only the 2+ state of vitamin B
12r
is paramagnetic and
gives rise to an EPR signal. The electronic absorption
spectrum of the coenzyme of ribonucleotide reductase
is also changed rapidly by substrate in a way that
suggests formation of B
12r
. Thus, it was proposed
that a homolytic cleavage occurs to form B
12r
and a
stabilized 5'-deoxyadenosyl radical (Eq. 16-35).
395396a
However, H
3
is not transferred from the 3' position of
the substrate into the deoxyadenosyl part of the coen-
zyme.
394
The enzyme has many properties in common
with the previously discussed irontyrosinate ribo-
nucleotide reductases (Fig. 16-21) including limited
peptide sequence homology.
350
Stubbe and coworkers
suggested that the deoxyadenosyl radical is formed as
a radical chain initiator
394,396b
and that the mechanism
of ribonucleotide reduction is as shown in Eq. 16-25.
Studies of enzyme-activated inhibitors support this
mechanism.
364
The isomerization reactions. At least 10 reac-
tions of the type described by Eq. 16-34 are known
397
(Table 16-1). They can be subdivided into three
groups. First, X = OH or NH
2
in Eq. 16-34; isomeriza-
tion gives a geminal-diol or aminoalcohol that can
eliminate H
2
O or NH
3
to give an aldehyde. All of
these enzymes, which are called hydro-lyases or
ammonia-lyases, specifi-
cally require K
+
as well as
the vitamin B
12
coenzyme.
Second, X = NH
2
in Eq.
16-34; For this group of
aminomutases PLP is
required as a second
coenzyme. Third, X is
attached via a carbon
atom; the enzymes are
called mutases. Methyl-
malonyl-Co mutase is
required for catabolism of
propionate in the human
body, and is one of only
two known vitamin B
12
-
dependent enzymes. The related isobutyryl-CoA
mutase participates in the microbial synthesis of such
polyether antibiotics as monensin A.
397a,b
Other mutases
are involved in anaerobic bacterial fermentations.
In these reactions the hydrogen is always trans-
ferred via the B
12
coenzyme. No exchange with the
medium takes place. Since X may be an electronegative
group such as OH, the reactions could all be treated
formally as hydride ion transfers but it is more likely
that they occur via homolytic cleavages. Such cleavage
is indicated by the observation of EPR signals for
several of the enzymes in the presence of their sub-
strates.
398
Abeles and associates showed that when
dioldehydratase (Table 16-1) catalyzes the conversion
of 1,2-[1-
3
H]propanediol to propionaldehyde, tritium
appears in the coenzyme as well as in the final product.
When
3
H-containing coenzyme is incubated with
unlabeled propanediol, the product also contains
3
H,
which was shown by chemical degradation to be
exclusively on C-5'. Synthetic 5'-deoxyadenosyl coen-
zyme containing
3
H in the 5' position transferred
3
H to
product. Most important, using a mixture of propan-
ediol and ethylene glycol, a small amount of inter-
molecular transfer was demonstrated; that is,
3
H was
transferred into acetaldehyde, the product of dehy-
dration of ethylene glycol. Similar results were also
obtained with ethanolamine ammonia-lyase.
399
Another important experiment
398
showed that
18
O from [2-
18
O]propanediol was transferred into the 1
position without exchange with solvent. Furthermore,
18
O from (S)-[1-
18
O]propanediol was retained in the
product while that from the (R) isomer was not. Thus,
it appears that the enzyme stereospecifically dehydrates
the final intermediate. From these and other experiments,
it was concluded that initially a 5'-deoxyadenosyl
radical is formed via Eq. 16-35. This radical then
abstracts the hydrogen atom marked by a shaded box
in Eq. 16-36 to form a substrate radical and 5'-deoxy-
adenosine. One proposal, illustrated in Eq. 16-36, is
that the substrate radical immediately recombines with
Co(II)
+
CH
2
R
5-Deoxyadenosyl
radical
CH
2
5-Deoxyadenosylcobalamin
Co(III)
+
R
nucleoside triphosphate
Dihydrolipoic acid +
(16-35)
a c
d
CH
3
CH
H
C OH
OH
H
*
CH
2
Co
R
(5-deoxyadenosyl
coenzyme)
CH
3
CHO CH
2
H
2
O
HCH
2
R
(5-deoxyadenosine)
b
Co
H
C C
H
3
C
OH
H
HO*
Co
C C
H
3
C
OH H
*OH
H
3
C C C
OH
H
*
H
H
HO
(16-36)
873
the Co(II) of the coenzyme to form an organo-cobalt
substrate compound (step a of Eq. 16-36). The 5'-deoxy-
adenosine now contains hydrogen from the substrate;
because of rotation of the methyl group this hydrogen
becomes equivalent to the two already present in the
coenzyme. The substratecobalamin compound formed
in this step then undergoes isomerization, which, in
the case of dioldehydratase, leads to intramolecular
transfer of the OH group (step b). In step c the hydrogen
atom is transferred back from the 5'-deoxyadenosine
to its new location in the product and in step d the
resulting gem-diol is dehydrated to form the aldehyde
product.
401a
Carbocation, carboanion, and free radical inter-
mediates have all been proposed for the isomerization
step in the reaction. A carbocation would presumably
be cyclized to an epoxide which could react with the
B
12s
(Eq. 16-37, step b) to complete the isomerization.
At present most evidence favors, for the isomer-
ization reactions, an enzyme-catalyzed rearrangement
of the substrate radical produced initially during
formation of the 5'-deoxyadenosine (Eq. 16-38).
400401c
According to this mechanism the Co(II) of the B
12r
formed in Eq. 16-35 has no active role in the isomeriza-
tion and does not form an organocobalt intermediate
as in Eq. 16-36. Its only role is to be available to re-
combine with the 5'-deoxyadenosyl radical at the end
of the reaction sequence. Support for this interpreta-
tion has been obtained from study of model reactions
and of organic radicals generated in other ways.
400
Recently, EPR spectroscopy with
2
H- and
13
C-labeled
glutamates as substrates for glutamate mutase permitted
identification of a 4-glutamyl radical as a probable
intermediate for that enzyme.
402,402a,b
On the other hand, for methylmalonyl-CoA
mutase Ingraham suggested cleavage of the Co C bond
of an organocobalt intermediate to form a carbanion,
the substratecobalamin compound serving as a sort
of biological Grignard reagent. The carbanion
would be stabilized by the carbonyl group of the
thioester forming a homoenolate anion (Eq. 16-39).
The latter could break up in either of two ways reform-
ing a C Co bond and causing the isomerization.
403
Some experimental results also favor an ionic or organo-
cobalt pathway.
404
The three-dimensional structure of methylmalonyl-
CoA mutase from Propionibacterium shermanii shows
that the vitamin B
12
is bound in a base-off conformation
with the dimethylbenzimidazole group bound to the
protein far from the corrin ring (Fig. 16-23; see also Fig.
16-24).
405,405a
A histidine of the protein coordinates the
cobalt, as also in methionine synthase. The entrance
to the deeply buried active site is blocked by the coen-
zyme part of the substrate. The buried active site may
be favorable for free radical rearrangement reactions.
The structure of substrate complexes shows that the
coenzyme is in the cob(II)alamin (B
12r
) form with the
5'-deoxyadenosyl group detached from the cobalt,
rotated, shifted, and weakly bound to the protein.
405a
Side chains from neighboring Y89, R207, and H244 all
hydrogen bond to the substrate. The R207 guanidinium
group makes an ion pair with the substrate carboxylate,
and the phenolic and imidazole groups may have
catalytic functions.
405b,c,d,406
Studies are also in progress
on crystalline glutamate mutase.
406,406a
B. Cobalt and Vitamin B
12
a
b
C C
O
H
OH
H
3
C
H
Co(I)
C
C
OH
H
H
O
H
3
C
Co(III)
+
C
C
HO
H
OH
H
H
3
C
Co(III)
+
B
12s
(16-37)
C C
OH
H
HO
H
3
C
H
C C
H
H
OH
H
3
C
OH
*
*
(16-38)
Co(III)
+
C
H
2
C
C O
S
CoA
H
COOH
C H
2
C
C
S O
H
COOH
CoA

Co(III)
2+
C H
2
C
C
S O
H
COOH
CoA

Co(III)
+
(16-39)
874 Chapter 16. Transition Metals in Catalysis and Electron Transport
Figure 16-23 Three-dimensional
structure of methylmalonyl-CoA
mutase from Propionobacterium
shermanii. The B
12
coenzyme is
deeply buried, as is the active site.
A molecule of bound desulfo-
coenzyme A, a substrate analog,
blocks the active site entrance on
the left side. From Mancia et al.
405
Courtesy of Philip R. Evans.
C C
OH
CH
3
H
HO
H
H
C CHO
CH
3
H
H
2 1
(S)-1,2-Propanediol

*
*

(16-40)
C C
OH
CH
3
H
H
H
HO
*
(R)-1,2-Propanediol

C CHO
H
3
C
H
H
*
(16-41)
According to all of these mechanisms, 5'-deoxy-
adenosine is freed from its bond to cobalt during the
action of the enzymes. Why then does the deoxy-
adenosine not escape from the coenzyme entirely,
leading to its inactivation? Substrate-induced inacti-
vation is not ordinarily observed with coenzyme B
12
-
dependent reactions, but some quasi-substrates do
inactivate their enzymes. Thus, glycolaldehyde con-
verts the coenzyme of dioldehydratase to 5'-deoxy-
adenosine and ethylene glycol does the same with
ethanolamine deaminase. When 5'-deoxyinosine
replaces the 5'-deoxyadenosine of the normal coen-
zyme in dioldehydratase, 5'-deoxyinosine is released
quantitatively by the substrate. This suggests that the
dehydratase may normally hold the adenine group of
5'-deoxyadenosine through hydrogen bonding to the
amino group. Because the OH group of inosine tauto-
merizes to C O, inosine may not be held as tightly.
The deeply buried active site (Fig. 16-23) may also
prevent escape of the deoxyadenosine.
Despite the evidence in its favor, there was initially
some reluctance to accept 5'-deoxyadenosine as an
intermediate in vitamin B
12
-dependent isomerization
reactions. It was hard to believe that a methyl group
could exchange hydrogen atoms so rapidly. It was
suggested that protonation of the oxygen of the ribose
ring as in Eq. 16-33 might facilitate release of a hydrogen
atom. However, substitution of the ring oxygen by
CH
2
in a synthetic analog did not destroy the coenzy-
matic activity.
407
Another possibility is that the methyl
group has an unusual reactivity if the cobalt is reduced
to Co(II).
Stereochemistry of the isomerization reactions.
Dioldehydratase acts on either the (R) or (S) isomers of
1,2-propanediol (Eqs. 16-40 and 16-41; asterisks and
daggers mark positions of labeled atoms in specific
experiments).
In both cases the reaction proceeds with retention of
configuration at C-2 and with stereochemical speci-
ficity
408
for one of the two hydrogens at C-1. The
reaction catalyzed by methylmalonyl-CoA mutase
likewise proceeds with retention of configuration at
C-2 (Table 16-1)
409
but the glutamate mutase reaction
is accompanied by inversion (Eq. 16-28).
Aminomutases. The enzymes L--lysine mutase
(which is also D--lysine mutase) and D-ornithine
mutase catalyze the transfer of an -amino group to
an adjacent carbon atom
410
(Table 16-1). Two proteins
are needed for the reaction; pyridoxal phosphate is
required and is apparently directly involved in the
amino group migration. In the -lysine mutase the
6-amino group of L--lysine replaces the pro-S hydro-
gen at C-5 but with inversion at C-5 to yield (3S, 5S)-
3,5-diaminohexanoic acid.
411
A bacterial D-lysine
5,6-aminomutase interconverts D-lysine with 2,5-
diaminohexanoic acid.
411a
Another related enzyme
875
is L-leucine 2,3-aminomutase, which catalyzes the
reversible interconversion of L-leucine and -leucine.
410,412
It was reported to be present in plants and also in the
human body, but the latter could not be confirmed.
413
The interconversion of L--lysine and L--lysine is
catalyzed by a lysine 2,3-aminomutase found in certain
clostridia.
414-415a
This enzyme also requires pyridoxal
phosphate and catalyzes a reaction with the same
stereochemistry as that of -lysine mutase. However,
it does not contain vitamin B
12
but depends upon
S-adenosylmethionine (AdoMet) and an ironsulfur
cluster. The adenosyl group of AdoMet may function
in the same manner as does the deoxyadenosyl group
of the adenosylcobalamin coenzyme. In these mutases
a 5'-deoxyadenosyl radical may abstract a hydrogen
atom from the position of a Schiff base of PLP with
the amino acid substrate. The radical isomerizes (Eq.
16-42) and then accepts a hydrogen atom back from
the 5'-deoxyadenosine to complete the reaction. Its
properties suggest that lysine 2,3-aminomutase is
related to the pyruvate formate-lyase of E. coli (Eq.
15-37),
410
class I ribonucleotide reductases, and other
enzymes that act by homolytic mechanisms.
Transfer reactions of methyl groups. The
generation and utilization of methyl groups is a quan-
titatively important aspect of the metabolism of all
cells. As we have seen (Fig. 15-18), methyl groups can
be created by the reduction of one-carbon compounds
attached to tetrahydrofolic acid. Methyl groups of
methyltetrahydrofolic acid (N
5
-CH
3
THF) can then be
transferred to the sulfur atom of homocysteine to form
methionine (Eq. 16-43). The latter is converted to
S-adenosylmethionine, the nearly universal methyl
group donor for transmethylation reactions (Eq. 12-3).
In some bacteria, fungi, and higher plants, the methyl-
THF-homocysteine transmethylase does not depend
upon vitamin B
12
but is a metalloenzyme with zinc at
the active center.
416
However, human beings share
with certain strains of E. coli and other bacteria the
need for methylcobalamin.
The structure of the E. coli enzyme (Fig. 16-24)
shows methylcobalamin bound in a base-off confor-
mation, with histidine 759 of the protein replacing
dimethylbenzimidazole in the distal coordination
position on the cobalt. This histidine is part of a
sequence Asp-X-His-X-X-Gly that is found not only in
methionine synthase but also in methylmalonyl-CoA
mutase, glutamate mutase, and 2-methyleneglutarate
mutase. However, diol dehydratase lacks this sequence
and binds adenosylcobalamin with the dimethylbenz-
imidazolecobalt bond intact.
417
The coenzyme evidently functions in a cyclic
process. The cobalt alternates between the +1 and +3
oxidation states as shown in Eq. 16-43. The first indi-
cation of such a cyclic process was the report by
Weissbach that
14
C-labeled methylcobalamin could
be isolated following treatment of the enzyme with
such methyl donors as AdoMet and methyl iodide
B. Cobalt and Vitamin B
12
Figure 16-24 Stereoscopic views
of the active site of methionine
synthase from E. coli. Methylco-
balamin (black) is in the active site
with His 759 of the protein in the
distal position of the coenzyme in a
base-off conformation. The dimethyl-
benzimidole nucleotide has been
omitted for clarity. Notice the
hydrogen-bonded His 759 D757
S810 triad. From Jarret et al.
418a
Courtesy of R. G. Matthews.
(16-42)
N
HC
OH
CH
3
N
CH COO CH R
N
CH
OH
CH
3
N
CH COO CH R
N
CH
OH
CH
3
N
CH COO CH R

O P

O P
O P
a
b
876 Chapter 16. Transition Metals in Catalysis and Electron Transport
lating agents such as AdoMet. At the conclusion of the
reaction in Eq. 16-44 the cobalt is in the +3 state. To be
remethylated, it must presumably be reduced to Co(II).
A second methyl group can be transferred by the same
type of reaction to form (CH
3
)
2
Hg.
Methylation of arsenic is an important pollution
problem because of the widespread use of arsenic
compounds in insecticides and because of the presence
of arsenate in the phosphate used in household deter-
gents.
421,422
After reduction to arsenite, methylation
occurs in two steps (Eq. 16-45). Additional reduction
steps result in the formation of dimethylarsine, one
of the principal products of action of methanogenic
bacteria on arsenate. The methyl transfer is shown as
occurring through CH
3
+
, with an accompanying loss of
a proton from the substrate. However, a CH
3
radical
may be transferred with formation of a cobalt(II)
corrinoid.
423
Corrinoid-dependent synthesis of acetyl-CoA.
The anaerobic bacterium Clostridium thermoaceticum
obtains its energy for growth by reduction of CO
2
with
hydrogen (Eq. 16-46). One of the CO
2
molecules is
reduced to formate which is converted via 5-methyl-THF
to the methyl corrinoid 5-methoxybenzimidazolyl-
(16-45)
O
As
HO
O
OH
HO
As
HO
O O As O
HO
As
O
CH
3
O
H
3
C
As
O
CH
3
O

Arsenate

Arsenite
H
3
C
As
CH
3
H

Methylarsonate

Dimethylarsinate
Dimethylarsine
2e

H
2
O
CH
3
+
H
+
H
2
O
+
CH
3
+
2e

4e

5 H
+
2 H
2
O
H
S COO
H
3
N H

+
H
3
C
THF N
5
CH
3
THF
HS COO
H
3
N H

+
Homocysteine
a
b
c
d
CH
3
Co(III)
+
N
N
O O
H

Co(I)
N
N
O O
H

Co(II)
+
e

Ado-Hcy e

AdoMet
+
H
+
H
+
(16-43)
Co(III)
+
HO
Co(III)
+
Hg
2+
CH
3
H
2
O
CH
3
Hg
+
+ H
+
(16-44)
after reduction (e.g., with reduced riboflavin phosphate).
The sequence parallels that of Eq. 16-31 for the labora-
tory synthesis of methylcobalamin. Nevertheless, the
transmethylase demonstrates some complexities.
Initially, it must be activated by AdoMet or methyl
iodide, after which it cycles according to Eq. 16-43,
steps a and b, but is gradually inactivated. This appar-
ently happens by oxidation to a Co(II) form of the
enzyme (Eq. 16-43, step c) that must be reductively
methylated with AdoMet and reduced flavodoxin
(step d) to regenerate the active form.
418
It is also
possible that the methyl group is not transferred as a
formal CH
3
+
as pictured in Eq. 16-43 but as a

CH
3
radical generated by homolytic cleavage of methyl-
cobalamin to cob(II)alamin. Whatever the mechanism,
a chiral methyl group is transferred from 5-methyl-
THF to homocysteine with overall retention of config-
uration.
419
Another important group of methyl transfer reac-
tions are those from methyl corrinoids to mercury, tin,
arsenic, selenium, and tellurium. For example, Eq.
16-44 describes the methylation of Hg
2+
. These reactions
are of special interest because of the generation of toxic
methyl and dimethyl mercury and dimethylarsine.
Notice that whereas in Eq. 16-43 the methyl group
is transferred as CH
3
+
by nucleophilic displacement on
a carbon atom, the transfer to Hg
2+
in Eq. 16-44 is that
of a carbanion, CH
3

, with no valence change occurring


in the cobalt. However, it is also possible that transfer
occurs as a methyl radical.
420
Methyl corrinoids are
able to undergo this type of reaction nonenzymatically,
and the ability to transfer a methyl anion is a property
of methyl corrinoids not shared by other transmethy-
877
obtained the Nobel Prize, his first reports were greeted
with skepticism and outright disbelief. The presence
of two atoms of nickel in each molecule of urease
435
was not discovered until 1975. The metal ions had
been overlooked previously, despite the fact that the
absorption spectrum of the purified enzyme contains
an absorption tail extending into the visible region
with a shoulder at 425 nm and weak maxima at 725
and 1060 nm. Urease catalyzes the hydrolytic cleavage
of urea to two molecules of ammonia and one of bicar-
bonate and is useful in the analytical determination of
urea.
Jack bean urease is a trimer or hexamer of identi-
cal 91-kDa subunits while that of the bacterium Kleb-
siella has an (
2

2
)
2
stoichiometry. Nevertheless, the
enzymes are homologous and both contain the same
binickel catalytic center (Fig. 16-25).
435 437a
The three-
dimensional structure of the Klebsiella enzyme revealed
that the two nickel ions are bridged by a carbamyl
group of a carbamylated lysine. Like ribulose bisphos-
phate carboxylase (Fig. 13-10), urease also requires
CO
2
for formation of the active enzyme.
438
Formation
of the metallocenter also requires four additional
proteins, including a chaperonin and a nickel-binding
protein.
438,439
The mechanism of urease action is probably related
to those of metalloproteases such as carboxypeptidase
A (Fig. 12-16) and of the zinc-dependent carbonic
(16-46)
cobamide. The methyl group of the latter
4 H
2
+ 2 CO
2
CH
3
COO

+ H
+
+ 2 H
2
O
G (pH 7) = 94.9 kJ mol
1
combines with another CO
2
to form acetyl-CoA. The
process, which requires the nickel-containing carbon
monoxide dehydrogenase, is discussed in Section C.
C. Nickel
It was not until the 1970s that nickel was first
recognized as a dietary essential for animals.
424 426
Nickel-deficient chicks grew poorly, had thickened
legs, and developed dermatitis. Tissues of deficient
animals contained swollen mitochondria and swollen
perinuclear space suggesting a function for Ni in
membranes. However, doubts have been raised about
the conclusions based on these experiments.
427
Within
tissues the nickel content ranges from 1 to 5 g / l.
Some of the metal in serum is present as complexes
of low molecular mass and some is bound to serum
albumin
428
and to a specific nickel-containing protein of
the macroglobulin class, known as nickeloplasmin.
429
Nickel is also present in plants; in some, e.g., Allysum,
it accumulates to high concentrations.
430
It is essential
for legumes and possibly for all plants.
431
Nickel
uptake proteins have been identified in bacteria and
fungi.
432,432a
Because of its ubiquitous occurrence it is
difficult to prepare a totally Ni-free diet. The acute
toxicity of orally ingested Ni(II) is low, and homeostatic
mechanisms exist in the animal body for regulating its
concentration. However, the volatile nickel carbonyl
Ni(CO)
4
is very toxic
426
and Ni from jewelry is a common
cause of dermatitis.
428,433
In its compounds nickel usually has the +2 oxi-
dation state but the +3 and +4 states occur rarely in
complexes. The Ni
2+
ion contains eight 3d electrons,
a configuration that favors square-planar coordination
of four ligands. However, the ion is also able to form a
complex with six ligands and an octahedral geometry.
It has been suggested that this ambivalence may be
of biochemical significance.
Nickel is found in at least four enzymes: urease,
certain hydrogenases, methyl-CoM reductase (in its
cofactor F
430
) of methanogenic bacteria, and carbon
monoxide dehydrogenase of acetogenic and metha-
nogenic bacteria.
434
1. Urease
Urease, which was first isolated from the jack bean
has a special place in biochemical history as the first
enzyme to be crystallized. This was accomplished by
J. B. Sumner in 1926, and although Sumner eventually
Figure 16-25 The active site of urease showing the two
Ni
+
ions held by histidine side chains and bridged by a
carbamylated lysine (K217*). A bound urea molecule is
shown in green. It has been placed in an open coordination
position on one nickel and is shown being attacked for
hydrolytic cleavage by a hydroxyl group bound to the other
nickel. Based on a structure by Jabri et al.
436
and drawing by
Lippard.
437
D360
H272
H134
H136
H246
K217*
N
H
N
N
O
O
H
N
C
O
H
H
H
N
N
NH
N
NH
O
O
O
H
HN
N
Ni
Ni
2+

H
C. Nickel
878 Chapter 16. Transition Metals in Catalysis and Electron Transport
C
O
NH
2
H
2
N
C
NH
2
H
2
N
HO
O
H
Urea
C
O
NH
2
O
HO

NH
3
H
2
O CO
2
NH
3
Carbamate
(16-47)
anhydrase (Fig. 13-1). In Fig. 16-25 one nickel ion is
shown polarizing the carbonyl group, while the second
provides a bound hydroxyl ion that serves as the
attacking nucleophile. A probable intermediate
product is a carbamate ion (Eq. 16-47).
Urease is an essential enzyme for bacteria and
other organisms that use urea as a primary source of
nitrogen. The peptic ulcer bacterium Helicobacter
pylori uses urease to hydrolyze urea in order to defend
against the high acidity of the stomach.
439a
The enzyme
is also present in plant leaves and may play a necessary
role in nitrogen metabolism.
431
In nitrogen-fixing
legumes urea derivatives, the ureides, have an impor-
tant function (Chapter 25) but urease may not be
involved in the catabolism of these compounds.
440
2. Hydrogenases
Many plants, animals, and microorganisms are able
to evolve H
2
by reduction of hydrogen ions (Eq. 16-48)
or to oxidize H
2
by the reverse of this reaction.
441,442
2H
+
+ 2e

H
2
Hydrogenases have been classified into two main
types: Fe-hydrogenases, which contain iron as the
only metal,
443
and Ni-hydrogenases, which contain
both iron and nickel.
444
In a few Ni-hydrogenases a
selenocysteine residue replaces a conserved cysteine
side chain.
445,446
Fe-hydrogenases are often extremely
active and are utilized to rid organisms of an excess of
electrons by evolution of H
2
. Since they may also be
used to acquire electrons by oxidation of H
2
, they are
often described as bidirectional. Ni-hydrogenases, as
well as some Fe-hydrogenases, are involved primarily
in uptake of H
2
.
447
All hydrogenases contain one or
more Fe S centers in addition to the H
2
-forming
catalytic center.
441
Some hydrogenases are membrane
bound and are often coupled through unidentified
carriers to formate dehydrogenase (Chapter 17). In
the strict anaerobes such as clostridia, hydrogenases
are linked to ferredoxins. Hydrogenases are inactivated
readily by O
2
, which oxidizes the catalytic centers but
can sometimes be reactivated by treatment with reduc-
ing agents.
448
The 60-kDa all-iron monomeric hydrogenase I of
Clostridium pasteurianum (mentioned on p. 861) contains
ferredoxin-like Fe
4
S
4
clusters plus additional Fe and
sulfur atoms organized as a special H cluster. The EPR
spectrum of the catalytic center, recognized because the
spectrum is altered by the binding of carbon monoxide,
is unusual. Its g values of 2.00, 2.04, and 2.10 are similar
to those of oxidized high-potential iron proteins.
449
The
C. pasteurianum hydrogenase II is a 53-kDa monomer con-
taining eight Fe and eight S
2
ions. These are organized
into one ferredoxin-like Fe
4
S
4
cluster plus a three-Fe
cluster and one iron ion in a unique environment.
449
In contrast to these iron-only hydrogenases, the
large periplasmic hydrogenase of Desulfovibrio gigas
consists of one 28-kDa subunit and one 60-kDa sub-
unit and contains two Fe
4
S
4
clusters, one Fe
3
S
4
cluster,
and another dimetal center containing a single atom of
Ni.
450,451
These are seen clearly in the three-dimensional
structure depicted in Fig. 16-26. The three Fe S clusters,
at 5 - to 6-nm intervals, form a chain from the external
surface to the deeply buried nickel iron center.
450453a
A plausible pathway for transport of protons to the
active center can also be seen.
450,451
The nickel center
also contains an atom of Fe. Four cysteine side chains
participate in forming the Ni Fe cluster, two of them
provide sulfur atoms that bridge between the metals
while two others are ligands to Ni (Fig. 16-26).
(16-48)
a b
c
d
H
2
C
S
Ni(III)
S
Fe(II)
S CH
2
Cys 530
CH
2
Cys 65
S
Ni(II)
S
Fe(II)
S CH
2
Cys 530
CH
2
Cys 65

H
2
H
+
H
+
e

2H
+
S
Ni(II)
S
Fe(II)
S CH
2
Cys 530
CH
2
Cys 65

+
H
H
S
Ni(II)
S
Fe(II)
S CH
2
Cys 530
CH
2
Cys 65
H
H
+

(16-49)
879
A hydrogenase from chromatium vinosum has a similar
structure.
453b
There are still uncertainties about other
nonprotein ligands such as H
2
O.
452453a
All of the Ni-hydrogenases display an EPR signal
that can be assigned to Ni(III).
452
However, the active
enzyme from D. gigas contains Ni(II). A proposed
mechanism
452
is indicated in Eq. 16-49. Step a of this
equation is a reductive activation. In step b a molecule
of H
2
is bound as a hydride ion on Ni and a proton on
a nearby sulfur. Protonation of a second sulfur ligand
to Ni is needed to promote the cleavage of H
2
prior to
the two-step oxidation of the bound H

. One of two Ni-


containing hydrogenases of Methanobacterium thermo-
autotrophicum contains FAD as well as FeS clusters.
454
It specifically reduces the 5-deazaflavin cofactor F
420
(Chapter 15). A major function of this deazaflavin is
reduction of the nickel-containing cofactor F
430
.
3. Cofactor F
430
and Methyl-Coenzyme
M Reductase
Cofactor F
430
is a nickel tetrapyrrole with a structure
(Fig. 16-27)
455,456
similar to those of vitamin B
12
and of
siroheme. The tetrapyrrole ring is the most highly
reduced in cofactor F
430
, which functions in reduction
of methyl-CoM to methane in methanogens (Fig. 16-28).
The methyl CoM reductase of Methanobacterium
thermoautotrophicum is a large 300-kDa protein with
subunit composition
2

2
and containing two mole-
cules of bound F
430
. The nickel in F
430
is first thought
to be reduced, in an activation step, to Ni(I),
456a
which
may attack the methyl group of methyl-CoM homolyt-
ically to yield a methyl nickel complex and a sulfur
radical.
457,458
Alkyl nickel compounds react with
protons, and in this case they would yield methane
and would regenerate the Ni(II) form of the cofactor.
The CoM radical could be reduced back to free CoM.
High-resolution crystal structures of the enzyme
in two inactive Ni(II) forms
458
show the two F
430
mole-
cules. Each is bound in an identical channel about 3
nm in length and extending from the surface deep into
the interior of the protein. The F
430
lies at the bottom
of this channel with its nickel atom coordinated with
the oxygen atom of a glutamine side chain. In one
form CoM lies directly above the nickel, with its
C. Nickel
Figure 16-26 (A) Stereo-
scopic view of the structure
of the Desulfovibrio gigas
hydrogenase as an -carbon
plot. The electron density
map at the high level of
8 is superimposed and
consists of dark spheres
representing the Fe and Ni
atoms. The iron atoms of
the two Fe
4
S
4
and one Fe
3
S
4
clusters are seen clearly
forming a chain from the
surface of the protein to
the Ni Fe center. (B) The
structure of the active site
Ni Fe pair. The two metals
are bridged by two cysteine
sulfur atoms and an uniden-
tified atom, perhaps O, and
the nickel is also coordinated
by two additional cysteine
sulfurs. Unidentified small
molecules L1, L2, and L3
are also present. From
Volbeda et al.
453
Courtesy
of M. Frey.
A
B
880 Chapter 16. Transition Metals in Catalysis and Electron Transport
Figure 16-27 (A) Structure of the nickel-containing prosthetic group F
430
as isolated in the esterified (methylated) form. From
Pfaltz et al.
459
The front face, which reacts with methyl-coenzyme M, is toward the reader.
458
(B) Structure of a representative
member of a family of tunichlorins isolated from marine tunicates.
460
For tunichlorin R = R = H. Related compounds have
R = CH
3
and/or R = an alkyl group with 13 21 carbon atoms and up to six double bonds.
N
Ni
N
N N
O
COOH
COOH
COOH
CH
3
H
HOOC
HN
H
H
3
C
C
H
O
COOH
O
H
2
N
+
Cofactor F
430
N
Ni
N
N N
CH
3 H
3
C
CH
3
O
CH
3
H
3
C
C
O O R'
O
R
1
2 3
4
5
10
7
8

Tunichlorins
a
b
c
d
e
Ni(III)
Ni(II)
Ni(I) Ni(I)
HTP
S
S
CoM
CH
4
Ni(I)
CH
4
S
S
O
3
S
HTP

HO
3
P
O
H
3
C H
NH
COO
H
O

SH

(HTP-SH)
H
3
C
S
SO
3

Methyl-CoM
HTP
SH
S
SO
3
CH
3

HTP
S
S
SO
3
2
CH
3

H
HTP
S
S
SO
3
CH
3

Figure 16-28 Proposed mechanism of action


of the methane-forming coenzyme M reductase.
Based on the crystal structure.
A B
881
thiolate sulfur providing the sixth ligand for the
nickel. The long SH-containing side chain of
heptanoyl threonine phosphate (HTP; also called
coenzyme B; Chapter 15, Section E) also lies within
the channel with its amino acid head group blocking
the entrance. In a second crystal form the mixed
disulfide HTP S S CoM, an expected product of
the reaction (Fig. 16-28), is present in the channel.
458
Because of the distance from the SH group of HTP
and the nickel atom it is clear that there must be some
motion of the methyl-CoM and that the methane
formed may stay trapped in the active site until the
HTPSSCoA product leaves.
A proposed mechanism for the catalytic cycle
based on the X-ray results as well as previous chemical
studies and EPR spectroscopy is shown in Fig. 16-28.
The substrates enter in step a. The position of the HTP,
with its extended side chain, is probably the same as
that seen in the X-ray structures of the Ni(II) complexes
but the conformation of the methyl-CoM is different.
The methyl transfer in step b is reminiscent of that of
methionine synthase (Eq. 16-43). Although the distance
from the CoM sulfur and the HTP SH is too great for
direct proton transfer between the two, as indicated
for step b, there are two tyrosine hydroxyls that could
provide a pathway for proton transfer. The region
around the surface of the nickel coenzyme is largely
hydrophobic and could facilitate formation of the
thiyl radical in step c. In the structure with the bound
HTPSSCoM heterodisulfide an oxygen atom of the
sulfonate group of CoM is bonded tightly to the Ni(II).
However, in the active Ni(I) form the nickel is nucleo-
philic and would probably repel the sulfonate, perhaps
assisting the product release.
458
The enzyme contains five posttranslationally
modified amino acids near the active site: N-methyl-
histidine, 5-methylarginine, 2-methylglutamine, 2-methyl-
cysteine, and thioglycine in a thiopeptide bond. The
latter may be the site of radical formation.
458a,b
4. Tunichlorins
Nickel is found in various marine invertebrates.
In the tunicates (sea squirts and their relatives) it occurs
in a fixed ratio with cobalt, suggesting a metabolic
role.
460
A new class of nickel chelates called tunichlorins
have been isolated. An example is shown in Fig. 16-27B.
The function of tunichlorins is unknown but their
existence suggests the possibility of unidentified
biochemical roles for nickel.
5. Carbon Monoxide Dehydrogenases and
Carbon Monoxide Dehydrogenase/Acetyl-
CoA Synthase
There are several bacterial carbon monoxide
dehydrogenases that catalyze the reversible oxidation
of CO to CO
2
:
CO + H
2
O CO
2
+ H
+
+ 2 e
-
CO + H
2
O CO
2
+ H
2
G

= 38.1 kJ / mol
Some bacteria use CO as both a source of energy and
for synthesis of carbon compounds. The purple photo-
synthetic bacterium Rhodospirillum rubrum employs a
relatively simple monomeric Ni-containing CO dehy-
drogenase containing one atom of Ni and seven or
eight iron atoms, apparently arranged in Fe
4
S
4
clusters.
These bacteria can grow anaerobically with CO as the
sole source of both energy and carbon.
461
Some aerobic
bacteria oxidize CO using a molybdenum-containing
enzyme (Section H). However, the most studied CO
dehydrogenase is a complex enzyme that also synthe-
sizes, reversibly, acetyl-CoA from CO and a methyl
corrin. Employed by methanogens, acetogens, and
sulfate-reducing bacteria, it is at the heart of the
WoodLjundahl pathway of autotrophic metabolism,
which is discussed further in Chapter 17.
Both oxidation of CO to CO
2
and reduction of CO
2
to CO are important activities of CO dehydrogenase /
acetyl-CoA synthase. During growth on CO, some
CO must be oxidized to CO
2
and then reduced by the
pathways of Fig. 15-22 to form a methyl-tetrahydro-
pterin which can be used to form the methyl group
of acetyl-CoA. During growth on any other carbon
compound CO
2
must be reduced to CO to form the
carbonyl group of acetyl-CoA which can serve as a
precursor to all other carbon compounds. Native CO
dehydrogenase/acetyl CoA synthase was isolated
from cells of Clostridium thermoaceticum grown in the
presence of radioactive
63
Ni. The protein is a 310-kDa

2
oligomer. Each dimer contains 2 atoms of Ni,
1 of Zn, ~ 12 of Fe and ~ 12 sulfide ions,
462 464
which
are organized into three metal clusters referred to as A,
B, and C. Each cluster contains 4 Fe atoms and clusters
A and C also contain 1 Ni each. Oxidation of CO occurs
in the subunits, each of which contains both cluster
B, an Fe
4
S
4
ferredoxin-type cluster, and cluster C, where
the oxidation of CO is thought to occur. Cluster C
contains 1 nickel ion as well as an Fe
4
S
4
cluster that
resembles that of aconitase (Fig. 13-4). Cluster A,
which is in the subunit, also contains 1 atom of Ni
and 4 Fe ions and is probably the site of synthesis of
acetyl-CoA.
Oxidation of CO may require cooperation of the
nickel ion and the Fe
4
S
4
group within the C cluster.
C. Nickel
(16-50a)
(16-50b)
882 Chapter 16. Transition Metals in Catalysis and Electron Transport
Fe(III)
Ni(III)
2e

N
i
2+
C
O
CH
3
CoA S

A cluster
Possible
catalytic
cycle
Fe(I) CH
3
Ni(III) C O
Fe(III)
Ni(III) C O
CoA S C CH
3
O
Acetyl-CoA
C cluster
H
+
+ CO
HO

Eq. 16-51,
reverse
M(III)
3+
C O
M(III)
3+
M(I)
+
Fe(III)
Ni(III)
CH
3
Co(III)
+
Co(I)
Corrinoid
Iron-sulfur
protein
H
+
CH
3
THF
THF
(16-52)
CO probably binds to one of these metals and is attacked
by a hydroxyl ion (Eq. 16-51, step b) which may be
donated by the other metal of the pair.
465,465a
CO
2
and
a proton are released rapidly (step c), after which the
reduced metal center is reoxidized (step d). One elec-
tron is thought to be transferred directly to the Fe
4
S
4
cluster B and the second by an alternative route.
465
A
multienzyme complex isolated from the methanogen
Methanosarcina has an ()
6
structure, with the
subunits having masses of 89, 60, 50, 48, and 20 kDa,
respectively.
466
In this complex the CO dehydrogenase/
acetyl-CoA synthase activity appears to reside in the

2
complex, the complex has a tetrahydropteridine:
cob(I)amideprotein transferase, and the subunit
has an acetyltransferase that binds acetyl-CoA and
transfers the acetyl group to a group on the sub-
unit.
466
Although the Clostridium and Methanosarcina
systems are not identical, similar mechanisms are
presumably involved.
467,467a
To generate acetyl-CoA
a methyl group is first transferred from a tetrahydro-
pterin such as tetrahydrofolate or, in methanogens,
tetrahydromethanopterin or tetrahydrosarcinapterin
(Fig. 15-17) to form a methylcorrinoid. At the A center
of the CO dehydrogenase a molecule of CO, which
may be bound to the Ni, equilibrates with the methyl
group, and with acetyl-CoA. As depicted in Eq. 16-52,
acetyl-Ni may be an intermediate. Other details shown
here are hypothetical. It is possible that the methyl
group is transferred to the Ni atom in the M cluster
before reaction with the CO, which might be bound
to either Ni of cluster C or to Fe. This reaction of two
transition-metal-bound ligands parallels a proposed
industrial process for synthesis of acetic acid from
methanol and CO and involving catalysis by rhodium
metal and methyl iodide. It is thought that rhodium-
bound CO is inserted into bound Rh CH
3
to
form an intermediate . The acetyl group is
released as acetyl iodide which is hydrolyzed to acetic
acid. An acetyl-nickel intermediate may be involved
in the corresponding biological reaction of Eq. 16-52.
The stereochemistry of the sequence has been investi-
gated using methyl-THF containing a chiral methyl
group. Overall retention of the configuration of the
methyl group in acetyl-CoA was observed.
468,469
D. Copper
Copper was recognized as nutritionally essential
by 1924 and has since been found to function in many
cellular proteins.
470 474
Copper is so broadly distributed
in foods that a deficiency has only
rarely been observed in humans.
474a
However, animals may sometimes
receive inadequate amounts because
absorption of Cu
2+
is antagonized by
Zn
2+
and because copper may be tied
up by molybdate as an inert complex.
There are copper-deficient desert areas
of Australia where neither plants nor
animals survive. Copper-deficient
animals have bone defects, hair color
is lacking, and hemoglobin synthesis is
impaired. Cytochrome oxidase activity
is low. The protein elastin of arterial
walls is poorly crosslinked and the
arteries are weak. Genetic defects in
copper metabolism can have similar
effects.
An adult human ingests ~ 2 5 mg
of copper per day, about 30% of which
is absorbed. The total body content of
copper is ~ 100 mg (~ 2 x 10
4
mol / kg),
and both uptake and excretion (via the
bile) are regulated. Since an excess of
copper is toxic, regulation is important.
a b
c
d
CO
M
+
2e

(to B and X centers)


M
3+
C O
OH

M C
O
O

3+
H
+ CO
2
H
+
C center
M
3+
(16-51)
Rh C CH
3
O
883
Because Cu
2+
is the most tightly bound metal ion in
most chelating centers (Table 6-9), almost all of the
copper present in living cells is complexed with pro-
teins. Copper is transported in the blood by a 132-
kDa, 1046-residue sky-blue glycoprotein called
ceruloplasmin.
471,475 477
This one protein contains
3% of the total body copper.
Regulation of copper uptake has been studied in
most detail in the yeast Saccharomyces cerevisiae. Uptake
of Cu
2+
is similar to that of Fe
3+
. The same plasma
membrane reductase system, consisting of proteins
Fre1p and Fre2p (encoded by genes FRE1 and FRE2),
acts to reduce both Fe
3+
and Cu
2+
.
478 481
These two
genes are controlled in part by a transcriptional acti-
vator that responds to the internal copper concentra-
tion.
410,482,483
Similar regulation is thought to occur in
both plants
484
and animals.
The human hereditary disorders Wilsons disease
and Menkes disease have provided further insight
into copper metabolism.
485,486
In Wilsons disease the
ceruloplasmin content is low and copper gradually
accumulates to high levels in the liver and brain. In
Menkes syndrome, there is also a low ceruloplasmin
level and an accumulation of copper in the form of
copper metallothionein.
487,488
Persons with this disease
have abnormalities of hair, arteries, and bones and die
in childhood of cerebral degeneration.
489,490
Similar
symptoms are seen in some patients with EhlersDanlos
syndrome (Box 8-E).
491
Genes for the proteins that are
defective in both Wilsons and Menkes diseases have
been cloned and both proteins have been identified as
P-type ATPase cation transporters (Chapter 8).
492 495c
The two proteins must be similar in structure as indi-
cated by a 55% sequence identity.
496
Homologous
genes involved in copper homeostasis have been
located in both yeast
497
and the cyanobacterium
Synechococcus.
498
The transporter encoded by this
yeast gene, designated Ccc2, apparently functions to
export copper from the cytosol into an extracytosolic
compartment. In a similar way the Wilson and Menkes
disease proteins, which reside in the trans-Golgi network,
are thought to export copper or to provide copper for
incorporation into essential proteins.
493,499
The Wilson
disease protein is also found in a shortened form in
mitochondrial membranes.
492
Other proteins associated
with intracellular copper metabolism seem to be chap-
erones for Cu(I).
500 501a
The ability of copper ions to undergo reversible
changes in oxidation state permits them to function in
a variety of oxidationreduction processes. Like iron,
copper also provides sites for reaction with O
2
, with
superoxide radicals, and with nitrite ions.
1. Electron-Transferring Copper Proteins
A large group of small, intensely blue copper
proteins function as single-electron carriers within
bacteria and plants. Best known is plastocyanin,
which is ubiquitous in green plants and functions in
the electron transport chain between the light-absorbing
photosynthetic centers I and II of chloroplasts (Chapter
23). The bacterial azurins
502
are thought to carry electrons
between cytochrome c
441
and cytochrome oxidase.
Amicyanin accepts electrons from the coenzyme TTQ
of methylamine dehydrogenase of methylotrophic
bacteria and passes them to a cytochrome c (Chapter
15).
168,503,504
A basic blue copper protein phytocyanin
of uncertain function occurs in cucumber seeds.
505
The 10.5-kDa peptide chain of plastocyanin is
folded into an eight-stranded barrel (Fig. 2-16),
which contains a single copper atom. In poplar plasto-
cyanin, the Cu is coordinated by the side chains of
His 37, His 87, Met 92, and Cys 84 in a tetrahedral but
distorted toward a trigonal bipyramidal geometry.
Since copper-free apoplastocyanin has essentially the
same structure, this geometry may be imposed by the
protein onto the Cu
2+
, which usually prefers square-
planar or tetrahedral coordination (Chapter 7).
506
Calculations suggest that there is little or no strain and
that the FranckCondon barrier to electron transfer
is low.
507,508
The three-dimensional structure and
copper environment of azurin are similar to those of
plastocyanin.
509 511
Messerschmidt et al.
512
suggested
that the copper site in these proteins is perfectly adapted
to its function because its geometry is a compromise
between the optimal geometries of the Cu(I) and Cu(II)
states between which it alternates. Stellacyanin,
present in the Japanese lac tree and some other plants,
is a mucoprotein; the 108-residue protein is over 40%
carbohydrate.
513
While its spectrum resembles that
of plastocyanins and azurins, stellacyanin contains no
methionine and this amino acid cannot be a ligand to
copper.
514
Rusticyanin functions in the periplasmic
space of some chemolithotropic sulfur bacteria to transfer
electrons from Fe
2+
to cytochrome c as part of the energy-
providing reaction for these organisms (Eq. 18-23).
515 517
Halocyanin functions in membranes of the archaeo-
bacteriumNatronobacterium,
518
and aurocyanin func-
tions in green photosynthetic bacteria.
519
The blue color of these type 1 copper proteins is
much more intense than are the well known colors of
the hydrated ion Cu(H
2
O)
4
2+
or of the more strongly
absorbing Cu(NH
3
)
4
2+
. The blue color of these simple
complexes arises from a transition of an electron from
one d orbital to another within the copper atom. The
absorption is somewhat more intense in copper peptide
chelates of the type shown in Eq. 6-85. However, the
~ 600 nm absorption bands of the blue proteins are an
order of magnitude more intense, as is illustrated by
the absorption spectrum of azurin (Fig. 23-8). The
intense blue is thought to arise as a result of transfer
of electronic charge from the cysteine thiolate to the
Cu
2+
ion.
520,521
D. Copper
884 Chapter 16. Transition Metals in Catalysis and Electron Transport
A third type of copper center, first recognized in
cytochrome c oxidase (see Fig. 18-10) is called Cu
A
or
purple CuA. Each copper ion is bonded to an imida-
zole and two cysteines serve as bridging ligands. The
two copper ions are about 0.24 nm apart, and the two
Cu
2+
ions together can accept a single electron from an
external donor such as cytochrome c or azurin to give
a half-reduced form.
521a,b
2. Copper, Zinc-Superoxide Dismutase
Although of similar topology to the blue electron-
transferring proteins, Cu, Zn-superoxide dismutase,
has a different function. This dimeric 153-residue
protein has been demonstrated in the cytoplasm of
virtually all eukaryotic cells
522
and in the periplasmic
space of some bacteria
523
where it converts superoxide
ions

O
2

to O
2
and H
2
O
2
. The enzyme, which has a
major protective role against oxidative damage to cells,
presumably functions in a manner similar to that
indicated in Eq. 16-27 for iron or manganese. How-
ever, copper cycles between Cu
2+
and Cu
+
, alternately
accepting and donating electrons.
The active site of cytosolic superoxide dismutase
(SOD) contains both Cu
2+
and Zn
2+
. The copper ion is
of type 2: nonblue and paramagnetic. It is surrounded
by four imidazole groups with an irregular square
planar geometry.
524 527
One of these imidazole groups
(that of His 61) is shared with the Zn
2+
, which is also
bonded to two additional imidazole groups and a side
chain carboxylate. The metal ions have evidently
replaced the hydrogen atom that would otherwise be
present on the imidazole of His 61 (see the following
diagram). It has been suggested, as is also indicated
in the diagram, that when the bound superoxide
donates an electron to the Cu(II) to become O
2
(first
step of Eq. 16-27), a proton becomes attached to the
bridging imidazole with breakage of its linkage to
the Cu(I). The structure of a new crystalline form of
reduced yeast SOD shows that the Cu(I) has moved
0.1 nm away from the bridging imidazole in agreement
with this possibility.
528
In the second half reaction the
imidazole proton, together with a second proton from
the medium and an electron from the Cu(I), would
react to convert the second O
2

into H
2
O
2
. The role
of the Zn
2+
may be in part structural but it may also
serve to ensure that His 61 is protonated on the correct
nitrogen atom. Arg 141 may assist in binding the O
2

as is shown in the diagram. However, the fact that a


mutant containing leucine in place of the active site
arginine has over 10% of the activity of the native
enzyme shows that the arginine is not absolutely
essential.
529
Additional nearby positively charged
arginine and lysine side chains may provide electro-
static guidance that increases the velocity of reaction
of superoxide ions.
530,531
Cu, Zn-SOD is one of the
fastest enzymes known.
In addition to the cytosolic SOD there is a longer
~ 222-residue extracellular form that binds to the pro-
teoglycans found on cell surfaces.
522,527
Manganese
SODs are found in mitochondria and in bacteria and
iron SODs in plants and bacteria. They all appear
to be important in protecting cells from superoxide
radicals.
522,532,533
This importance was dramatically
emphasized when it was found that a defective SOD is
present in persons (about 1 in 100,000) with a hereditary
form of amylotrophic lateral sclerosis (ALS), which
is also called Lou Gehrigs disease after the baseball
hero who was stricken with this terrible disease of
motor neurons in 1939 at the age of 36.
534 536
3. Nitrite and Nitrous Oxide Reductases
Copper enzymes participate in two important
reactions catalyzed by denitrifying bacteria. Nitrite
reductases from species of Achromobacter
537,538
and
Alcaligenes
539 542
are trimeric proteins
543
made up of
37-kDa subunits, each of which contains one type 1
(blue) copper and one type 2 (nonblue) copper. The
first copper serves as an electron acceptor from a
small blue pseudoazurin.
544,544a
The second copper,
which is in the active site, is thought to bind to nitrite
throught its nitrogen atom and to reduce it to NO.
NO
2

+ 2H
+
+ e

NO + H
2
O
Crystallographic studies on the 343-residue Alcaligenes
enzyme reveal two barrel domains with the type 1
copper embedded in one of them and the type 2 cop-
per in an interface between the domains. Studies of
EPR
541
and ENDOR
545
spectra and of various mutant
forms have shown that, as for other copper enzymes,
the type 1 copper is an electron-transferring center,
accepting electrons from the pseudoazurin and passing
them to the type 2 copper which binds and reduces
nitrite.
540,545a
The immediate product of nitrite reductase is NO,
which is reduced in two one-electron steps to N
2
O,
N
N N
Arg 141
H H
H
O
O
N N Cu
2
H
His
His
His
H
His 61
H
Zn
2
His
His
Asp
+
+
+
+

(16-53)
885
and then to N
2
. The second of these steps is catalyzed
by another copper enzyme, nitrous oxide reductase.
546
N
2
O + 2H
+
+ 2e

N
2
+ H
2
O
The biological significance of these reactions is con-
sidered further in Chapters 18 and 24. The 132-kDa
dimeric N
2
O reductase from Pseudomonas stutzeri
contains four copper atoms per subunit.
546
One of its
copper centers resembles the Cu
A
centers of cytochrome
c oxidase. A second copper center consists of four
copper ions, held by seven histidine side chains in a
roughly tetrahedral array around one sulfide (S
2
) ion.
Rasmussen et al. speculate that this copper-sulfide
cluster may be an acceptor of the oxygen atoms of
N
2
O in the formation of N
2
.
546a
There is also a cyto-
chrome cd
1
type of nitrite reductase.
143a
4. Hemocyanins
While many copper proteins are catalysts for
oxidative reactions of O
2
, hemocyanin reacts with O
2
reversibly. This water-soluble O
2
carrier is found in
the blue blood of many molluscs and arthropods,
including snails, crabs, spiders, and scorpions. Hemo-
cyanins are large oligomers ranging in molecular mass
from 450 to 13,000 kDa. Molluscan hemocyanins are
cylindrical oligomers which have a striking appearance
under the electron microscope. Simpler hemocyanins,
found in arthropods, are hexamers of 660-residue 75-kDa
subunits. Each subunit of the hemocyanin from the
spiny lobster is folded into three distinct domains, one
of which contains a pair of Cu(I) atoms which bind
the O
2
. Each copper ion is held by three imidazole
groups without any bridging groups between them,
the CuCu distance being 0.36 0.46 nm.
547
Octopus
hemocyanin has a different fold and forms oligomers
of ten subunits. However, the active sites are very
similar.
548
The O
2
is thought to bind between the two
copper atoms. An allosteric mechanism may involve
changes in the distance between the copper atoms.
549
The oxygenated compound is distinctly blue with a
molar extinction coefficient 5 10 times greater than
that of cupric complexes. This fact suggests that the
Cu(I) has been oxidized to Cu(II) and that the O
2
has
been reduced to the peroxide dianion O
2
2
in the com-
plex.
550 552
Further support for this idea comes from
the observation that treatment of oxygenated hemocy-
anin with glacial acetic acid leads to the formation of
equal amounts of Cu
2+
and Cu
+
and protonated super-
oxide.
5. Copper Oxidases
A large group of copper-containing proteins acti-
vate oxygen toward chemical reactions of dehydroge-
nation, hydroxylation, or oxygenation. Galactose
oxidase (Fig. 16-29), from the mushroom Polyporus, is
a dehydrogenase which converts the 6-hydroxymethyl
group of galactose to an aldehyde while O
2
is reduced
to H
2
O
2
.
Galactose oxidase has been used frequently to label
glycoproteins of external cell membrane surfaces.
Exposed terminal galactosyl or N-acetylgalactosaminyl
residues are oxidized to the corresponding C-6 aldehydes
and the latter are reduced under mild conditions with
tritiated sodium borohydride.
553
The single 639-residue polypeptide chain contains
one type 2 copper ion.
554
Neither oxygen nor galactose
affects the absorption spectrum of the light murky
green enzyme, but the combination of the two does,
suggesting that both substrates bind to the enzyme
before a reaction takes place. A side reaction releases
D. Copper
(16-55)
(CuO
2
Cu)
2+
+ H
+
Cu
2+
+ Cu
+
+ HO
2
R CH
2
OH R CHO
O
2
H
2
O
2 (16-56)
(16-54)
Cu
S
Cu
Cu Cu
His
His
OH
His His
His
His OH
His

Figure 16-29 Drawing of the active site of galactose oxi-


dase showing both the Cu(II) atom and the neighboring free
radical on tyrosine 272, which has been modified by addition
of the thiol of cysteine 228 and oxidation. See Halfen et al.
557
Based on a crystal structure of Ito et al.
558
O

Y495
H661
H496
N H
O
NH

N
H
3
C
O
Cu
2+
N
Y272
C228
S
O
Acetate
886 Chapter 16. Transition Metals in Catalysis and Electron Transport
OH OOC
H
3
N H
OH OOC
H
3
N H
OH
+
O OOC
H
3
N H

3,4-Dihydroxyphenylalanine (dopa)
+
+
Dopaquinone

Tyrosine
(16-57)
superoxide ion and leaves the enzyme in the inactive
Cu(II) state. EPR spectroscopic observations on the
enzyme were puzzling. The active enzyme shows
no EPR signal but a one-electron reduction gives an
inactive form with an EPR signal that arises from
Cu(II). Experimental studies eventually pointed to the
presence of a second reducible center which contains
an organic free radical. In the active form this radical
is antiferromagnetically coupled (spin-coupled)
giving an EPR-silent enzyme able to accept two
electrons.
555557
Another surprise was the discovery, from the X-ray
structure,
558559
that a tyrosine side chain at the active
site has been modified by addition of a thiolate group
from a cysteine residue. This structure, which is also
the site of the organic free radical, is shown in Fig. 16-29.
A possible mechanism of action is portrayed in Fig.
16-30. The substrate binds in step a and its OH group
is deprotonated, perhaps by transfer of H
+
to the phe-
nolate oxygen of tyrosine 495 (Fig. 16-29) in step b of
Fig. 16-30. In step c a free radical hydrogen transfer
occurs to form a ketyl radical which is immediately
oxidized by Cu(II) in step d. In steps c and f the oxi-
dant O
2
is converted to H
2
O and the aldehyde product
is also released. A fungal glyoxal oxidase has similar
characteristics.
560
The galactose oxidase proenzyme is
self-processing. The Tyr-Cys cofactor arises as a result
of copper-catalyzed oxidative modification via a tyrosine
free radical.
560a
Several copper-containing amine oxidases
561,561a
convert amines to aldehydes and H
2
O
2
. They also
contain one of the organic quinone cofactors discussed
in Chapter 15. The dimeric plasma amine oxidase
contains a molecule of the coenzyme TPQ and one
Cu
2+
per 90-kDa subunit.
562563a
Whether the O
2
binds
only to the single atom Cu(I) or also interacts directly
with a cofactor radical in step d of Eq. 15-53 is uncer-
tain.
562
Lysyl oxidase, which is responsible for con-
version of -amino groups of side chains of lysine into
aldehyde groups in collagen and elastin (Chapter 8)
contains coenzyme LTQ as well as Cu.
564
The enzyme
is specifically inhibited by -aminopropionitrile (Box
8-G) and its activity is decreased in genetic diseases of
copper metabolism. The glycerol oxidase of Asper-
gillus is a large 400 kDa protein containing one heme
and two atoms of Cu.
565
It converts glycerol + O
2
into
glyceraldehyde and H
2
O
2
. Also containing copper is
urate oxidase, whose action is indicated in Fig. 25-18.
Tyrosinase catalyzes hydroxylation followed by
dehydrogenation (Eq. 16-57). First identified in mush-
rooms, the enzyme has a widespread distribution in
Figure 16-30 Possible reaction cycle for catalysis of the
6OH of D-galactose or other suitable alcohol substrate by
galactose oxidase. See Wachter et al.
566
and Whittaker et
al.
567
S Y272
O
R
H
H
O
Cu(II)
2
O
2
S Y272
O
R
H
H
O
Cu(II)
2
O
2
S Y272
O
R
H
O
Cu(II)
2
O
2
S Y272
O
R
H
O
Cu(I)
O
2
S
O
R
H
O
H
Cu(II)
2
+
+
O
2
H

S Y272
O
+ +

+
H H

a
e
HO
CH
2
R O
2
H
+
from Y495
H
2
O
2
O
C R
H
f
Cu(II)
2+
b
H
+
to Y495
c
d
887
nature. It is present in large amounts in plant tissues
and is responsible for the darkening of cut fruits. In
animals tyrosinase participates in the synthesis of
dihydroxyphenylalanine (dopa) and in the formation
of the black melanin pigment of skin and hair. Either
a lack of or inhibition of this enzyme in the melanin-
producing melanocytes causes albinism (Chapter 25).
The 46-kDa monomeric tyrosinase of Neurospora
contains a pair of spin-coupled Cu(II) ions.
568,569
The
structure of this copper pair (type 3 copper) has many
properties in common with the copper pair in hemo-
cyanin.
569a
For example, in the absence of other sub-
strates, tyrosinase binds O
2
to form oxytyrosinase,
a compound with properties resembling those of
oxyhemocyanin and containing a bound peroxide
dianion.
569
Tyrosinase is both an oxidase and a hydroxylase.
Some other copper enzymes have only a hydroxylase
function. One of the best understood of these is the
peptidylglycine -hydroxylating monoxygenase,
which catalyzes the first step of the reaction of Eq. 10-
11. The enzyme is a colorless two-copper protein but
the copper atoms are 1.1 nm apart and do not form a
binuclear center.
570
Ascorbate is an essential cosubstrate,
with two molecules being oxidized to the semidehydro-
ascorbate radical as both coppers are reduced to Cu(I).
A ternary complex of reduced enzyme, peptide, and
O
2
is formed and reacts to give the hydroxylated
product.
570
A related two-copper enzyme is dopamine
-monooxygenase, which utilizes O
2
and ascorbate
to hydroxylate dopamine to noradrenaline (Chapter
25).
571,572
These and other types of hydroxylases are
compared in Chapter 18.
The blue multicopper oxidases couple the oxi-
dation of substrates to the four-electron reduction
of molecular oxygen to H
2
O.
573
In this respect they
resemble cytochrome c oxidase, which also contains
copper. However, they do not contain iron. The best
known member of this group is the plant enzyme
ascorbate oxidase, which dehydrogenates ascorbic
acid to dehydroascorbic acid (See Box 18-D). It is a
dimeric blue copper with identical 70-kDa subunits.
The three-dimensional structure revealed one type 1
copper ion held in a typical blue copper environment
as in plastocyanin or azurin and also a three-copper
center. In this center a pair of copper ions, each held
by three imidazole groups, and bridged by a -oxo
group as in hemerythrin (Fig. 16-20), lie 0.51 nm apart
and 0.41 0.44 nm away from the third copper, which
is held by two other imidazoles.
574
The type 1 copper
shows typical intense 600-nm absorption and charac-
teristic EPR signal, while the pair with the oxo bridge
are antiferromagnetically coupled and EPR silent but
with strong near ultraviolet light absorption (type 3
copper). The additional metal ion in the trinuclear
center is a type 2 copper which lacks characteristic
spectroscopic features.
575,576
Reduction of O
2
to 2 H
2
O
is thought to procede via superoxide radical inter-
mediates. When substrate is added to the enzyme,
the blue color fades and it can be shown that the copper
is reduced to the +1 state. The reduced enzyme then
reacts with O
2
, converting it into two molecules of H
2
O.
Similar to ascorbate oxidase in structure and properties
are laccase, found in the latex of the Japanese lac tree
and in the mushroom Polyporus, and the previously
discussed ceruloplasmin.
477
Laccase is a catalyst
for oxidation of phenolic compounds by a free radical
mechanism involving the trinuclear copper center.
577
Studied by Gabriel Bertrand in the 1890s, it was one
of the first oxidative enzymes investigated.
578
In addition to its previously mentioned role in
copper transport, ceruloplasmin is an amine oxidase,
a superoxide dismutase, and a ferrooxidase able to
catalyze the oxidation of Fe
2+
to Fe
3+
. Ceruloplasmin
contains three consecutive homologous 350-residue
sequences which may have originated from an ancestral
copper oxidase gene. Like ascorbate oxidase, this blue
protein contains copper of the three different types.
Blood clotting factors V and VIII (Fig. 12-17), and the
iron uptake protein Fet3 (Section A,1) are also closely
related.
6. Cytochrome c Oxidase
The most studied of all copper-containing oxidases
is cytochrome c oxidase of mitochondria. This multi-
subunit membrane-embedded enzyme accepts four
electrons from cytochrome c and uses them to reduce
O
2
to 2 H
2
O. It is also a proton pump. Its structure and
functions are considered in Chapter 18. However, it is
appropriate to mention here that the essential catalytic
centers consist of two molecules of heme a (a and a
3
)
and three Cu
+
ions. In the fully oxidized enzyme two
metal centers, one Cu
2+
(of the two-copper center Cu
A
)
and one Fe
3+
(heme a), can be detected by EPR spectro-
scopy. The other Cu
2+
(Cu
B
) and heme a
3
exist as an
EPR-silent exchange-coupled pair just as do the two
copper ions of hemocyanin and of other type 3 binuclear
copper centers.
E. Manganese
Tissues usually contain less than one part per
million of manganese on a dry weight basis, less than
0.01 mM in fresh tissues. This compares with a total
content in animal tissues of the more abundant Mg
2+
of 10 mM. A somewhat higher Mn content (3.5 ppm)
is found in bone. Nevertheless, manganese is nutri-
tionally essential
579,580
and its deficiency leads to
well-defined symptoms. These include ovarian and
testicular degeneration, shortening and bowing of
legs, and other skeletal abnormalities such as the
E. Manganese
888 Chapter 16. Transition Metals in Catalysis and Electron Transport
slipped tendon disease of chicks. In Mn deficiency
the organic matrix of bones and cartilage develops
poorly. The galactosamine, hexuronic acids, and
chondroitin sulfates content of cartilage is decreased.
Manganese is also essential for plant growth and
plays a unique and essential role in the photosynthetic
reaction centers of chloroplasts. Two magnetically
coupled pairs of manganese ions bound in a protein
act as the O
2
evolving center in photosynthetic system
II. This function is considered in Chapter 23. An ABC
transporter for managanese uptake has been identified
in the cyanobacterium Synechocystis.
581
Manganese lies in the center of the first transition
series of elements. The stable Mn
2+
(manganous ion)
contains five 3d electrons in a high-spin configuration.
The less stable Mn
3+
(manganic ion) appears to be of
importance in some enzymes and is essential to the
photosynthetic evolution of oxygen. Many enzymes
specifically require or prefer Mn
2+
. These include
galactosyl and N-acetylgalactosaminyltransferases
582
needed for synthesis of mucopolysaccharides (Chapter
20), lactose synthetase (Eq. 20-15), and a muconate-
lactonizing enzyme (Eq. 13-23).
583
Arginase, essential to the production of urea in
the human body (Fig. 24-11), specifically requires
Mn
2+
which exists as a spin-coupled dimetal center
with a bridging water or

OH ion. The Mn
2+
may act
much as does the Ni
2+
ions of urease (Fig. 16-25).
584,585
Pyruvate carboxylase (Eq. 14-3) contains four atoms
of tightly bound Mn
2+
, one for each biotin molecule
present. This manganese is essential for the transcar-
boxylation step in the action of this enzyme. Either
Mn
2+
or Mg
2+
is also needed in the initial step of carboxy-
lation of biotin (Eq. 14-5). Another Mn
2+
-containing
protein is the lectin concanavalin A (Chapter 4). The
joining of O-linked oligosaccharides to secreted glyco-
proteins also seems to require manganese.
586
Manganese is a component of a pseudocatalase
of Lactobacillus,
204
of lignin-degrading peroxidases,
257,258
and of the wine-red superoxide dismutases found in
bacteria and in the mitochondria of eukaryotes.
376,587
The dimeric dismutase from E. coli has a structure
nearly identical to that of bacterial iron SOD (Fig. 16-22).
The manganese ions are presumed to alternate between
the +3 and +2 states during catalysis (Eq. 16-27).
Knockout mice with inactivated Mn SOD genes
live no more than three weeks, indicating that this
enzyme is essential to life. However, mice lacking
CuZn SOD appear normal in most circumstances.
588
Some dioxygenases contain manganese.
589
Many
enzymes that require Mg
2+
can utilize Mn
2+
in its
place, a fact that has been exploited in study of the
active sites of enzymes.
590
The highly paramagnetic
Mn
2+
is the most useful ion for EPR studies (Box 8-C)
and for investigations of paramagnetic relaxation of
NMR signals. Manganese can also replace Zn
2+
in
some enzymes and may alter catalytic properties.
Manganese may function in the regulation of some
enzymes. For example, glutamine synthetase (Fig. 24-7)
in one form requires Mg
2+
for activity but upon adeny-
lylation binds Mn
2+
tightly.
591
Nucleases and DNA
polymerases often show altered specificity when Mn
2+
substitutes for Mg
2+
. However, the significance of
these differences in vivo is uncertain. Manganese is
mutagenic in living organisms, apparently because it
diminishes the fidelity of DNA replication.
592
A striking accumulation of Mn
2+
often occurs
within bacterial spores (Chapter 32). Bacillus subtilus
absolutely requires Mn
2+
for initiation of sporulation.
During logarithmic growth the bacteria can concentrate
Mn
2+
from 1 M in the external medium to 0.2 mM
internally; during sporulation the concentrations
become much higher.
593
F. Chromium
Animals deficient in chromium grow poorly
and have a reduced life span.
594 596
They also have
decreased glucose tolerance, i.e., glucose injected
into the blood stream is removed only half as fast as
it is normally.
597,598
This is similar to the effect of a
deficiency of insulin. Fractionation of yeast led to the
isolation of a chromium-containing glucose tolerance
factor which appeared to be a complex of Cr
3+
, nico-
tinic acid, and amino acids.
597
The chromium in this
material is apparently well absorbed by the body but
is probably not an essential cofactor.
596
Nevertheless,
dietary supplementation with chromium appears to
improve glucose utilization, apparently by enhancing
the action of insulin.
596
Ingestion of glucose not only
increases insulin levels in blood but also causes
increased urinary loss of chromium,
599
perhaps as
a result of insulin-induced mobilization of stored
chromium.
596
It has been suggested that a specific
chromium-binding oligopeptide isolated from
mammalian liver
596,600
may be released in response
to insulin and may activate a membrane phospho-
tyrosine phosphatase.
596
Chromium concentrations in animal tissues are
usually less than 2 M but tend to be much higher in
the caudate nucleus of the brain. High concentrations
of Cr
3+
have also been found in RNAprotein com-
plexes.
601
While several oxidation states, including +2,
+3, and +6, are known for chromium, only Cr(III) is
found to a significant extent in tissues. The Cr(VI)
complex ions, chromate and dichromate, are toxic and
chronic exposure to chromate-containing dust can lead
to lung cancer. Ascorbate is a principal biological
reductant of chromate and can create mutagenic Cr(V)
compounds that include a Cr(V) ascorbateperoxo
complex.
602
However, Cr(III) compounds administered
orally are not significantly toxic. Evidently, the Cr(VI)
compounds can cross cell membranes and be reduced
889
to Cr(III), which forms stable complexes with many
constituents of cells including DNA.
603
The use of
such exchange-inert Cr(III) complexes of ATP in
enzymology was considered in Chapter 12.
Most forms of Cr(III) are not absorbed and uti-
lized by the body. For this reason, and because of the
increased use of sucrose and other refined foods, a
marginal human chromium deficiency may be wide-
spread.
604,605
This may result not only in poor utiliza-
tion of glucose but also in other effects on lipid and
protein metabolism.
597
However, questions have
been raised about the use of chromium picolinate as
a dietary supplement. High concentrations have been
reported to cause chromosome damage
606
and there
may be danger of excessive accumulation of chromium
in the body.
607
G. Vanadium
Vanadium is a dietary essential for goats and
presumably also for human beings,
427
who typically
consume ~ 2 mg / day. However, because vanadium
compounds have powerful pharmacological effects it
has been difficult to establish the nutritional require-
ment for animals.
427,604,607a
The adult body contains
only about 0.1 mg of vanadium. Typical tissue concen-
trations are 0.1 0.7 M
608,609
and serum concentration
may be 10 nM or less. Vanadium can assume oxidation
states ranging from +2 to +5, the vanadate ion VO
4
3
being the predominant form of V(V) in basic solution
and in dilute solutions at pH 7. However, at millimolar
concentrations, V
3
O
9
3
, V
4
O
12
4
, and other polynuclear
forms predominate.
610 612
In plasma most exists as
metavanadate, VO
2

, but within cells it is reducted to


the vanadyl cation VO
2+
which is an especially stable
double-bonded unit in compounds of V(IV).
609
Only
at very low pH is V(III) stable.
The first suggestion of a possible biochemical
function for vanadium came from the discovery that
vanadocytes, the green blood cells of tunicates (sea
squirts), contain ~ 1.0 M V(III) and 1.5 2 M H
2
SO
4
.
613
It was proposed that a V-containing protein is an
oxygen carrier. However, the V
3+
appears not to be
associated with proteins
612
and it does not carry O
2
.
It may be there to poison predators.
614
The vanadium-
accumulating species also synthesize several complex,
yellow catechol-type chelating agents (somewhat
similar to enterobactin; Fig. 16-1) which presumably
complex V(V) and perhaps also reduce it to V(III).
615
Vanadium is also accumulated by other marine organ-
isms and by the mushroom Amanita muscaria.
Vanadoproteins are found in most marine algae
and seaweed and in some lichens.
616
Among these are
haloperoxidases,
252,253,617618b
enzymes that are quite
different from the corresponding heme peroxidases
discussed in Section A,6. The vanadium is bound as
hydrogen vanadate, HVO
4
2
, in trigonal bipyrimidal
coordination with the three oxygens in equatorial
positions and a histidine in one axial position. In the
crystal structure an azide (N
3

) ion occupies the other


axial position, but it is presumably the site of interaction
with peroxide.
619
The structure is similar to that of
acid phosphatases inhibited by vanadate.
620,621
Many
nitrogen-fixing bacteria contain genes for a vanadium-
dependent nitrogenase that is formed only if molyb-
denum is not available.
622
The nitrogenases are
discussed in Chapter 24.
Much of current interest in vanadium stems from
the discovery that vanadate (HVO
4
2
at pH 7) is a
powerful inhibitor of ATPases such as the sodium
pump protein (Na
+
+ K
+
)ATPase (Chapter 8), of phos-
phatases,
623
and of kinases.
624
This can be readily
understood from comparison of the structure of phos-
phate and vanadate ions.
Other enzymes such as the cyclic AMP-dependent
protein kinase are stimulated by vanadium.
624
Vanadate
seems to inhibit most strongly those enzymes that
form a phosphoenzyme intermediate. This inhibition
may be diminished within cells because vanadate is
readily reduced by glutathione and other intracellular
reductants. The resulting vanadyl ion is a much
weaker inhibitor and also stimulates several meta-
bolic processes.
608
Also of great interest is an insulin-like action of
vanadium
607a,625
and evidence that vanadium may be
essential to proper cardiac function.
626
A role in lipid
metabolism was suggested by the observation that in
high doses vanadium inhibits cholesterol synthesis
and lowers the phospholipid and cholesterol content
of blood. Vanadium is reported to inhibit development
of caries by stimulating mineralization of teeth. Unlike
tungsten, vanadium does not compete with molybde-
num in the animal body.
627
The sometimes dramatic
effects of vanadate as an inhibitor, activator, and meta-
bolic regulator are shared also by molybdate and
tungstate.
628,629
Even greater effects are observed
with vanadate, molybdate, or tungstate plus H
2
O
2
.
630
The resulting pervanadate, permolybdate, and
pertungstate are often assumed to be monoperoxo
compounds, e.g., vanodyl hydroperoxide. However,
there is some uncertainty.
631
G. Vanadium
O
P
O
OH
HO
O
V
O
OH
HO
Phosphate

Vanadate
890 Chapter 16. Transition Metals in Catalysis and Electron Transport
xanthine oxidase family (or molybdenum hydroxyl-
ase family).
632,641,643
Also included in the family are
aldehyde oxidoreductases from the sulfate-reducing
Desulfovibrio gigas
633
and from the tomato.
644
Two other families of molybdoenzymes are the
sulfite oxidase family
646a,b
and the dimethylsulfoxide
reductase family.
632,641
Nitrogenase (Chapter 24)
constitutes a fourth family. Sulfite oxidase (Eq. 16-60)
is an essential human liver enzyme (see also Chapter
24).
645,646
SO
3
2
+ H
2
O SO
4
2
+ 2 e

+ 2 H
+
The assimilatory nitrate reductase (Eq. 16-61) of
fungi and green plants (Chapter 24) also belongs to
the sulfite oxidase family.
NO
3
2
+ 2 e

+ 2 H
+
NO
2

+ H
2
O
DMSO reductase reduces dimethylsulfoxide to
dimethylsulfide (Eq. 16-62) as part of the biological
sulfur cycle.
647648d
(H
3
C)
2
S=O + 2 H
+
+ 2 e

(CH
3
)
2
S
A number of other reductases and dehydrogenases,
including dissimilatory nitrate reductases of E. coli
and of denitrifying bacteria (Chapter 18), belong to the
DMSO reductase family. Other members are reductases
for biotin S-oxide,
649
trimethylamine N-oxide, and
polysulfides as well as formate dehydrogenases (Eq.
16-63), formylmethanofuran dehydrogenase (Fig. 15-22,
HCOO

+ 2 e

+ 3 H
+
CO
2
+ 2 H
2
O
step b), and arsenite oxidase.
632
Several other molyb-
doenzymes, such as pyridoxal oxidase, had not been
classified by 1996.
632
1. Molybdenum Ions and Coenzyme Forms
Molybdenum is a metal of the second transition
series, one of the few heavy elements known to be
essential to life. Its most stable oxidation state, Mo(VI),
has 4d orbitals available for coordination with anionic
ligands. Coordination numbers of 4 and 6 are preferred,
but molybdenum can accommodate up to eight ligands.
Most of the complexes are formed from the oxycation
Mo(VI)O
2
2+
. If two molecules of water are coordinated
with this ion, the protons are so acidic that they disso-
ciate completely to give Mo(VI)O
4
2
, the molybdate ion.
Other oxidation states vary from Mo(III) to Mo(V).
In these lower oxidation states, the tendency for protons
to dissociate from coordinated ligands is less, e.g.,
Mo(III)(H
2
O)
6
3+
does not lose protons even in a very
basic medium. Molybdenum tends to form dimeric
(16-60)
(16-61)
(16-62)
H. Molybdenum
Long recognized as an essential element for the
growth of plants, molybdenum has never been directly
demonstrated as a necessary animal nutrient. Never-
theless, it is found in several enzymes of the human
body, as well as in 30 or more additional enzymes of
bacteria and plants.
632
Aldehyde oxidases,
633
xanthine
oxidase of liver and the related xanthine dehydro-
genase, catalyze the reactions of Eqs. 16-58 and 16-59
and contain molybdenum that is essential for catalytic
activity. Xanthine oxidase also contains two Fe
2
S
2
clusters and bound FAD. The enzymes can also
oxidize xanthine further (Eq. 16-59, step b) by a repeti-
tion of the same type of oxidation process at positions
8 and 9 to form uric acid. The much studied xanthine
dehydrogenase has been isolated from milk,
634,635
liver,
fungi,
636
and some bacteria.
637
In the dehydrogenase
NAD
+
is the electron acceptor that oxidizes the bound
FADH
2
formed in Eq. 16-59. Xanthine dehydrogenase,
in the absence of thiol compounds, is converted spon-
taneously into xanthine oxidase, probably as a result of
a conformational change and formation of a disulfide
bridge within the protein. Treatment with thiol com-
pounds such as dithiothreitol reconverts the enzyme
to the dehydrogenase. Evidently in the oxidase form
the NAD
+
binding site has moved away from the FAD,
permitting oxidation of FADH
2
by O
2
with formation
of hydrogen peroxide.
635,638
A purine hydroxylase from fungi,
639
bacterial
quinoline and isoquinoline oxidoreductases,
640,641
and a selenium-containing nicotinic acid hydroxylase
from Clostridium barberei
642
are members of the
(16-63)
R C
O
H
R C
O
OH
H
2
O
2e

2H
+
(16-58)
HN
N
N
N
O
HN
N
N
N
O
O
H
HN
N
N
N
O
Hypoxanthine
H
9
Xanthine
8
H
O
H
H
H
2
O
2H
+
2e

H
2
O
2H
+
+ 2e

(to FAD)
(to FAD)
a
b
(16-59)
891 H. Molybdenum
Figure 16-31 (A) Structure of molybdopterin cytosine dinucleotide complexed with an atom of molybdenum. (B) Stereo-
scopic ribbon drawing of the structure of one subunit of the xanthine oxidase-related aldehyde oxidoreductase from Desulfo-
vibrio gigas. Each 907-residue subunit of the homodimeric protein contains two Fe
2
S
2
clusters visible at the top and the molybdenum
molybdopterin coenzyme buried in the center. (C) Alpha-carbon plot of portions of the protein surrounding the molybdenum
molybdopterin cytosine dinucleotide and (at the top) the two plant-ferredoxin-like Fe
2
S
2
clusters. Each of these is held by a
separate structural domain of the protein. Two additional domains bind the molybdopterin coenzyme and there is also an
intermediate connecting domain. In xanthine oxidase the latter presumably has the FAD binding site which is lacking in the
D. gigas enzyme. From Romo et al.
633
Courtesy of R. Huber.
HN
N N
N
O H
2
N
O
H
H
H
S
Mo
S
O
P
O
P
O
O
O
O
O
N
N
O
O
H
O
HO
HO
H
H

7
Molybdopterin cytosine dinucleotide
or polymeric oxygen-bridged ions. However, within
the enzymes it exists as the unique molybdenum
coenzymes. The Mo-containing enzymes usually also
contain additional bound cofactors, including Fe S
clusters and flavin coenzymes or heme.
The recognition that the Mo in the molybdoproteins
exists in organic cofactor forms came from studies of
mutants of Aspergillus and Neurospora.
650
In 1964,
Pateman and associates discovered mutants that lacked
both nitrate reductase and xanthine dehydrogenase.
Later, it was shown that acid-treated molybdoenzymes
released a material that would restore activity to the
inactived nitrate reductase from the mutant organisms.
This new coenzyme, a phosphate ester of molybdo-
pterin (Fig. 15-17), was characterized by Rajagopalan
and coworkers.
650,651
A more complex form of the
coenzyme, molybdopterin cytosine dinucleotide
(Fig. 16-31), is found in the D. gigas aldehyde oxido-
reductase. Related coenzyme forms include nucleotides
of adenine, guanine (see chapter banner, p. 837), and
hypoxanthine.
651a,651b
The structure of molybdopterin
is related to that of urothione (Fig. 15-17), a normal
urinary constituent. The relationship to urothione was
strengthened by the fact that several children with
severe neurological and other symptoms were found
to lack both sulfite oxidase and xanthine dehydrogenase
as well as the molybdenum cofactor and urinary
urothione.
646,646a,646b
Study by X-ray absorption spectroscopy of the
extended X-ray absorption fine structure (EXAFS)
has provided estimates of both the nature and the
number of the nearest neighboring atoms around the
Mo. The EXAFS spectra of xanthine dehydrogenase
and of nitrate reductase from Chlorella confirmed the
C
A B
892 Chapter 16. Transition Metals in Catalysis and Electron Transport
presence of both the Mo(VI)O
2
unit with Mo O dis-
tances of 0.17 nm and two or three sulfur atoms at
distances of 0.24 nm.
652,653
The two sulfur atoms were
presumed to come from the molybdopterin. A pecu-
liarity of the xanthine oxidase family is the presence
on the molybdenum of a cyanolyzable sulfur.
654
This is a sulfide attached to the molybdenum, which is
present as Mo(VI)OS rather than Mo(VI)O
2
. Reaction
with cyanide produces thiocyanate (Eq. 16-64).
The active site structures of the three classes of
molybdenum-containing enzymes are compared in
Fig. 16-32. In the DMSO reductase family there are
two identical molybdopterin dinucleotide coenzymes
complexed with one molybdenum. However, only
one of these appears to be functionally linked to the
Fe
2
S
2
center.
Nitrogenase, which catalyzes the reduction of N
2
to two molecules of NH
3
, has a different molybdenum
iron cofactor (FeMo-co). It can be obtained by acid
denaturation of the very oxygen-labile ironmolybde-
num protein of nitrogenase followed by extraction
with dimethylformamide.
655,656
The coenzyme is a
complex Fe S Mo cluster also containing homo-
citrate with a composition MoFe
7
S
9
homocitrate
(see Fig. 24-3). Nitrogenase and this coenzyme are
considered further in Chapter 24.
(16-65)
2. Enzymatic Mechanisms
Although several of the reactions catalyzed by
molybdoenzymes are classified as dehydrogenases,
all of them except nitrogenase involve H
2
O as either a
reactant or a product. The EXAFS spectra suggest that
the Mo(VI)O
2
unit is converted to Mo(IV)O during
reaction with a substrate Sub (Eq. 16-65, step a). Reac-
tion of the Mo(IV)O with water (step b) completes the
catalysis.
a
[Mo(VI)O
2
]
2+
+ Sub [Mo(IV)O]
2+
+ SubO
b
[Mo(IV)O]
2+
+ H
2
O 2H
+
+ 2e

+ [Mo(VI)]
2+
O
2
SumSub + H
2
O SubO + 2H
+
+ 2e

Step a of all of these reactions can be regarded as an


oxo-transfer.
653
To complete the reaction, two electrons must be
passed from Mo(IV) to a suitable acceptor, usually an
Fe S cluster or a bound heme group. FAD is also often
present. Xanthine oxidase
634,635,643,657,657a
contains two
Fe
2
S
2
clusters and a FAD for each of the two atoms of
Mo in the dimer. Since this enzyme acts like a typical
flavin oxidase that generates H
2
O
2
from O
2
, it may
be that electrons pass from Mo to the Fe S center and
then to the flavin. Since the EPR signal of the para-
magnetic Mo(V), with its characteristic six-line hyper-
fine structure, is seen during the action of xanthine
oxidase and other molybdenum-containing enzymes,
single-electron transfers are probably involved.
In bacteria such as E. coli a dissimilatory nitrate
reductase allows nitrate to serve as an oxidant in place
of O
2
. An oxygen atom is removed from the nitrate
to form nitrite as two electrons are accepted from a
membrane-bound cytochrome b. The nitrate reductase
consists of a 139-kDa Mo-containing catalytic subunit,
a 58-kDa electron-transferring subunit that contains
both Fe
3
S
4
and Fe
4
S
4
centers, and a 26-kDa heme-
containing membrane anchor subunit.
658 660
The
assimilatory nitrate reductase of fungi, green algae,
and higher plants contains both a b-type cytochrome
and FAD and a molybdenum coenzyme in a large
oligomeric complex.
661 663a
Formate dehydrogenases from many bacteria
contain molybdopterin and also often selenium (Table
15-4).
664,665
A membrane-bound Mo-containing formate
dehydrogenase is produced by E. coli grown anaerobi-
cally in the presence of nitrate. Under these circum-
stances it is coupled to nitrate reductase via an electron-
transport chain in the membranes which permits oxi-
dation of formate by nitrate (Eq. 18-26). This enzyme
is also a multisubunit protein.
665,666
Two other Mo- and
Se- containing formate dehydrogenases are produced
Figure 16-32 Structures surrounding molybdenum in three
families of molybdoenzymes. See Hille.
632
Mo
S
O
S
OH
2
S
S
S
S
S
Mo
O X Ser
VI
Molybdo-
pterin
Xanthine oxidase
(molybdenum hydroxylase)
family
VI
Mo
O
O
S
S
S
OH
S Cys,
Sulfite oxidase family
Molybdopterin
dinucleotide
Se Cys
Molybdopterin
dinucleotide
(X = S, Se)
VI
(
Cys
)
DMSO reductase family
Mo
O S
Mo (VI) (IV)
O
+ 2 + 2
CN

SCN

(16-64)
893 I. Tungsten
by E. coli.
667,668
The three-dimensional structure is
known for one of them, formate dehydrogenase H,
a component of the anaerobic formate hydrogen lyase
complex (Eq. 17-25).
669,670
The structure shows Mo
held by the sulfur atoms of two molybdopterin mole-
cules, as in DMSO reductase. The Se atom of SeCys
140 is also coordinated with the Mo atom, and the
imidazole of His 141 is in close proximity. When
13
C-labeled formate was oxidized in
18
O-enriched
water no
18
O was found in the released product, CO
2
.
671
This suggested that formate may be bound to Mo and
dehydrogenated, with Mo(VI) being reduced to Mo(IV).
The formate hydrogen might be transferred as H
+
to
the His 140 side chain. Mo(IV) could then be reoxidized
by electron transfer in two one-electron steps.
670
How-
ever, recent X-ray absorption spectra suggest the presence
in the enzyme of a selenosulfide ligand to Mo.
672
Mechanistic uncertainties remain!
A flavin-dependent formate dehydrogenase sys-
tem found in Methanobacterium passes electrons from
dehydrogenation of formate to FAD and then to the
deazaflavin coenzyme F
420
.
673
In contrast to these Mo-
containing enzymes, the formate dehydrogenase from
Pseudomonas oxalaticus, which oxidizes formate with
NAD
+
(Eq. 16-66), contains neither Mo or Se.
674
HCOO

+ NAD
+
CO
2
+ NADH
It is a large 315-kDa oligomer containing 2 FMN and
~ 20 Fe / S. Formate dehydrogenases of green plants
and yeasts are smaller 70 - to 80-kDa proteins lacking
bound prosthetic groups.
674
A key enzyme in the
metabolism of carbon monoxide-oxidizing bacteria
is CO oxidase, another membrane-bound molybdo-
enzyme.
675 676c
It also contains selenium, which is
attached to a cysteine side chain as S-selanylcysteine.
A proposed reaction requence
676a
is shown in Eq. 16-67.
3. Nutritional Need for Mo
The first hint of an essential role of molybdenum
in metabolism came from the discovery that animals
raised on a diet deficient in molybdenum had decreased
liver xanthine oxidase activity. There is no evidence
that xanthine oxidase is essential for all life, but a
human genetic deficiency of sulfite oxidase or of its
molybdopterin coenzyme can be lethal.
646,646a,b
The
conversion of molybdate into the molybdopterin
cofactor in E. coli depends upon at least five genes.
677
In Drosophila the addition of the cyanolyzable sulfur
(Eq. 16-64) is the final step in formation of xanthine
dehydrogenase.
678
It is of interest that sulfur (S
0
) can
be transferred from rhodanese (see Eq. 24-45), or from
a related mercaptopyruvate sulfurtransferase
679
into
the desulfo form of xanthine oxidase to generate an
active enzyme.
680
Uptake of molybdate by cells of E. coli is accom-
plished by an ABC-type transport system.
681
In some
bacteria, e.g., the nitrogen-fixing Azotobacter, molybde-
num can be stored in protein-bound forms.
682
I. Tungsten
For many years tungsten was considered only as
a potential antagonist for molybdenum. However, in
1970 growth stimulation by tungsten compounds was
observed for some acetogens, some methanogens, and
a few hyperthermophilic bacteria. Since then over a
dozen tungstoenzymes have been isolated.
683,683a
These can be classified into three categories: aldehyde
oxidoreductases, formaldehyde oxidoreductases, and
the single enzyme acetylene hydratase. In most cases
the tungstoenzymes resemble the corresponding
molybdoenzymes and in most instances organisms
containing a tungsten-requiring enzyme also contain
the corresponding molybdenum enzyme. However,
a few hyperthermophilic archaea appear to require W
and are unable to use Mo.
The aldehyde ferredoxin oxidoreductase from the
hyperthermophile Pyrococcus furiosus was the first
molybdopterin-dependent enzyme for which a three-
dimensional structure became available.
683,684
The
tungstoenzyme resembles that of the related molybdo-
enzyme (Fig. 16-31). A similar ferredoxin-dependent
enzyme reduces glyceraldehyde-3-phosphate.
685
Another member of the tungstoenzyme aldehyde
oxidoreductase family is carboxylic acid reductase,
an enzyme found in certain acetogenic clostridia. It is
able to use reduced ferredoxin to convert unactivated
carboxylic acids into aldehydes, even though E
o
for
the acetaldehyde / acetate couple is 0.58 V.
686
Tungsten- and sometimes Se-containing formate
dehydrogenases together with N-formylmethanofuran
dehydrogenases (Fig. 15-22, step b) form a second family.
(16-66)
S
Mo
S
O
O
O
H
C
Se S
Cys
O
Mo O
O
O
H
C
Se S
Cys
O
Mo
O
O
C
Se S
Cys
O

+VI

O
0
H
+
S-selanylcysteine
anion
(16-67)
894 Chapter 16. Transition Metals in Catalysis and Electron Transport
1. Ochiai, E. I. (1977) Bioinorganic Chemistry. An
Introduction, Allyn and Bacon, Boston
2. Frasto da Silva, J. J. R., and Williams, R. J. P.
(1991) The Biological Chemistry of the Elements:
The Inorganic Chemistry of Life, Clarendon
Press, Oxford
3. Lippard, S. J., and Berg, J. M. (1994) Principles
of Bioinorganic Chemistry, Univ. Science Books,
Mill Valley, California
4. Holm, R. H., Kennepohl, P., and Solomon, E. I.
(1996) Chem. Rev. 96, 2239 2314
5. Neilands, J. B., ed. (1974) Microbial Iron
Metabolism, Academic Press, New York
6. Jacobs, A., and Woodwood, M., eds. (1974)
Iron in Biochemistry and Medicine, Academic
Press, New York
7. Neilands, J. B. (1973) in Inorganic Biochemistry,
Vol. 1 (Eichhorn, G. L., ed), pp. 167202,
Elsevier, Amsterdam
8. Bergeron, R. J. (1986) Trends Biochem. Sci. 11,
133136
9. Neilands, J. B. (1995) J. Biol. Chem. 270, 26723
26726
10. Sakaitani, M., Rusnak, F., Quinn, N. R., Tu, C.,
Frigo, T. B., Berchtold, G. A., and Walsh, C. T.
(1990) Biochemistry 29, 67896798
11. Harris, W. R., Carrano, C. J., Cooper, S. R.,
Sofen, S. R., Avdeef, A. E., McArdle, J. V., and
Raymond, K. N. (1979) J. Am. Chem. Soc. 101,
60976104
11a. Ferguson, A. D., Hofmann, E., Coulton, J. W.,
Diederichs, K., and Welte, W. (1998) Science
282, 22152220
11b. Braun, V., and Killmann, H. (1999) Trends
Biochem. Sci. 24, 104109
11c. Stintzi, A., Barnes, C., Xu, J., and Raymond,
K. N. (2000) Proc. Natl. Acad. Sci. U.S.A. 97,
1069110696
11d. Cohen, S. M., Meyer, M., and Raymond, K. N.
(1998) J. Am. Chem. Soc. 120, 62776286
11e. Adrait, A., Jacquamet, L., Le Pape, L.,
Gonzalez de Peredo, A., Aberdam, D.,
Hazemann, J.-L., Latour, J.-M., and Michaud-
Soret, I. (1999) Biochemistry 38, 62486260
12. Neilands, J. B., Erickson, T. J., and Rastetter,
W. H. (1981) J. Biol. Chem. 256, 38313832
Again, these appear to resemble the corresponding
Mo-dependent enzymes. The unique acetylene
hydratase from the acetylene-utilizing Pelobacter
acetylenicus catalyzes the hydration of acetylene to
acetaldehyde.
687
H C C H + H
2
O H
3
C CHO
In Thermotoga maritima, the most thermophilic organism
known, tungsten promotes synthesis of an Fe-contain-
ing hydrogenase as well as some other enzymes but
seems to have a regulatory rather than a structural
role.
688
(16-68)
References
13. Newton, S. M. C., Allen, J. S., Cao, Z., Qi, Z.,
Jiang, X., Sprencel, C., Igo, J. D., Foster, S. B.,
Payne, M. A., and Klebba, P. E. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 45604565
14. Persmark, M., Expert, D., and Neilands, J. B.
(1989) J. Biol. Chem. 264, 31873193
15. Khalil-Rizvi, S., Toth, S. I., van der Helm, D.,
Vidavsky, I., and Gross, M. L. (1997)
Biochemistry 36, 41634171
16. Wong, G. B., Kappel, M. J., Raymond, K. N.,
Matzanke, B., and Winkelmann, G. (1983) J.
Am. Chem. Soc. 105, 810815
17. Shanzer, A., Libman, J., Lifson, S., and Felder,
C. E. (1986) J. Am. Chem. Soc. 108, 76097619
18. Reid, R. T., Live, D. H., Faulkner, D. J., and
Butler, A. (1993) Nature (London) 366, 455458
19. Llins, M., Wilson, D. M., and Neilands, J. B.
(1973) Biochemistry 12, 38363843
20. Eng-Wilmot, D. L., and van der Helm, D.
(1980) J. Am. Chem. Soc. 102, 77197725
21. Cox, C. D., Rinehart, K. L., Jr., Moore, M. L.,
and Cook, J. C., Jr. (1981) Proc. Natl. Acad. Sci.
U.S.A. 78, 42564260
22. Neilands, J. B. (1952) J. Am. Chem. Soc. 74,
48464847
23. Braun, V. (1985) Trends Biochem. Sci. 10, 7578
24. Letendre, E. D. (1985) Trends Biochem. Sci. 10,
166168
25. Kster, W., and Braun, V. (1990) J. Biol. Chem.
265, 2140721410
26. Jiang, X., Payne, M. A., Cao, Z., Foster, S. B.,
Feix, J. B., Newton, S. M. C., and Klebba, P. E.
(1997) Science 276, 12611264
27. Coy, M., and Neilands, J. B. (1991) Biochemistry
30, 82018210
28. Kammler, M., Schoin, C., and Hantke, D.
(1993) J. Bacteriol. 175, 62126219
29. Dix, D. R., Bridgham, J. T., Broderius, M. A.,
Byersdorfer, C. A., and Eide, D. J. (1994) J.
Biol. Chem. 269, 2609226099
30. Kaplan, J., and OHalloran, T. V. (1996) Science
271, 15101512
31. Dix, D., Bridgham, J., Broderius, M., and Eide,
D. (1997) J. Biol. Chem. 272, 1177011777
32. de Silva, D., Davis-Kaplan, S., Fergestad, J.,
and Kaplan, J. (1997) J. Biol. Chem. 272, 14208
14213
33. Radisky, D., and Kaplan, J. (1999) J. Biol. Chem.
274, 44814484
34. Stearman, R., Yuan, D. S., Yamaguchi-Iwai, Y.,
Klausner, R. D., and Dancis, A. (1996) Science
271, 15521557
35. Hassett, R. F., Romeo, A. M., and Kosman, D.
J. (1998) J. Biol. Chem. 273, 76287636
36. Mukhopadhyay, C., Attieh, Z. K., and Fox, P.
L. (1998) Science 279, 714717
37. Li, L., and Kaplan, J. (1997) J. Biol. Chem. 272,
2848528493
37a. Lange, H., Kispal, G., and Lill, R. (1999) J. Biol.
Chem. 274, 1898918996
37b. Urbanowski, J. L., and Piper, R. C. (1999) J.
Biol. Chem. 274, 3806138070
38. Octave, J.-N., Schneider, Y.-J., Trouet, A., and
Crichton, R. R. (1983) Trends Biochem. Sci. 8,
217222
39. Conrad, M. E., Umbreit, J. N., Moore, E. G.,
Peterson, R. D. A., and Jones, M. B. (1990) J.
Biol. Chem. 265, 52735279
40. Ponka, P., Schulman, H. M., Woodworth, R. C.,
and Richter, G. W., eds. (1990) Iron Transport
and Storage, CRC Press, Boca Raton, Florida
41. Yu, J., and Wessling-Resnick, M. (1998) J. Biol.
Chem. 273, 69096915
42. Gunshin, H., Mackenzie, B., Berger, U. V.,
Gunshin, Y., Romero, M. F., Boron, W. F.,
Nussberger, S., Gollan, J. L., and Hediger, M.
A. (1997) Nature (London) 388, 482488
43. Toth, I., Rogers, J. T., McPhee, J. A., Elliott, S.
M., Abramson, S. L., and Bridges, K. R. (1995)
J. Biol. Chem. 270, 28462852
44. Finch, C. A., and Huebers, H. (1982) N. Engl.
J. Med. 306, 15201528
45. Scrimshaw, N. S. (1991) Sci. Am. 265(Oct), 4652
46. Kaplan, J., and Kushner, J. P. (2000) Nature
(London) 403, 711,713
46a. Khn, L. C. (1999) Trends Biochem. Sci. 24,
164166
46b. Attieh, Z. K., Mukhopadhyay, C. K., Seshadri,
V., Tripoulas, N. A., and Fox, P. L. (1999) J. Biol.
Chem. 274, 11161123
47. Gordeuk, V., Thuma, P., Brittenham, G.,
McLaren, C., Parry, D., Backenstose, A.,
Biemba, G., Msiska, R., Holmes, L., McKinley,
E., Vargas, L., Gilkeson, R., and Poltera, A. A.
(1992) N. Engl. J. Med. 327, 15181521
895
References
70. Bauminger, E. R., Treffry, A., Quail, M. A.,
Zhao, Z., Nowik, I., and Harrison, P. M. (1999)
Biochemistry 38, 77917802
71. Yang, X., Chen-Barrett, Y., Arosio, P., and
Chasteen, N. D. (1998) Biochemistry 37, 9743
9750
72. Pereira, A. S., Small, W., Krebs, C., Tavares, P.,
Edmondson, D. E., Theil, E. C., and Huynh, B.
H. (1998) Biochemistry 37, 98719876
72a. Hwang, J., Krebs, C., Huynh, B. H.,
Edmondson, D. E., Theil, E. C., and Penner-
Hahn, J. E. (2000) Science 287, 122125
73. Treffry, A., Zhao, Z., Quail, M. A., Guest, J. R.,
and Harrison, P. M. (1997) Biochemistry 36,
432441
74. Bourne, P. E., Harrison, P. M., Rice, D. W.,
Smith, J. M. A., and Stansfield, R. F. D. (1982)
in The Biochemistry and Physiology of Iron
(Saltman, P., and Hegenauer, J., eds), pp.
427, Elsevier North Holland, Amsterdam
75. Bridges, K. R., and Hoffman, K. E. (1986) J.
Biol. Chem. 261, 1427314277
76. Medlock, A. E., and Dailey, H. A. (2000)
Biochemistry 39, 74617467
76a. Franco, R., Ma, J.-G., Lu, Y., Ferreira, G. C.,
and Shelnutt, J. A. (2000) Biochemistry 39,
25172529
76b. Lecerof, D., Fodje, M., Hansson, A., Hansson,
M., and Al-Karadaghi, S. (2000) J. Mol. Biol.
297, 221232
77. Eaton, S. S., and Eaton, G. R. (1977) J. Am. Chem.
Soc. 99, 16011604
78. Crossley, M. J., Harding, M. M., and Sternhell,
S. (1986) J. Am. Chem. Soc. 108, 36083613
78a. Braun, J., Schwesinger, R., Williams, P. G.,
Morimoto, H., Wemmer, D. E., and Limbach,
H.-H. (1996) J. Am. Chem. Soc. 118, 1110111110
78b. Treibs, A. (1979) Trends Biochem. Sci. 4, 7172
79. Wu, W., Chang, C. K., Varotsis, C., Babcock,
G. T., Puustinen, A., and Wikstrm, M. (1992)
J. Am. Chem. Soc. 114, 11821187
80. Sone, N., Ogura, T., Noguchi, S., and
Kitagawa, T. (1994) Biochemistry 33, 849855
81. Fenna, R., Zeng, J., and Davey, C. (1995) Arch.
Biochem. Biophys. 316, 653656
82. Taylor, K. L., Strobel, F., Yue, K. T., Ram, P.,
Pohl, J., Woods, A. S., and Kinkade, J. M., Jr.
(1995) Arch. Biochem. Biophys. 316, 635642
83. Callahan, P. M., and Babcock, G. T. (1983)
Biochemistry 22, 452461
84. Petke, J. D., and Maggiora, G. M. (1984) J. Am.
Chem. Soc. 106, 31293133
85. Anraku, Y., and Gennis, R. B. (1987) Trends
Biochem. Sci. 12, 262266
86. Margoliash, E., and Schejter, H. (1984) Trends
Biochem. Sci. 9, 364367
87. Sotiriou, C., and Chang, C. K. (1988) J. Am.
Chem. Soc. 110, 22642270
88. Timkovich, R., Cork, M. S., and Taylor, P. V.
(1984) J. Biol. Chem. 259, 1508915093
89. Kaufman, J., Spicer, L. D., and Siegel, L. M.
(1993) Biochemistry 32, 28532867
90. Matthews, J. C., Timkovich, R., Liu, M.-Y., and
Le Gall, J. (1995) Biochemistry 34, 52485251
91. Chang, C. K., Timkovich, R., and Wu, W.
(1986) Biochemistry 25, 84478453
92. Mylrajan, M., Andersson, L. A., Loehr, T. M.,
Wu, W., and Chang, C. K. (1991) J. Am. Chem.
Soc. 113, 50005005
93. Meyer, T. E., and Kamen, M. D. (1982) Adv.
Prot. Chem. 35, 105212
94. Margoliash, E., and Schejter, A. (1984) Trends
Biochem. Sci. 9, 364367
95. Dickerson, R. E., and Timkovich, R. (1975) in
The Enzymes, 3rd ed., Vol. 11 (Boyer, P. D., ed),
pp. 397547, Academic Press, New York
96. Dickerson, R. E. (1972) Sci. Am. 226(Apr),
5872
97. Takano, T., Kallei, O. B., Swanson, R., and
Dickerson, R. E. (1973) J. Biol. Chem. 248,
52345255
97a. Sebban-Kreuzer, C., Blackledge, M., Dolla, A.,
Marion, D., and Guerlesquin, F. (1998)
Biochemistry 37, 83318340
97b. Dolla, A., Arnoux, P., Protasevich, I.,
Lobachov, V., Brugna, M., Giudici-Orticoni,
M. T., Haser, R., Czjzek, M., Makarov, A., and
Bruschi, M. (1999) Biochemistry 38, 3341
98. Margoliash, E., and Frohwirt, N. (1959)
Biochem. J. 71, 570572
99. Margoliash, E., and Bosshard, H. A. (1983)
Trends Biochem. Sci. 8, 316320
100. Poerio, E., Parr, G. R., and Taniuchi, H. (1986)
J. Biol. Chem. 261, 1097610989
101. Salemme, F. R., Kraut, J., and Kamen, M. D.
(1973) J. Biol. Chem. 248, 77017716
102. Almassy, R. J., and Dickerson, R. E. (1978)
Proc. Natl. Acad. Sci. U.S.A. 75, 26742678
102a. Benini, S., Gonzlez, A., Rypniewski, W. R.,
Wilson, K. S., Van Beeumen, J. J., and Ciurli, S.
(2000) Biochemistry 39, 1311513126
103. Sogabe, S., and Miki, K. (1995) J. Mol. Biol.
252, 235247
104. Simes, P., Matias, P. M., Morais, J., Wilson,
K., Dauter, Z., Carrondo, M. A., (1998)
Inorganic Chimica Acta 273, 213224
105. Sczekan, S. R., and Joshi, J. G. (1987) J. Biol.
Chem. 262, 1378013788
106. Weber, P. C. (1982) Biochemistry 21, 51165119
107. La Mar, G. N., Jackson, J. T., Dugad, L. B.,
Cusanovich, M. A., and Bartsch, R. G. (1990) J.
Biol. Chem. 265, 1617316180
108. Ren, Z., Meyer, T., and McRee, D. E. (1993) J.
Mol. Biol. 234, 433445
109. Weber, P. C., Salemme, F. R., Mathews, F. S.,
and Bethge, P. H. (1981) J. Biol. Chem. 256,
77027704
110. Hamada, K., Bethge, P. H., and Mathews, F. S.
(1995) J. Mol. Biol. 247, 947962
111. Huang, D., Everly, R. M., Cheng, R. H.,
Heymann, J. B., Schgger, H., Sled, V.,
Ohnishi, T., Baker, T. S., and Cramer, W. A.
(1994) Biochemistry 33, 44014409
112. Prince, R. C., and George, G. N. (1995) Trends
Biochem. Sci. 20, 217218
113. Higuchi, Y., Bando, S., Kusunoki, M.,
Matsurura, Y., Yasuska, N., Kakuda, M.,
Yamanaka, T., Yagi, T., and Inokuchi, H.
(1981) J. Biochem. 89, 16591662
114. Bruschi, M., Woudstra, M., Guigliarelli, B.,
Asso, M., Lojou, E., Retillot, Y., and Abergel,
C. (1997) Biochemistry 36, 1060110608
115. Czjzek, M., Payan, F., Guerlesquin, F.,
Bruschi, M., and Haser, R. (1994) J. Mol. Biol.
243, 653667
116. Liu, M.-C., and Peck, H. D., Jr. (1981) J. Biol.
Chem. 256, 1315913164
117. Banci, L., Bertini, I., Bruschi, M.,
Sompornpisut, P., and Turano, P. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1439614400
118. Jenney, F. E., Jr., Prince, R. C., and Daldal, F.
(1994) Biochemistry 33, 24962502
119. Menin, L., Schoepp, B., Garcia, D., Parot, P.,
and Vermglio, A. (1997) Biochemistry 36,
1217512182
120. Menin, L., Schoepp, B., Parot, P., and
Vermglio, A. (1997) Biochemistry 36, 12183
12188
121. Adir, N., Axelrod, H. L., Beroza, P., Isaacson,
R. A., Rongey, S. H., Okamura, M. Y., and
Feher, G. (1996) Biochemistry 35, 25352547
122. Kerfeld, C. A., Anwar, H. P., Interrante, R.,
Merchant, S., and Yeates, T. O. (1995) J. Mol.
Biol. 250, 627647
123. Gao, F., Qin, H., Simpson, M. C., Shelnutt, J.
A., Knaff, D. B., and Ondrias, M. R. (1996)
Biochemistry 35, 1281212819
48. Faller, B., and Nick, H. (1994) J. Am. Chem. Soc.
116, 38603865
49. Baker, E. N., Rumball, S. V., and Anderson, B.
F. (1987) Trends Biochem. Sci. 12, 350353
50. Welch, S. (1992) Transferrin: The Iron Carrier,
CRC Press, Boca Raton, Florida
51. Baker, H. M., Anderson, B. F., Brodie, A. M.,
Shongwe, M. S., Smith, C. A., and Baker, E. N.
(1996) Biochemistry 35, 90079013
51a. Sharma, A. K., Paramasivam, M., Srinivasan,
A., Yadav, M. P., and Singh, T. P. (1998) J. Mol.
Biol. 289, 303317
51b. Peterson, N. A., Anderson, B. F., Jameson, G.
B., Tweedie, J. W., and Baker, E. N. (2000)
Biochemistry 39, 66256633
51c. Bou Abdallah, F., and El Hage Chahine, J.-M.
(2000) J. Mol. Biol. 303, 255 266
52. Kurokawa, H., Mikami, B., and Hirose, M.
(1995) J. Mol. Biol. 254, 196207
52a. Mizutani, K., Yamashita, H., Mikami, B., and
Hirose, M. (2000) Biochemistry 39, 32583265
53. Moore, S. A., Anderson, B. F., Groom, C. R.,
Haridas, M., and Baker, E. N. (1997) J. Mol.
Biol. 274, 222236
54. Bailey, S., Evans, R. W., Garratt, R. C., Gorinsky,
B., Hasnain, S., Horsburgh, C., Jhoti, H.,
Lindley, P. F., Mydin, A., Sarra, R., and Watson,
J. L. (1988) Biochemistry 27, 58045812
54a. He, Q.-Y., Mason, A. B., Tam, B. M.,
MacGillivray, R. T. A., and Woodworth, R. C.
(1999) Biochemistry 38, 97049711
55. Randell, E. W., Parkes, J. G., Olivieri, N. F.,
and Templeton, D. M. (1994) J. Biol. Chem. 269,
1604616053
56. Grossmann, J. G., Mason, A. B., Woodworth,
R. C., Neu, M., Lindley, P. F., and Hasnain, S.
S. (1993) J. Mol. Biol. 231, 554558
57. Zak, O., Tam, B., MacGillivray, R. T. A., and
Aisen, P. (1997) Biochemistry 36, 1103611043
58. Zak, O., Aisen, P., Crawley, J. B., Joannou, C.
L., Patel, K. J., Rafiq, M., and Evans, R. W.
(1995) Biochemistry 34, 1442814434
59. Mecklenburg, S. L., Donohoe, R. J., and Olah,
G. A. (1997) J. Mol. Biol. 270, 739750
60. Fleming, M. D., Romano, M. A., Su, M. A.,
Garrick, L. M., Garrick, M. D., and Andrews,
N. C. (1998) Proc. Natl. Acad. Sci. U.S.A. 95,
11481153
61. Theil, E. C. (1987) Ann. Rev. Biochem. 56, 289
315
62. Crichton, R. R. (1984) Trends Biochem. Sci. 9,
283286
63. Trikha, J., Theil, E. C., and Allewell, N. M.
(1995) J. Mol. Biol. 248, 949967
64. Hempstead, P. D., Yewdall, S. J., Fernie, A. R.,
Lawson, D. M., Artymiuk, P. J., Rice, D. W.,
Ford, G. C., and Harrison, P. M. (1997) J. Mol.
Biol. 268, 424448
65. Crichton, R. R., Soruco, J.-A., Roland, F.,
Michaux, M.-A., Gallois, B., Prcigoux, G.,
Mahy, J.-P., and Mansuy, D. (1997) Biochemistry
36, 1504915054
66. Watt, G. D., Frankel, R. B., Papaefthymiou, G.
C., Spartalian, K., and Stiefel, E. I. (1986)
Biochemistry 25, 43304336
67. Garg, R. P., Vargo, C. J., Cui, X., and Kurtz, D.
M., Jr. (1996) Biochemistry 35, 62976301
67a. Romo, C. V., Regalla, M., Xavier, A. V.,
Teixeira, M., Liu, M.-Y., and Le Gall, J. (2000)
Biochemistry 39, 68416849
68. Dickey, L. F., Sreedharan, S., Theil, E. C.,
Didsbury, J. R., Wang, H.-H., and Kaufman, R.
E. (1987) J. Biol. Chem. 262, 79017907
69. Sayers, D. E., Theil, E. C., and Rennick, F. J.
(1983) J. Biol. Chem. 258, 1407614079
69a. Johnson, J. L., Cannon, M., Watt, R. K., Frankel,
R. B., and Watt, G. D. (1999) Biochemistry 38,
67066713
896 Chapter 16. Transition Metals in Catalysis and Electron Transport
184. McMahon, M. T., deDios, A. C., Godbout, N.,
Salzmann, R., Laws, D. D., Le, H., Havlin, R.
H., and Oldfield, E. (1998) J. Am.
Chem. Soc. 120, 47844797
185. Collman, J. P., Gagne, R. R., Reed, C. A.,
Halbert, T. R., Lang, G., and Robinson, W. T.
(1975) J. Am. Chem. Soc. 97, 14271439
186. Hoard, J. L. (1971) Science 174, 12951302
187. Fermi, G., Perutz, M. F., and Shulman, R. G.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 61676168
188. Klotz, I. M., Klippenstein, G. L., and
Hendrickson, W. A. (1976) Science 192, 335344
189. Dou, T., Admiraal, S. J., Ikeda-Saito, M.,
Krzywda, S., Wilkinson, A. J., Li, T., Olson, J.
S., Prince, R. C., Pickering, I. J., and George,
G. N. (1995) J. Biol. Chem. 270, 1599316001
190. Shiro, Y., Iizuka, T., Marubayashi, K., Ogura,
T., Kitagawa, T., Balasubramanian, S., and
Boxer, S. G. (1994) Biochemistry 33, 1498614992
191. Kaminaka, S., Takizawa, H., Handa, T.,
Kihara, H., and Kitagawa, T. (1992)
Biochemistry 31, 69977002
192. Zhang, J.-H., and Kurtz, D. M., Jr. (1991)
Biochemistry 30, 91219125
193. Lukat, G. S., Kurtz, D. M. J., Shiemke, A. K.,
Loehr, T. M., and Sanders-Loehr, J. (1984)
Biochemistry 23, 64166422
194. Stenkamp, R. E., Sieker, L. C., Jensen, L. H.,
McCallum, J. D., and Sanders-Loehr, J. (1985)
Proc. Natl. Acad. Sci. U.S.A. 82, 713716
194a. Dunford, H. B. (2000) Heme Peroxidases, Wiley-
VCH, New York
194b. Mat, M. J., Zamocky, M., Nykyri, L. M.,
Herzog, C., Alzari, P. M., Betzel, C., Koller, F.,
and Fita, I. (1999) J. Mol. Biol. 268, 135149
195. Shaffer, J. B., Sutton, R. B., and Bewley, G. C.
(1987) J. Biol. Chem. 262, 1290812911
196. Eaton, J. W., and Ma, M. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2371
2383, McGraw-Hill, New York
197. Bravo, J., Fita, I., Ferrer, J. C., Ens, W., Hillar,
A., Switala, J., and Loewen, P. C. (1997)
Protein Sci. 6, 10161023
198. Fita, I., and Rossmann, M. G. (1985) J. Mol.
Biol. 185, 2137
198a. Putnam, C. D., Arvai, A. S., Bourne, Y., and
Tainer, J. A. (2000) J. Mol. Biol. 296, 295309
199. Vainshtein, B. K., Melik-Adamyan, W. R.,
Barynin, V. V., Vagin, A. A., and Grebenko, A.
I. (1981) Nature (London) 293, 411412
200. Fita, I., and Rossmann, M. G. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 16041608
201. Kirkman, H. N., Rolfo, M., Ferraris, A. M.,
and Gaetani, G. F. (1999) J. Biol. Chem. 274,
1390813914
202. Gouet, P., Jouve, H.-M., and Dideberg, O.
(1995) J. Mol. Biol. 249, 933954
203. Jacob, G. S., and Orme-Johnson, W. H. (1979)
Biochemistry 18, 29752980
204. Kono, Y., and Fridovich, I. (1983) J. Biol. Chem.
258, 60156019
205. Khangulov, S., Sivaraja, M., Barynin, V. V.,
and Dismukes, G. C. (1993) Biochemistry 32,
49124924
206. Holzbaur, I. E., English, A. M., and Ismail, A.
A. (1996) J. Am. Chem. Soc. 118, 33543359
207. Ryan, O., Smyth, M. R., and Fgin, C. O.
(1994) Essays in Biochemistry 28, 129146
208. Newmyer, S. L., and Ortiz de Montellano, P.
R. (1995) J. Biol. Chem. 270, 1943019438
208a. Rodrguez-Lpez, J. N., Gilabert, M. A.,
Tudela, J., Thorneley, R. N. F., and Garca-
Cnovas, F. (2000) Biochemistry 39, 13201
13209
References
124. Okkels, J. S., Kjaer, B., Hansson, ., Svendsen,
I., Mller, B. L., and Scheller, H. V. (1992) J.
Biol. Chem. 267, 2113921145
125. Link, T. A., Hatzfeld, O. M., Unalkat, P.,
Shergill, J. K., Cammack, R., and Mason, J. R.
(1996) Biochemistry 35, 75467552
126. Xia, D., Yu, C.-A., Kim, H., Xia, J.-Z.,
Kachurin, A. M., Zhang, L., Yu, L., and
Deisenhofer, J. (1997) Science 277, 6066
127. von Jagow, G., and Sebald, W. (1980) Ann.
Rev. Biochem. 49, 281314
128. Kiel, J. L. (1995) Type-B Cytochromes: Sensors
and Switches, CRC Press, Boca Raton, Florida
129. Mathews, F. S., Levine, M., and Argos, P.
(1972) J. Mol. Biol. 64, 449464
129a. Kostanjevecki, V., Leys, D., Van Driessche, G.,
Meyer, T. E., Cusanovich, M. A., Fischer, U.,
Guisez, Y., and Van Beeumen, J. (1999) J. Biol.
Chem. 274, 3561435620
130. Vergres, G., Ramsden, J., and Waskell, L.
(1995) J. Biol. Chem. 270, 34143422
131. Nagi, M., Cook, L., Prasad, M. R., and Cinti,
D. L. (1983) J. Biol. Chem. 258, 1482314828
132. Nishida, H., Inaka, K., Yamanaka, M., Kaida,
S., Kobayashi, K., and Miki, K. (1995)
Biochemistry 34, 27632767
133. Lindqvist, Y., Brndn, C.-I., Mathews, F. S.,
and Lederer, F. (1991) J. Biol. Chem. 266, 3198
3207
134. Guiard, B., and Lederer, F. (1979) J. Mol. Biol.
135, 639650
135. Garrett, R. M., and Rajagopalan, K. V. (1996)
J. Biol. Chem. 271, 73877391
136. Tegoni, M., Begotti, S., and Cambillau, C.
(1995) Biochemistry 34, 98409850
137. Weber, P. C., Salemme, F. R., Mathews, F. C.,
and Bethge, P. H. (1981) J. Biol. Chem. 257,
77027704
138. Yu, L., Wei, Y.-Y., Usui, S., and Yu, C.-A. (1992)
J. Biol. Chem. 267, 2450824515
139. Knaff, D. B. (1990) Trends Biochem. Sci. 15,
289291
140. Jalukar, V., Kelley, P. M., and Njus, D. (1991)
J. Biol. Chem. 266, 68786882
141. Kita, K., Konishi, K., and Anraku, Y. (1984)
J. Biol. Chem. 259, 33683374
142. Nakamura, K., Yamaki, M., Sarada, M.,
Nakayama, S., Vibat, C. R. T., Gennis, R. B.,
Nakayashiki, T., Inokuchi, H., Kojima, S., and
Kita, K. (1996) J. Biol. Chem. 271, 521527
143. Yang, X., Yu, L., and Yu, C.-A. (1997) J. Biol.
Chem. 272, 96839689
143a. Ranghino, G., Scorza, E., Sjgren, T., Williams,
P. A., Ricci, M., and Hajdu, J. (2000) Biochemistry
39, 1095810966
144. Gray, H. B., and Winkler, J. R. (1996) Ann. Rev.
Biochem. 65, 537561
145. Farver, O., and Pecht, I. (1991) FASEB J. 5,
25542559
146. Komar-Panicucci, S., Sherman, F., and
McLendon, G. (1996) Biochemistry 35, 4878
4885
147. Page, C. C., Moser, C. C., Chen, X., and Dutton,
P. L. (1999) Nature (London) 402, 4752
148. DeVault, D. (1984) Quantum-Mechanical
Tunnelling in Biological Systems, Cambridge
Univ. Press, Cambridge, UK
149. Closs, G. L., and Miller, J. R. (1988) Science
240, 440447
150. Evenson, J. W., and Karplus, M. (1993) Science
262, 12471249
151. Moser, C. C., Keske, J. M., Warncke, K., Farid,
R. S., and Dutton, P. L. (1992) Nature (London)
355, 796802
152. Williams, R. J. P. (1992) Nature (London) 355,
770771
153. Pappa, H. S., and Poulos, T. L. (1995)
Biochemistry 34, 65736580
154. Mayo, S. L., Ellis, W. R., Jr., Crutchley, R. J.,
and Gray, H. B. (1986) Science 233, 948952
155. Wuttke, D. S., Bjerrum, M. J., Winkler, J. R.,
and Gray, H. B. (1992) Science 256, 10071009
156. Banci, L., Bertini, I., De la Rosa, M. A.,
Koulougliotis, D., Navarro, J. A., and Walter,
O. (1998) Biochemistry 37, 48314843
157. Moreira, I., Sun, J., Cho, M. O.-K., Wishart, J.
F., and Isied, S. S. (1994) J. Am. Chem. Soc. 116,
83968397
158. Ahmed, A. J., and Millett, F. (1981) J. Biol.
Chem. 256, 16111615
159. Koppenol, W. H., and Margoliash, E. (1982) J.
Biol. Chem. 257, 44264437
160. Hahm, S., Miller, M. A., Geren, L., Kraut, J.,
Durham, B., and Millett, F. (1994) Biochemistry
33, 14731480
161. Willie, A., Stayton, P. S., Sligar, S. G., Durham,
B., and Millett, F. (1992) Biochemistry 31, 7237
7242
162. Qin, L., and Kostic, N. M. (1994) Biochemistry
33, 1259212599
163. Liang, N., Mauk, A. G., Pielak, G. J., Johnson,
J. A., Smith, M., and Hoffman, B. M. (1988)
Science 240, 311314
164. Pelletier, H., and Kraut, J. (1992) Science 258,
17481755
165. Miller, M. A., Vitello, L., and Erman, J. E.
(1995) Biochemistry 34, 1204812058
166. Mei, H., Wang, K., Peffer, N., Weatherly, G.,
Cohen, D. S., Miller, M., Pielak, G., Durham,
B., and Millett, F. (1999) Biochemistry 38,
68466854
167. Falzon, L., and Davidson, V. L. (1996)
Biochemistry 35, 1211112118
168. Merli, A., Brodersen, D. E., Morini, B., Chen,
Z.-w, Durley, R. C. E., Mathews, F. S.,
Davidson, V. L., and Rossi, G. L. (1996) J. Biol.
Chem. 271, 91779180
169. Mutz, M. W., McLendon, G. L., Wishart, J. F.,
Gaillard, E. R., and Corin, A. F. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 95219526
169a. Castleman, A. W., Jr., Zhong, Q., and Hurley,
S. M. (1999) Proc. Natl. Acad. Sci. U.S.A. 96,
42194220
170. Daizadeh, I., Medvedev, E. S., and
Stuchebrukhov, A. A. (1997) Proc. Natl. Acad.
Sci. U.S.A. 94, 37033708
171. Langen, R., Chang, I.-J., Germanas, J. P.,
Richards, J. H., Winkler, J. R., and Gray, H. B.
(1995) Science 268, 17331735
172. Bishop, G. R., and Davidson, V. L. (1997)
Biochemistry 36, 1358613592
173. Ivkovic-Jensen, M. M., and Kostic, N. M.
(1997) Biochemistry 36, 81358144
174. Ingraham, L. L. (1966) Comprehensive
Biochemistry 14, 424446
175. Pauling, L. (1948) The Nature of the Chemical
Bond, 2nd ed., Cornell Univ. Press, Ithaca,
New York
176. Smulevich, G., Mantini, A. R., Paoli, M.,
Coletta, M., and Geraci, G. (1995) Biochemistry
34, 75077516
177. Van Dyke, B. R., Saltman, P., and Armstrong,
F. A. (1996) J. Am. Chem. Soc. 118, 34903492
178. Sugawara, Y., Matsuoka, A., Kaino, A., and
Shikama, K. (1995) Biophys. J. 69, 583592
179. Geibel, J., Chang, C. K., and Traylor, T. G.
(1975) J. Am. Chem. Soc. 97, 59245926
180. Gerothanassis, I. P., and Momenteau, M.
(1987) J. Am. Chem. Soc. 109, 69446947
181. Phillips, S. E. V., and Schoenborn, B. P. (1981)
Nature (London) 292, 8184
182. Perutz, M. F. (1989) Trends Biochem. Sci. 14,
4244
183. Brzozowski, A., Derewenda, Z., Dodson, E.,
Dodson, G., Grabowski, M., Liddington, R.,
Skarzynski, T., and Vallely, D. (1984) Nature
(London) 307, 7476
897
References
209. Rodriguez-Lopez, J. N., Hernndez-Ruiz, J.,
Garcia-Cnovas, F., Thorneley, R. N. F.,
Acosta, M., and Arnano, M. B. (1997) J. Biol.
Chem. 272, 54695476
210. Abelskov, A. K., Smith, A. T., Rasmussen, C.
B., Dunford, H. B., and Welinder, K. G. (1997)
Biochemistry 36, 94539463
211. Fukuyama, K., Sato, K., Itakura, H.,
Takahashi, S., and Hosoya, T. (1997) J. Biol.
Chem. 272, 57525756
212. Nissum, M., Neri, F., Mandelman, D., Poulos,
T. L., and Smulevich, G. (1998) Biochemistry
37, 80808087
213. Patterson, W. R., and Poulos, T. L. (1995)
Biochemistry 34, 43314341
214. Patterson, W. R., Poulos, T. L., and Goodin, D.
B. (1995) Biochemistry 34, 43424345
215. Finzel, B. C., Poulos, T. L., and Kraut, J. (1984)
J. Biol. Chem. 259, 1302713036
216. Bonagura, C. A., Sundaramoorthy, M., Pappa,
H. S., Patterson, W. R., and Poulos, T. L. (1996)
Biochemistry 35, 61076115
217. Wang, J., Larsen, R. W., Moench, S. J.,
Satterlee, J. D., Rousseau, D. L., and Ondrias,
M. R. (1996) Biochemistry 35, 453463
218. Sinclair, R., Copeland, B., Yamazaki, I., and
Powers, L. (1995) Biochemistry 34, 1317613182
218a. Doyle, W. A., Blodig, W., Veitch, N. C.,
Piontek, K., and Smith, A. T. (1998)
Biochemistry 37, 1509715105
218b. Choinowski, T., Blodig, W., Winterhalter, K.
H., and Piontek, K. (1999) J. Mol. Biol. 286,
809827
219. Yamaguchi, Y., Zhang, D.-E., Sun, Z., Albee, E.
A., Nagata, S., Tenen, D. G., and Ackerman, S.
J. (1994) J. Biol. Chem. 269, 1941019419
220. Andersson, L. A., Bylkas, S. A., and Wilson, A.
E. (1996) J. Biol. Chem. 271, 34063412
221. Rae, T. D., and Goff, H. M. (1996) J. Am. Chem.
Soc. 118, 21032104
222. Kooter, I. M., Moguilevsky, N., Bollen, A., van
der Veen, L. A., Otto, C., Dekker, H. L., and
Wever, R. (1999) J. Biol. Chem. 274, 26794
26802
222a. Fiedler, T. J., Davey, C. A., and Fenna, R. E.
(2000) J. Biol. Chem. 275, 1196411971
223. Nagano, S., Tanaka, M., Ishimori, K.,
Watanabe, Y., and Morishima, I. (1996)
Biochemistry 35, 1425114258
224. Tanaka, M., Ishimori, K., and Morishima, I.
(1998) Biochemistry 37, 26292638
225. Low, D. W., Gray, H. B., and Duus, J. . (1997)
J. Am. Chem. Soc. 119, 15
226. Makino, R., Uno, T., Nishimura, Y., Iuzuka, T.,
Tsuboi, M., and Ishimura, Y. (1986) J. Biol.
Chem. 261, 83768382
227. Hashimoto, S., Tatsuno, Y., and Kitagawa, T.
(1986) Proc. Natl. Acad. Sci. U.S.A. 83, 2417
2421
228. Nakajima, R., and Yamazaki, I. (1987) J. Biol.
Chem. 262, 25762581
229. Morishima, I., Takamuki, Y., and Shiro, Y.
(1984) J. Am. Chem. Soc. 106, 76667672
230. Loew, G., and Dupuis, M. (1996) J. Am. Chem.
Soc. 118, 1058410587
230a. Filizola, M., and Loew, G. H. (2000) J. Am.
Chem. Soc. 122, 1825
230b. Roach, M. P., Ozaki, S.-i, and Watanabe, Y.
(2000) Biochemistry 39, 14461454
231. Olson, L. P., and Bruice, T. C. (1995)
Biochemistry 34, 73357347
232. Nakamura, M., Yamazaki, I., Kotani, T., and
Ohtaki, S. (1985) J. Biol. Chem. 260, 1354613552
233. Kobayashi, S., Nakano, M., Kimura, T., and
Schaap, A. P. (1987) Biochemistry 26, 50195022
234. Ortiz de Montellano, P. R., Choe, Y. S.,
DePillis, G., and Catalano, C. E. (1987) J. Biol.
Chem. 262, 1164111646
235. Dawson, J. H. (1988) Science 240, 433439
236. Libby, R. D., Thomas, J. A., Kaiser, L. W., and
Hager, L. P. (1982) J. Biol. Chem. 257, 50305037
237. Hosten, C. M., Sullivan, A. M., Palaniappan,
V., Fitzgerald, M. M., and Terner, J. (1994) J.
Biol. Chem. 269, 1396613978
238. Sono, M., Eble, K. S., Dawson, J. H., and
Hager, L. P. (1985) J. Biol. Chem. 260, 15530
15535
239. Sono, M., Dawson, J. H., and Hager, L. P.
(1984) J. Biol. Chem. 259, 1320913216
240. Dawson, J. H., and Sono, M. (1987) Chem. Rev.
87, 12551276
241. Geigert, J., Neidleman, S. L., and Dalietos, D.
J. (1983) J. Biol. Chem. 258, 22732277
242. Mueller, T. J., and Morrison, M. (1974) J. Biol.
Chem. 259, 75687573
243. Andersson, E., Hellman, L., Gullberg, U., and
Olsson, I. (1998) J. Biol. Chem. 273, 47474753
244. van Dalen, C. J., Winterbourn, C. C.,
Senthilmohan, R., and Kettle, A. J. (2000) J.
Biol. Chem. 275, 1163811644
245. Nauseef, W. M., Brigham, S., and Cogley, M.
(1994) J. Biol. Chem. 269, 12121216
246. Davey, C. A., and Fenna, R. E. (1996)
Biochemistry 35, 1096710973
247. Thomas, E. L., Bozeman, P. M., Jefferson, M.
M., and King, C. C. (1995) J. Biol. Chem. 270,
29062913
248. Kanofsky, J. R. (1983) J. Biol. Chem. 258, 5991
5993
249. Khan, A. U., Gebauer, P., and Hager, L. P.
(1983) Proc. Natl. Acad. Sci. U.S.A. 80, 51955197
250. McCormick, M. L., Roeder, T. L., Railsback,
M. A., and Britigan, B. E. (1994) J. Biol. Chem.
269, 2791427919
251. Manthey, J. A., and Hager, L. P. (1981) J. Biol.
Chem. 256, 1123211238
252. Colpas, G. J., Hamstra, B. J., Kampf, J. W., and
Pecoraro, V. L. (1996) J. Am. Chem. Soc. 118,
34693478
253. Messerschmidt, A., and Wever, R. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 392396
254. Mande, S. S., Parsonage, D., Claiborne, A., and
Hol, W. G. J. (1995) Biochemistry 34, 69856992
255. Crane, E. J., III, Parsonage, D., Poole, L. B.,
and Claiborne, A. (1995) Biochemistry 34,
1411414124
255a. Claiborne, A., Yeh, J. I., Mallett, T. C., Luba,
J., Crane, E. J., III, Charrier, V., and Parsonage,
D. (1999) Biochemistry 38, 15407 15416
256. Miki, K., Renganathan, V., and Gold, M. H.
(1986) Biochemistry 25, 47904796
257. Sundaramoorthy, M., Kishi, K., Gold, M. H.,
and Poulos, T. L. (1994) J. Biol. Chem. 269,
3275932767
258. Mauk, M. R., Kishi, K., Gold, M. H., and
Mauk, A. G. (1998) Biochemistry 37, 67676771
259. Beinert, H. (1990) FASEB J. 4, 24832491
260. Beinert, H., Holm, R. H., and Mnck, E. (1997)
Science 277, 653659
261. Carter, C. W., Jr., Kraut, J., Freer, S. T., Xuong,
N., Alden, R. A., and Bartsch, R. G. (1974) J.
Biol. Chem. 249, 42124225
262. Breiter, D. R., Meyer, T. E., Rayment, I., and
Holden, H. M. (1991) J. Biol. Chem. 266,
1866018667
263. Agarwal, A., Li, D., and Cowan, J. A. (1996) J.
Am. Chem. Soc. 118, 927928
264. Banci, L., Bertini, I., Ciurli, S., Ferretti, S.,
Luchinat, C., and Piccioli, M. (1993) Biochemistry
32, 93879397
265. Benning, M. M., Meyer, T. E., Rayment, I., and
Holden, H. M. (1994) Biochemistry 33, 2476
2483
266. Soriano, A., Li, D., Bian, S., Agarwal, A., and
Cowan, J. A. (1996) Biochemistry 35, 12479
12486
266a. Mulholland, S. E., Gibney, B. R., Rabanal, F.,
and Dutton, P. L. (1999) Biochemistry 38,
1044210448
267. Due, E. D., Fanchon, E., Vicat, J., Sieker, L.
C., Meyer, J., and Moulis, J.-M. (1994) J. Mol.
Biol. 243, 683695
268. Dauter, Z., Wilson, K. S., Sieker, L. C., Meyer,
J., and Moulis, J.-M. (1997) Biochemistry 36,
1606516073
269. Jensen, L. H. (1974) Ann. Rev. Biochem. 43,
461474
270. Sry, A., Housset, D., Serre, L., Bonicel, J.,
Hatchikian, C., Frey, M., and Roth, M. (1994)
Biochemistry 33, 1540815417
270a. Kyritsis, P., Kmmerle, R., Huber, J. G.,
Gaillard, J., Guigliarelli, B., Popescu, C.,
Mnck, E., and Moulis, J.-M. (1999)
Biochemistry 38, 63356345
271. Brereton, P. S., Verhagen, M. F. J. M., Zhou, Z.
H., and Adams, M. W. W. (1998) Biochemistry
37, 73517362
272. Bruschi, M. H., Guerlesquin, F. A., Bovier-
Lapience, G. E., Bonicel, J. J., and Couchoud,
P. M. (1985) J. Biol. Chem. 260, 82928296
273. Bruice, T. C., Maskiewicz, R., and Job, R.
(1975) Proc. Natl. Acad. Sci. U.S.A. 72, 231234
274. Dauter, Z., Wilson, K. S., Sieker, L. C., Moulis,
J.-M., and Meyer, J. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 88368840
275. Xiao, Z., Lavery, M. J., Ayhan, M., Scrofani, S.
D. B., Wilce, M. C. J., Guss, J. M., Tregloan, P.
A., George, G. N., and Wedd, A. G. (1998) J.
Am. Chem. Soc. 120, 41354150
276. Fukuyama, K., Hase, T., Matsumoto, S.,
Tsukihara, T., Katsube, Y., Tanaka, N.,
Kakudo, M., Wada, K., and Matsubara, H.
(1980) Nature (London) 286, 522524
277. Kok, M., Oldenhuis, R., van der Linden, M. P.
G., Meulenberg, C. H. C., Kingma, J., and
Witholt, B. (1989) J. Biol. Chem. 264, 54425451
278. Archer, M., Huber, R., Tavares, P., Moura, I.,
Moura, J. J. G., Carrondo, M. A., Sieker, L. C.,
LeGall, J., and Romao, M. J. (1995) J. Mol. Biol.
251, 690702
279. Hurley, J. K., Weber-Main, A. M., Stankovich,
M. T., Benning, M. M., Thoden, J. B., Vanhooke,
J. L., Holden, H. M., Chae, Y. K., Xia, B., Cheng,
H., Markley, J. L., Martinez-Jlvez, M., Gmez-
Moreno, C., Schmeits, J. L., and Tollin, G.
(1997) Biochemistry 36, 1110011117
279a. Mller, J. J., Mller, A., Rottmann, M.,
Bernhardt, R., and Heinemann, U. (1999)
J. Mol. Biol. 294, 501513
280. Hurley, J. K., Weber-Main, A. M., Hodges, A.
E., Stankovich, M. T., Benning, M. M., Holden,
H. M., Cheng, H., Xia, B., Markley, J. L.,
Genzor, C., Gomez-Moreno, C., Hafezi, R., and
Tollin, G. (1997) Biochemistry 36, 1510915117
280a. Morales, R., Chron, M.-H., Hudry-Clergeon,
G., Ptillot, Y., Norager, S., Medina, M., and
Frey, M. (1999) Biochemistry 38, 1576415773
281. Ta, D. T., and Vickery, L. E. (1992) J. Biol. Chem.
267, 1112011125
282. Xia, B., Cheng, H., Skjeldal, L., Coghlan, V.
M., Vickery, L. E., and Markley, J. L. (1995)
Biochemistry 34, 180187
282a. Uhlmann, H., and Bernhardt, R. (1995) J. Biol.
Chem. 270, 2995929966
282b. Ziegler, G. A., and Schulz, G. E. (2000)
Biochemistry 39, 1098610995
283. Meyer, J., Fujinaga, J., Gaillard, J., and Lutz,
M. (1994) Biochemistry 33, 1364213650
284. Pochapsky, T. C., and Ye, X. M. (1991)
Biochemistry 30, 38503856
285. Stout, C. D. (1988) J. Biol. Chem. 263, 92569260
286. Duff, J. L. C., Breton, J. L. J., Butt, J. N.,
Armstrong, F. A., and Thomson, A. J. (1996) J.
Am. Chem. Soc. 118, 85938603
898 Chapter 16. Transition Metals in Catalysis and Electron Transport
343. Bender, C. J., Rosenzweig, A. C., Lippard,
S. J., and Peisach, J. (1994) J. Biol. Chem. 269,
1599315998
344. Nesheim, J. C., and Lipscomb, J. D. (1996)
Biochemistry 35, 1024010247
345. Shu, L., Nesheim, J. C., Kauffmann, K.,
Mnck, E., Lipscomb, J. D., and Que, L., Jr.
(1997) Science 275, 515518
346. Pikus, J. D., Studts, J. M., McClay, K., Steffan,
R. J., and Fox, B. G. (1997) Biochemistry 36,
92839289
347. Herold, S., and Lippard, S. J. (1997) J. Am.
Chem. Soc. 119, 145156
348. Jordan, A., and Reichard, P. (1998) Ann. Rev.
Biochem. 67, 7198
349. Stubbe, J. (1990) J. Biol. Chem. 265, 53295332
350. Reichard, P. (1997) Trends Biochem. Sci. 22,
8185
351. Stubbe, J. (1998) Proc. Natl. Acad. Sci. U.S.A.
95, 27232724
351a. Eriksson, M., Jordan, A., and Eklund, H.
(1998) Biochemistry 37, 1335913369
352. Sahlin, M., Petersson, L., Grslund, A.,
Ehrenberg, A., Sjberg, B.-M., and Thelander,
L. (1987) Biochemistry 26, 55415548
353. Uhlin, U., and Eklund, H. (1994) Nature
(London) 370, 533539
354. Uhlin, U., and Eklund, H. (1996) J. Mol. Biol.
262, 358369
354a. Parkin, S. E., Chen, S., Ley, B. A., Mangravite,
L., Edmondson, D. E., Huynh, B. H., and
Bollinger, J. M., Jr. (1998) Biochemistry 37,
11241130
354b. Rova, U., Adrait, A., Ptsch, S., Grslund, A.,
and Thelander, L. (1999) J. Biol. Chem. 274,
2374623751
354c. Andersson, M. E., Hgbom, M., Rinaldo-
Matthis, A., Andersson, K. K., Sjberg, B.-M.,
and Nordlund, P. (1999) J. Am. Chem. Soc. 121,
23462352
355. Fieschi, F., Torrents, E., Toulokhonova, L.,
Jordan, A., Hellman, U., Barbe, J., Gibert, I.,
Karlsson, M., and Sjberg, B.-M. (1998)
J. Biol. Chem. 273, 43294337
356. Ling, J., Sahlin, M., Sjberg, B.-M., Loehr, T.
M., and Sanders-Loehr, J. (1994) J. Biol. Chem.
269, 55955601
357. Silva, K. E., Elgren, T. E., Que, L., Jr., and
Stankovich, M. T. (1995) Biochemistry 34,
1409314103
358. Sturgeon, B. E., Burdi, D., Chen, S., Huynh,
B.-H., Edmondson, D. E., Stubbe, J., and
Hoffman, B. M. (1996) J. Am. Chem. Soc. 118,
75517557
359. Kauppi, B., Nielsen, B. B., Ramaswamy, S.,
Larson, I. K., Thelander, M., Thelander, L., and
Eklund, H. (1996) J. Mol. Biol. 262, 706720
360. Katterle, B., Sahlin, M., Schmidt, P. P., Ptsch,
S., Logan, D. T., Grslund, A., and Sjberg, B.-
M. (1997) J. Biol. Chem. 272, 1041410421
361. Sealy, R. C., Harman, L., West, P. R., and
Mason, R. P. (1985) J. Am. Chem. Soc. 107,
34013406
362. Sjberg, B.-M., Karlsson, M., and Jrnvall, H.
(1987) J. Biol. Chem. 262, 97369743
363. Ekberg, M., Sahlin, M., Eriksson, M., and
Sjberg, B.-M. (1996) J. Biol. Chem. 271, 20655
20659
364. Ong, S. P., McFarlan, S. C., and Hogenkamp,
H. P. C. (1993) Biochemistry 32, 1139711404
365. Gerfen, G. J., Licht, S., Willems, J.-P., Hoffman,
B. M., and Stubbe, J. (1996) J. Am. Chem. Soc.
118, 81928197
366. Reichard, P. (1993) J. Biol. Chem. 268, 83838386
367. Sun, X., Eliasson, R., Pontis, E., Andersson, J.,
Buist, G., Sjberg, B.-M., and Reichard, P.
(1995) J. Biol. Chem. 270, 24432446
References
287. Schipke, C. G., Goodin, D. B., McRee, D. E.,
and Stout, C. D. (1999) Biochemistry 38, 8228
8239
288. George, G. N., and George, S. J. (1988) Trends
Biochem. Sci. 13, 369370
289. Kent, T. A., Emptage, M. H., Merkle, H.,
Kennedy, M. C., Beinert, H., and Mnck, E.
(1985) J. Biol. Chem. 260, 68716881
290. Tong, J., and Feinberg, B. A. (1994) J. Biol.
Chem. 269, 2492024927
291. Lane, R. W., Ibers, J. A., Frankel, R. B., and
Holm, R. H. (1975) Proc. Natl. Acad. Sci. U.S.A.
72, 28682872
291a. Camba, R., and Armstrong, F. A. (2000)
Biochemistry 39, 1058710598
292. Gardner, P. R., and Fridovich, I. (1991) J. Biol.
Chem. 266, 1932819333
293. Kennedy, M. C., Antholine, W. E., and Beinert,
H. (1997) J. Biol. Chem. 272, 2034020347
294. Duin, E. C., Lafferty, M. E., Crouse, B. R.,
Allen, R. M., Sanyal, I., Flint, D. H., and
Johnson, M. K. (1997) Biochemistry 36, 11811
11820
295. Magliozzo, R. S., McIntosh, B. A., and Sweeney,
W. V. (1982) J. Biol. Chem. 257, 35063509
296. Prince, R. C., and Adams, M. W. W. (1987) J.
Biol. Chem. 262, 51255128
297. Orme-Johnson, W. H. (1972) Ann. Rev.
Biochem. 42, 159204
298. Orme-Johnson, W. H., Hansen, R. E., Beinert,
H., Tsibris, J. C. M., Bartholomaus, R. C., and
Gunsalus, I. C. (1968) Proc. Natl. Acad. Sci.
U.S.A. 60, 368372
299. Scrofani, S. D. B., Brereton, P. S., Hamer, A.
M., Lavery, M. J., McDowall, S. G., Vincent, G.
A., Brownlee, R. T. C., Hoogenraad, N. J., Sadek,
M., and Wedd, A. G. (1994) Biochemistry 33,
1448614495
300. Holz, R. C., Small, F. J., and Ensign, S. A.
(1997) J. Biol. Chem. 36, 1469014696
301. Mouesca, J.-M., Chen, J. L., Noodleman, L.,
Bashford, D., and Case, D. A. (1994) J. Am.
Chem. Soc. 116, 1189811914
302. Swartz, P. D., Beck, B. W., and Ichiye, T. (1996)
Biophys. J. 71, 29582969
303. Kemper, M. A., Stout, C. D., Lloyd, S. E. J.,
Prasad, G. S., Fawcett, S., Armstrong, F. A.,
Shen, B., and Burgess, B. K. (1997) J. Biol.
Chem. 272, 1562015627
304. Bominaar, E. L., Hu, Z., Mnck, E., Girerd, J.-
J., and Borshch, S. A. (1995) J. Am. Chem. Soc.
117, 69766989
305. Scott, M. P., and Biggins, J. (1997) Protein Sci.
6, 340346
306. Coldren, C. D., Hellinga, H. W., and
Caradonna, J. P. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 66356640
307. Rieske, J. S., MacLennan, D. H., and Coleman,
R. (1964) Biochem. Biophys. Res. Commun. 15,
338344
308. Beckmann, J. D., Ljungdahl, P. O., Lopez, J. L.,
and Trumpower, B. L. (1987) J. Biol. Chem. 262,
89018909
309. Brasseur, G., Sled, V., Liebl, U., Ohnishi, T.,
and Daldal, F. (1997) Biochemistry 36, 11685
11696
310. Mosser, G., Breyton, C., Olofsson, A., Popot,
J.-L., and Rigaud, J.-L. (1997) J. Biol. Chem.
272, 20263 20268
311. Liebl, U., Sled, V., Brasseur, G., Ohnishi, T.,
and Daldal, F. (1997) Biochemistry 36, 11675
11684
312. Crane, B. R., Siegel, L. M., and Getzoff, E. D.
(1995) Science 270, 5967
313. Wilkerson, J. O., Janick, P. A., and Siegel, L.
M. (1983) Biochemistry 22, 50485054
314. Crane, B. R., Siegel, L. M., and Getzoff, E. D.
(1997) Biochemistry 36, 1212012137
315. Covs, J., Zeghouf, M., Macherel, D.,
Guigliarelli, B., Asso, M., and Fontecave, M.
(1997) Biochemistry 36, 59215928
316. Crane, B. R., Siegel, L. M., and Getzoff, E. D.
(1997) Biochemistry 36, 1210112119
316a. Peters, J. W., Lanzilotta, W. N., Lemon, B. J.,
and Seefeldt, L. C. (1998) Science 282, 1853
1858
316b. Popescu, C. V., and Mnck, E. (1999) J. Am.
Chem. Soc. 121, 78777884
316c. Nicolet, Y., Lemon, B. J., Fontecilla-Camps, J.
C., and Peters, J. W. (2000) Trends Biochem. Sci.
25, 138143
317. Grabowski, R., Hofmeister, A. E. M., and Buckel,
W. (1993) Trends Biochem. Sci. 18, 297300
318. Hofmeister, A. E. M., Grabowski, R., Linder,
D., and Buckel, W. (1993) Eur. J. Biochem. 215,
341349
319. Hofmeister, A. E. M., Berger, S., and Buckel,
W. (1992) Eur. J. Biochem. 205, 743749
320. Flint, D. H., and Emptage, M. H. (1988) J. Biol.
Chem. 263, 35583564
321. Flint, D. H., Tuminello, J. F., and Miller, T. J.
(1996) J. Biol. Chem. 271, 1605316067
322. Hofmeister, A. E. M., and Buckel, W. (1992)
Eur. J. Biochem. 206, 547552
323. Mh, U., Cinkaya, I., Albracht, S. P. J., and
Buckel, W. (1996) Biochemistry 35, 1171011718
324. Scherf, U., Shling, B., Gottschalk, G., Linder,
D., and Buckel, W. (1994) Arch. Microbiol. 161,
239245
325. Klees, A.-G., Linder, D., and Buckel, W. (1992)
Arch. Microbiol. 158, 294301
326. Vollmer, S. J., Switzer, R. L., and Debrunner, P.
G. (1983) J. Biol. Chem. 258, 1428414293
327. Ramsay, R. R., Dreyer, J.-L., Schloss, J. V.,
Jackson, R. H., Coles, C. J., Beinert, H.,
Cleland, W. W., and Singer, T. P. (1981)
Biochemistry 20, 74767482
328. Gaudu, P., and Weiss, B. (1996) Proc. R. Soc.
(London) 93, 1009410098
329. Ding, H., Hidalgo, E., and Demple, B. (1996)
J. Biol. Chem. 271, 3317333175
330. Hentze, M. W. (1996) Trends Biochem. Sci. 21,
282283
331. Feig, A. L., Masschelein, A., Bakac, A., and
Lippard, S. J. (1997) J. Am. Chem. Soc. 119,
334342
332. Pulver, S. C., Froland, W. A., Lipscomb, J. D.,
and Solomon, E. I. (1997) J. Am. Chem. Soc.
119, 387395
333. Lindqvist, Y., Huang, W., Schneider, G., and
Shanklin, J. (1996) EMBO J. 15, 40814092
334. Que, L., Jr. (1991) Science 253, 273274
335. Doi, K., Gupta, R., and Aisen, P. (1987) J. Biol.
Chem. 262, 69826985
336. Hayman, A. R., and Cox, T. M. (1994) J. Biol.
Chem. 269, 12941300
336a. Lindqvist, Y., Johansson, E., Kaija, H., Vihko,
P., and Schneider, G. (1999) J. Mol. Biol. 291,
135147
336b. Uppenberg, J., Lindqvist, F., Svensson, C., Ek-
Rylander, B., and Andersson, G. (1999) J. Mol.
Biol. 290, 201211
337. Wang, D. L., Holz, R. C., David, S. S., Que, L.,
Jr., and Stankovich, M. T. (1991) Biochemistry
30, 81878194
338. Cohen, S. S. (1984) Trends Biochem. Sci. 9, 334
336
339. Merkx, M., and Averill, B. A. (1998)
Biochemistry 37, 84908497
340. Klabunde, T., Strter, N., Frhlich, R., Witzel,
H., and Krebs, B. (1996) J. Mol. Biol. 259, 737
748
341. Strter, N., Klabunde, T., Tucker, P., Witzel, H.,
and Krebs, B. (1995) Science 268, 14891492
342. Baumbach, G. A., Ketcham, C. M.,
Richardson, D. E., Bazer, F. W., and Roberts,
R. M. (1986) J. Biol. Chem. 261, 1286912878
899
References
368. Sun, X., Ollagnier, S., Schmidt, P. P., Atta, M.,
Mulliez, E., Lepape, L., Eliasson, R., Grslund,
A., Fontecave, M., Reichard, P., and Sjberg,
B.-M. (1996) J. Biol. Chem. 271, 68276831
369. Ollagnier, S., Mulliez, E., Schmidt, P. P.,
Eliasson, R., Gaillard, J., Deronzier, C.,
Bergman, T., Grslund, A., Reichard, P., and
Fontecave, M. (1997) J. Biol. Chem. 272, 24216
24223
369a. Logan, D. T., Andersson, J., Sjberg, B.-M.,
and Nordlund, P. (1999) Science 283, 1499
1504
370. Young, P., Andersson, J., Sahlin, M., and
Sjberg, B.-M. (1996) J. Biol. Chem. 271, 20770
20775
371. van der Donk, W. A., Stubbe, J., Gerfen, G. J.,
Bellew, B. F., and Griffin, R. G. (1995) J. Am.
Chem. Soc. 117, 89088916
371a. Lawrence, C. C., Bennati, M., Obias, H. V.,
Bar, G., Griffin, R. G., and Stubbe, J. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 89798984
372. Covs, J., Le Hir de Fallois, L., Le Pape, L.,
Dcout, J.-L., and Fontecave, M. (1996)
Biochemistry 35, 85958602
373. Lenz, R., and Giese, B. (1997) J. Am. Chem. Soc.
119, 27842794
374. Trant, N. L., Meshnick, S. R., Kitchener, K.,
Eaton, J. W., and Cerami, A. (1983) J. Biol.
Chem. 258, 125130
375. Cooper, J. B., McIntyre, K., Badasso, M. O.,
Wood, S. P., Zhang, Y., Garbe, T. R., and
Young, D. (1995) J. Mol. Biol. 246, 531544
376. Lah, M. S., Dixon, M. M., Pattridge, K. A.,
Stallings, W. C., Fee, J. A., and Ludwig, M. L.
(1995) Biochemistry 34, 16461660
377. Stallings, W. C., Pattridge, K. A., Strong, R. K.,
and Ludwig, M. L. (1985) J. Biol. Chem. 260,
1642416432
378. Sorkin, D. L., Duong, D. K., and Miller, A.-F.
(1997) Biochemistry 36, 82028208
378a. Ursby, T., Adinolfi, B. S., Al-Karadaghi, S., De
Vendittis, E., and Bocchini, V. (1999) J. Mol.
Biol. 286, 189205
379. Carlioz, A., Ludwig, M. L., Stallings, W. C.,
Fee, J. A., Steinman, H. M., and Touati, D.
(1988) J. Biol. Chem. 263, 15551562
380. Barker, H. A. (1972) in The Enzymes, 3rd ed.,
Vol. 6 (Boyer, P. D., ed), pp. 509537,
Academic Press, New York
381. Barker, H. A. (1976) in Relections on
Biochemistry (Kornberg, A., ed), pp. 95,
Pergamon Press, New York
382. Lenhert, P. G., and Hodgkin, D. C. (1961)
Nature (London) 192, 937938
383. Lenhert, P. G., and Hodgkin, D. C. (1961)
Nature (London) 192, 937938
384. Zagalak, B., and Friedrich, W., eds. (1979)
Vitamin B12, de Gruyter, Berlin
385. Dolphin, D., ed. (1982) B12, Wiley, New York
(2 vols.)
386. Brown, K. L. (1987) J. Am. Chem. Soc. 109,
22772284
387. Anton, D. L., Hogenkamp, H. P. C., Walker, T.
E., and Matwiyoff, N. A. (1982) Biochemistry
21, 23722378
388. Wagner, F. (1966) Ann. Rev. Biochem. 35, 405
428
389. Schrauzer, G. N., Deutsch, E., and Windgassen,
R. J. (1968) J. Am. Chem. Soc. 90, 24412442
390. Needham, T. E., Matwiyoff, N. A., Walker, T.
E., and Hogenkamp, H. P. C. (1973) J. Am.
Chem. Soc. 95, 50195024
391. Parry, R. J., Ostrander, J. M., and Arzu, I. Y.
(1985) J. Am. Chem. Soc. 107, 21902191
391a. Walker, L. A., II, Shiang, J. J., Anderson, N. A.,
Pullen, S. H., and Sension, R. J. (1998) J. Am.
Chem. Soc. 120, 72867292
392. Hogenkamp, H. P. C. (1982) in B
12
, Vol. I
(Dolphin, D., ed), pp. 295323, Wiley, New York
393. Hay, B. P., and Finke, R. G. (1986) J. Am. Chem.
Soc. 108, 48204829
394. Ashley, G. W., Harris, G., and Stubbe, J. (1986)
J. Biol. Chem. 261, 39583964
395. Sando, G. N., Blakely, R. L., Hogenkamp, H. P.
C., and Hoffman, P. J. (1975) J. Biol. Chem. 250,
87748779
396. Thelander, L., and Reichard, P. (1979) Ann.
Rev. Biochem. 48, 133158
396a. Brown, K. L., and Li, J. (1998) J. Am. Chem.
Soc. 120, 94669474
396b. Licht, S. S., Lawrence, C. C., and Stubbe, J.
(1999) J. Am. Chem. Soc. 121, 74637468
397. Pratt, J. M. (1982) in B
12
, Vol. I (Dolphin, D., ed),
pp. 325392, Wiley, New York
397a. Zerbe-Burkhardt, K., Ratnatilleke, A.,
Philippon, N., Birch, A., Leiser, A., Vrijbloed,
J. W., Hess, D., Hunziker, P., and Robinson, J.
A. (1998) J. Biol. Chem. 273, 65086517
397b. Ratnatilleke, A., Vrijbloed, J. W., and
Robinson, J. A. (1999) J. Biol. Chem. 274,
3167931685
398. Rety, J., Umani-Ronchi, A., Seibl, J., and
Arigoni, D. (1966) Experientia 22, 502503
399. Sato, K., Orr, J. C., Babior, B. M., and Abeles,
R. H. (1976) J. Biol. Chem. 251, 37343737
400. Wollowitz, S., and Halpern, J. (1984) J. Am.
Chem. Soc. 106, 83198321
401. Graves, S. W., Krouwer, J. S., and Babior, B. M.
(1980) J. Biol. Chem. 255, 74447448
401a. Smith, D. M., Golding, B. T., and Radom, L.
(1999) J. Am. Chem. Soc. 121, 5700 5704
401b. Ke, S.-C., and Warncke, K. (1999) J. Am.
Chem. Soc. 121, 9922 9927
401c. Warncke, K., Schmidt, J. C., and Ke, S.-C.
(1999) J. Am. Chem. Soc. 121, 10522 10528
402. Bothe, H., Darley, D. J., Albracht, S. P. J.,
Gerfen, G. J., Golding, B. T., and Buckel, W.
(1998) Biochemistry 37, 41054113
402a. Chih, H.-W., and Marsh, E. N. G. (1999)
Biochemistry 38, 13684 13691
402b. Roymoulik, I., Chen, H.-P., and Marsh, E. N.
G. (1999) J. Biol. Chem. 274, 11619 11622
403. Lowe, J. N., and Ingraham, L. L. (1971) J. Am.
Chem. Soc. 93, 38013802
404. He, M., and Dowd, P. (1998) J. Am. Chem. Soc.
120, 11331137
405. Mancia, F., Keep, N. H., Nakagawa, A.,
Leadlay, P. F., McSweeney, S., Rasmussen, B.,
Bsecke, P., Diat, O., and Evans, P. R. (1996)
Structure 4, 339350
405a. Mancia, F., Smith, G. A., and Evans, P. R.
(1999) Biochemistry 38, 79998005
405b. Thom, N. H., Meier, T. W., Evans, P. R., and
Leadlay, P. F. (1998) Biochemistry 37, 14386
14393
405c. Smith, D. M., Golding, B. T., and Radom, L.
(1999) J. Am. Chem. Soc. 121, 93889399
405d. Maiti, N., Widjaja, L., and Banerjee, R. (1999)
J. Biol. Chem. 274, 3273332737
406. Reitzer, R., Gruber, K., Jogl, G., Wagner, V.
G., Bothe, H., Buckel, W., and Kratky, C.
(1999) Structure 7, 891 902
406a. Champloy, F., Jogl, G., Reitzer, R., Buckel, W.,
Bothe, H., Beatrix, B., Broeker, G., Michalowicz,
A., Meyer-Klaucke, W., and Kratky, C. (1999)
J. Am. Chem. Soc. 121, 11780 11789
407. Stadtman, T. C. (1971) Science 171, 859867
408. Zagalak, B., Frey, P. A., Karabatsos, G. L., and
Abeles, R. H. (1966) J. Biol. Chem. 241, 3028
3035
409. Sprecher, M., Clark, M. J., and Sprinson, D. B.
(1966) J. Biol. Chem. 241, 872877
410. Baker, J. R., and Stadtman, T. C. (1982) in B
12
,
Vol. II (Dolphin, D., ed), pp. 203232, Wiley,
New York
411. Kunz, F., Rety, J., Arigoni, D., Tsai, L., and
Stadtman, T. C. (1978) Helv. Chim. Acta 61,
11391145
411a. Chang, C. H., and Frey, P. A. (2000) J. Biol.
Chem. 275, 106 114
412. Poston, J. M. (1977) Science 195, 301302
413. Stabler, S. P., Lindenbaum, J., and Allen, R. H.
(1988) J. Biol. Chem. 263, 55815588
414. Ballinger, M. D., Frey, P. A., Reed, G. H., and
LoBrutto, R. (1995) Biochemistry 34, 10086
10093
415. Wu, W., Lieder, K. W., Reed, G. H., and Frey,
P. A. (1995) Biochemistry 34, 1053210537
415a. Wu, W., Booker, S., Lieder, K. W., Bandarian,
V., Reed, G. H., and Frey, P. A. (2000)
Biochemistry 39, 95619570
416. Zhou, Z. S., Peariso, K., Penner-Hahn, J. E.,
and Matthews, R. G. (1999) Biochemistry 38,
1591515926
417. Yamanishi, M., Yamada, S., Muguruma, H.,
Murakami, Y., Tobimatsu, T., Ishida, A.,
Yamauchi, J., and Toraya, T. (1998) Biochemistry
37, 47994803
418. Hall, D. A., Jordan-Starck, T. C., Loo, R. O.,
Ludwig, M. L., and Matthews, R. G. (2000)
Biochemistry 39, 1071110719
418a. Jarrett, J. T., Amaratunga, M., Drennan, C. L.,
Scholten, J. D., Sands, R. H., Ludwig, M. L.,
and Matthews, R. G. (1996) Biochemistry 35,
24642475
419. Zydowsky, T. M., Courtney, L. F., Frasca, V.,
Kobayashi, K., Shimizu, H., Yuen, L.,
Matthews, R. G., Benkovic, S. J., and Floss, H.
G. (1986) J. Am. Chem. Soc. 108, 31523153
420. Wood, J. M. (1982) in B
12
, Vol. II (Dolphin, D.,
ed), pp. 151164, Wiley, New York
421. Wood, J. M. (1974) Science 183, 10491052
422. McBride, B. C., and Wolfe, R. S. (1971)
Biochemistry 10, 43124317
423. Parker, D. J., Wood, H. G., Ghambeer, R. K.,
and Ljungdahl, L. G. (1972) Biochemistry 11,
30743080
424. Thauer, R. K., Diekert, G., and Schonheit, P.
(1980) Trends Biochem. Sci. 5, 304306
425. Nielson, F. H. (1974) in Trace Element
Metabolism in Animals-2 (Hoekstra, W. G.,
Suttie, J. W., Ganther, H. E., and Mertz, W.,
eds), pp. 381395, Univ. Park Press,
Baltimore, Maryland
426. Schnegg, A., and Kirchgessner, M. (1976) Int.
J. Vitamins Nutr. Res. 46, 9699
427. Nielsen, F. H. (1991) FASEB J. 5, 26612667
428. Patel, S. U., Sadler, P. J., Tucker, A., and Viles,
J. H. (1993) J. Am. Chem. Soc. 115, 92859286
429. Nomoto, S., McNeely, M. D., and Sunderman,
F. W., Jr. (1971) Biochemistry 10, 16471651
430. Severne, B. C. (1974) Nature (London) 248,
807808
431. Eskew, D. L., Welch, R. M., and Cary, E. E.
(1983) Science 222, 621623
432. Eitinger, T., and Friedrich, B. (1991) J. Biol.
Chem. 266, 32223227
432a. Eitinger, T., Degen, O., Bhnke, U., and
Mller, M. (2000) J. Biol. Chem. 275, 18029
18033
433. Frieden, E., ed. (1984) Biochemistry of the
Essential Ultratrace Elements, Plenum, New York
434. Walsh, C. T., and Orme-Johnson, W. H. (1987)
Biochemistry 26, 49014906
435. Todd, M. J., and Hausinger, R. P. (1987) J. Biol.
Chem. 262, 59635967
436. Jabri, E., Carr, M. B., Hausinger, R. P., and
Karplus, P. A. (1995) Science 268, 9981003
437. Lippard, S. J. (1995) Science 268, 996997
437a. Barrios, A. M., and Lippard, S. J. (1999) J. Am.
Chem. Soc. 121, 1175111757
438. Park, I.-S., and Hausinger, R. P. (1995) Science
267, 11561158
900 Chapter 16. Transition Metals in Catalysis and Electron Transport
485. Danks, D. M. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 2 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 22112235, McGraw-
Hill, New York
486. Lutsenko, S., and Cooper, M. J. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 60046009
487. Nielson, K. B., and Winge, D. R. (1984) J. Biol.
Chem. 259, 49414946
488. Hamer, D. H., Thiele, D. J., and Lemontt, J. E.
(1985) Science 228, 685690
489. Holtzman, N. A. (1976) Fed. Proc. 35, 22762280
490. Riordan, J. R., and Jolicoeur-Paquet, L. (1982)
J. Biol. Chem. 257, 46394645
491. Peltonen, L., Kuivaniemi, H., Palotie, A.,
Horn, N., Kaitila, I., and Kivirikko, K. I. (1983)
Biochemistry 22, 61566163
492. Hung, I. H., Suzuki, M., Yamaguchi, Y., Yuan,
D. S., Klausner, R. D., and Gitlin, J. D. (1997)
J. Biol. Chem. 272, 2146121466
493. Payne, A. S., and Gitlin, J. D. (1998) J. Biol.
Chem. 273, 37653770
494. Zhou, B., and Gitschier, J. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 74817486
495. Lutsenko, S., Petrukhin, K., Cooper, M. J.,
Gilliam, C. T., and Kaplan, J. H. (1997) J. Biol.
Chem. 272, 1893918944
495a. La Fontaine, S., Firth, S. D., Camakaris, J.,
Englezou, A., Theophilos, M. B., Petris, M. J.,
Howie, M., Lockhart, P. J., Greenough, M.,
Brooks, H., Reddel, R. R., and Mercer, J. F. B.
(1998) J. Biol. Chem. 273, 31375 31380
495b. Forbes, J. R., Hsi, G., and Cox, D. W. (1999) J.
Biol. Chem. 274, 12408 12413
495c. Cobine, P. A., George, G. N., Winzor, D. J.,
Harrison, M. D., Mogahaddas, S., and Dameron,
C. T. (2000) Biochemistry 39, 6857 6863
496. Yamaguchi, Y., Heiny, M. E., Suzuki, M., and
Gitlin, J. D. (1996) Proc. Natl. Acad. Sci. U.S.A.
93, 1403014035
497. Yuan, D. S., Stearman, R., Dancis, A., Dunn,
T., Beeler, T., and Klausner, R. D. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 26322636
498. Phung, L. T., Ajlani, G., and Haselkorn, R. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 96519654
499. Klomp, L. W. J., Lin, S.-J., Yuan, D. S.,
Klausner, R. D., Culotta, V. C., and Gitlin, J. D.
(1997) J. Biol. Chem. 272, 92219226
500. Rae, T. D., Schmidt, P. J., Pufahl, R. A.,
Culotta, V. C., and OHalloran, T. V. (1999)
Science 284, 805 808
500a. OHalloran, T. V., and Culotta, V. C. (2000) J.
Biol. Chem. 275, 2505725060
501. Valentine, J. S., and Gralla, E. B. (1997) Science
278, 817818
501a. Harrison, M. D., Jones, C. E., Solioz, M., and
Dameron, C. T. (2000) Trends Biochem. Sci. 25,
29 32
502. Hoitnik, C. W. G., Driscoll, P. C., Hill, H. A.
O., and Canters, G. W. (1994) Biochemistry 33,
35603571
503. Romero, A., Nar, H., Huber, R.,
Messerschmidt, A., Kalverda, A. P., Canters,
G. W., Durley, R., and Mathews, F. S. (1994) J.
Mol. Biol. 236, 11961211
504. Kalverda, A. P., Wymenga, S. S., Lomman, A.,
van de Ven, F. J. M., Hilbers, C. W., and
Canters, G. W. (1994) J. Mol. Biol. 240, 358371
505. Guss, J. M., Merritt, E. A., Phizackerley, R. P.,
and Freeman, H. C. (1996) J. Mol. Biol. 262,
686705
506. Garrett, T. P. J., Clingeleffer, D. J., Guss, J. M.,
Rogers, S. J., and Freeman, H. C. (1984) J. Biol.
Chem. 259, 28222825
507. Ryde, U., Olsson, M. H. M., Pierloot, K., and
Roos, B. O. (1996) J. Mol. Biol. 261, 586596
508. Guckert, J. A., Lowery, M. D., and Solomon, E.
I. (1995) J. Am. Chem. Soc. 117, 28172844
References
439. Lee, M. H., Pankratz, H. S., Wang, S., Scott, R.
A., Finnegan, M. G., Johnson, M. K., Ippolito,
J. A., Christianson, D. W., and Hausinger, R. P.
(1993) Protein Sci. 2, 10421052
439a. Doolittle, R. F. (1997) Nature (London) 388,
515516
440. Winkler, R. G., Blevins, D. G., Polacco, J. C.,
and Randall, D. D. (1988) Trends Biochem. Sci.
13, 97100
441. Mayhew, S. G., and OConnor, M. E. (1982)
Trends Biochem. Sci. 7, 1821
442. Elsden, S. R. (1981) Trends Biochem. Sci. 6, 252253
443. Fu, W., Drozdzewski, P. M., Morgan, T. V.,
Mortenson, L. E., Juszczak, A., Adams, M. W.
W., He, S.-H., Peck, H. D., Jr., DerVartanian,
D. V., LeGall, J., and Johnson, M. K. (1993)
Biochemistry 32, 48134819
444. Bagley, K. A., Van Garderen, C. J., Chen, M.,
Duin, E. C., Albracht, S. P. J., and Woodruff,
W. H. (1994) Biochemistry 33, 92299236
445. He, S. H., Teixeira, M., LeGall, J., Patil, D. S.,
Moura, I., Moura, J. J. G., DerVartanian, D. V.,
Huynh, B. H., and Peck, H. D., Jr. (1989) J.
Biol. Chem. 264, 26782682
446. Sorgenfrei, O., Duin, E. C., Klein, A., and
Albracht, S. P. J. (1996) J. Biol. Chem. 271,
2379923806
447. Arp, D. J., and Burris, R. H. (1981) Biochemistry
20, 22342240
448. Seefeldt, L. C., and Arp, D. J. (1989)
Biochemistry 28, 15881596
449. Telser, J., Benecky, M. J., Adams, M. W. W.,
Mortensen, L. E., and Hoffman, B. M. (1986)
J. Biol. Chem. 261, 1353613541
450. Volbeda, A., Charon, M.-H., Piras, C., Hatch-
ikian, E. C., Frey, M., and Fontecilla-Camps, J.
C. (1995) Nature (London) 373, 580587
451. Cammack, R. (1995) Nature (London) 373,
556557
452. Dole, F., Fournel, A., Magro, V., Hatchikian, E.
C., Bertrand, P., and Guigliarelli, B. (1997)
Biochemistry 36, 78477854
453. Volbeda, A., Garcin, E., Piras, C., de Lacey, A.
L., Fernandez, V. M., Hatchikian, E. C., Frey,
M., and Fontecilla-Camps, J. C. (1996)
J. Am. Chem. Soc. 118, 1298912996
453b. Davidson, G., Choudhury, S. B., Gu, Z., Bose,
K., Roseboom, W., Albracht, S. P. J., and
Maroney, M. J. (2000) Biochemistry 39, 7468
7479
453a. Amara, P., Volbeda, A., Fontecilla-Camps, J.
C., and Field, M. J. (1999) J. Am. Chem. Soc.
121, 4468 4477
454. Tau, S. L., Fox, J. A., Kojima, N., Walsh, C. T.,
and Orme-Johnson, W. H. (1984) J. Am. Chem.
Soc. 106, 30643066
455. Won, H., Olson, K. D., Wolfe, R. S., and
Summers, M. F. (1990) J. Am. Chem. Soc. 112,
21782184
456. Telser, J., Fann, Y.-C., Renner, M. W., Fajer, J.,
Wang, S., Zhang, H., Scott, R. A., and
Hoffman, B. M. (1997) J. Am. Chem. Soc. 119,
733743
456a. Telser, J., Horng, Y.-C., Becker, D. F.,
Hoffman, B. M., and Ragsdale, S. W. (2000)
J. Am. Chem. Soc. 122, 182 183
457. Varadarajan, R., and Richards, F. M. (1992)
Biochemistry 31, 1231512327
458. Ermler, U., Grabarse, W., Shima, S.,
Goubeaud, M., and Thauer, R. K. (1997)
Science 278, 14571462
458a. Selmer, T., Kahnt, J., Goubeaud, M., Shima,
S., Grabarse, W., Ermler, U., and Thauer, R.
K. (2000) J. Biol. Chem. 275, 3755 3760
458b. Grabarse, W., Mahlert, F., Shima, S., Thauer,
R. K., and Ermler, U. (2000) J. Mol. Biol. 303,
329 344
459. Pfaltz, A., Jaun, B., Fssler, A., Eschenmoser,
A., Jaenchen, R., Gilles, H. H., Diekert, G.,
and Thauer, R. K. (1982) Helv. Chim. Acta 65,
828865
460. Sings, H. L., Bible, K. C., and Rinehart, K. L.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 10560
10565
461. Ensign, S. A. (1995) Biochemistry 34, 53725381
462. Xia, J., Sinclair, J. F., Baldwin, T. O., and Lindahl,
P. A. (1996) Biochemistry 35, 19651971
463. Seravalli, J., Kumar, M., Lu, W.-P., and Ragsdale,
S. W. (1997) Biochemistry 36, 1124111251
464. Xia, J., Hu, Z., Popescu, C. V., Lindahl, P. A.,
and Mnck, E. (1997) J. Am. Chem. Soc. 119,
83018312
465. Anderson, M. E., and Lindahl, P. A. (1996)
Biochemistry 35, 83718380
465a. Fraser, D. M., and Lindahl, P. A. (1999)
Biochemistry 38, 15706 15711
466. Grahame, D. A., and DeMoll, E. (1996) J. Biol.
Chem. 271, 83528358
467. Menon, S., and Ragsdale, S. W. (1998)
Biochemistry 37, 56895698
467a. Murakami, E., and Ragsdale, S. W. (2000) J.
Biol. Chem. 275, 4699 4707
468. Lebertz, H., Simon, H., Courtney, L. F.,
Benkovic, S. J., Zydowsky, L. D., Lee, K., and
Floss, H. G. (1987) J. Am. Chem. Soc. 109,
31733174
469. Raybuck, S. A., Bastian, N. R., Zydowsky, L.
D., Kobayashi, K., Floss, H. G., Orme-
Johnson, W. H., and Walsh, C. T. (1987)
J. Am. Chem. Soc. 109, 31713173
470. Howell, J. M., and Gawthorne, J. M., eds. (1987)
Copper in Animals and Man, Vols. I and II, CRC
Press, Boca, Raton, Florida
471. Frieden, E. (1968) Sci. Am. 218(May), 103114
472. Spiro, T. G., ed. (1981) Copper Proteins, Wiley,
New York
473. Sigel, H., ed. (1981) Metal Ions in Biological
Systems, Vol. 13, (Copper Proteins) Dekker,
New York
474. Linder, M. C. (1991) Biochemistry of Copper,
Plenum, New York
474a. Turnland, J. R. (1999) in Modern Nutrition in
Health and Disease, 9th ed. (Shils, M. E.,
Olson, J. A., Shike, M., and Ross, A. C., eds),
pp. 241 252, Williams & Wilkins, Baltimore,
Maryland
475. Calabrese, L., Carbonaro, M., and Musci, G.
(1988) J. Biol. Chem. 263, 64806483
476. Yang, F., Friedrichs, W. E., Cupples, R. L.,
Bonifacio, M. J., Sanford, J. A., Horton, W. A.,
and Bowman, B. H. (1990) J. Biol. Chem. 265,
1078010785
477. Mukhopadhyay, C. K., Mazumder, B.,
Lindley, P. F., and Fox, P. L. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 1154611551
478. Hassett, R., and Kosman, D. J. (1995) J. Biol.
Chem. 270, 128134
479. Georgatsou, E., Mavrogiannis, L. A., and
Fragiadakis, G. S. (1997) J. Biol. Chem. 272,
1378613792
480. Glerum, D. M., Shtanko, A., and Tzagoloff, A.
(1996) J. Biol. Chem. 271, 2053120535
481. Askwith, C., and Kaplan, J. (1998) Trends
Biochem. Sci. 23, 135138
482. Yamaguchi-Iwai, Y., Serpe, M., Haile, D., Yang,
W., Kosman, D. J., Klausner, R. D., and Dancis,
A. (1997) J. Biol. Chem. 272, 1771117718
483. Zhu, Z., Labb, S., Pena, M. M. O., and Thiele,
D. J. (1998) J. Biol. Chem. 273, 12771280
484. Kampfenkel, K., Kushnir, S., Babiychuk, E.,
Inz, D., and Van Montagu, M. (1995) J. Biol.
Chem. 270, 2847928486
901
References
509. Groeneveld, C. M., and Canters, G. W. (1988)
J. Biol. Chem. 263, 167173
510. Karlsson, B. G., Tsai, L.-C., Nar, H., Sanders-
Loehr, J., Bonander, N., Langer, V., and Sjlin,
L. (1997) Biochemistry 36, 40894095
511. van de Kamp, M., Canters, G. W., Wijmenga,
S. S., Lommen, A., Hilbers, C. W., Nar, H.,
Messerschmidt, A., and Huber, R. (1992)
Biochemistry 31, 1019410207
512. Messerschmidt, A., Prade, L., Kroes, S. J.,
Sanders-Loehr, J., Huber, R., and Canters, G.
W. (1998) Proc. Natl. Acad. Sci. U.S.A. 95,
34433448
513. Farver, O., Licht, A., and Pecht, I. (1987)
Biochemistry 26, 73177321
514. Vila, A. J., and Fernndez, C. O. (1996) J. Am.
Chem. Soc. 118, 72917298
515. Casimiro, D. R., Toy-Palmer, A., Blake, R. C.,
II, and Dyson, H. J. (1995) Biochemistry 34,
66406648
516. Grossmann, J. G., Engledew, W. J., Harvey, I.,
Strange, R. W., and Hasnain, S. S. (1995)
Biochemistry 34, 84068414
517. Botuyan, M. V., Toy-Palmer, A., Chung, J.,
Blake, R. C., II, Beroza, P., Case, D. A., and
Dyson, H. J. (1996) J. Mol. Biol. 263, 752767
518. Hildebrandt, P., Matysik, J., Schrader, B.,
Scharf, B., and Engelhard, M. (1994)
Biochemistry 33, 1142611431
519. McManus, J. D., Brune, D. C., Han, J.,
Sanders-Loehr, J., Meyer, T. E., Cusanovich,
M. A., Tollin, G., and Blankenship, R. E. (1992)
J. Biol. Chem. 267, 65316540
520. Penfield, K. W., Gerwith, A. A., and Solomon,
E. I. (1985) J. Am. Chem. Soc. 107, 45194529
521. Blair, D. F., Campbell, G. W., Schoonover, J. R.,
Chan, S. I., Gray, H. B., Malmstrom, B. G., Pecht,
I., Swanson, B. I., Woodruff, W. H., Cho, W. K.,
English, A. M., Fry, H. A., Lum, V., and Norton,
K. A. (1985) J. Am. Chem. Soc. 107, 57555766
521a. Robinson, H., Ang, M. C., Gao, Y.-G., Hay,
M. T., Lu, Y., and Wang, A. H.-J. (1999)
Biochemistry 38, 5677 5683
521b. Holz, R. C., Bennett, B., Chen, G., and Ming,
L.-J. (1998) J. Am. Chem. Soc. 120, 6329 6335
522. Fridovich, I. (1995) Ann. Rev. Biochem. 64, 97
112
523. Beck, B. L., Tabatabai, L. B., and Mayfield, J.
E. (1990) Biochemistry 29, 372376
524. Tainer, J. A., Getzoff, E. D., Richardson, J. S.,
and Richardson, D. C. (1983) Nature (London)
306, 284287
525. Pesce, A., Capasso, C., Battistoni, A., Folcarelli,
S., Rotilio, G., Desideri, A., and Bolognesi, M.
(1997) J. Mol. Biol. 274, 408420
526. Rypniewski, W. R., Mangani, S., Bruni, B.,
Orioli, P. L., Casati, M., and Wilson, K. S.
(1995) J. Mol. Biol. 251, 282296
527. Stenlund, P., Andersson, D., and Tibell, L. A.
E. (1997) Protein Sci. 6, 23502358
528. Ogihara, N. L., Parge, H. E., Hart, P. J., Weiss,
M. S., Goto, J. J., Crane, B. R., Tsang, J., Slater,
K., Roe, J. A., Valentine, J. S., Eisenberg, D.,
and Tainer, J. A. (1996) Biochemistry 35, 2316
2321
529. Beyer, W. F., Jr., Fridovich, I., Mullenbach, G.
T., and Hallewell, R. (1987) J. Biol. Chem. 262,
1118211187
530. Banci, L., Bertini, I., Bauer, D., Hallewell, R.
A., and Viezzoli, M. S. (1993) Biochemistry 32,
43844388
531. Getzoff, E. D., Cabelli, D. E., Fisher, C. L.,
Parge, H. E., Viezzoli, M. S., Banci, L., and
Hallewell, R. A. (1992) Nature (London) 358,
347351
532. Afanasev, I. B. (1989,1991) Superoxide Ion:
Chemistry and Biological Implications, Vol. 2,
CRC Press, Boca Raton, Florida
533. Hiraishi, H., Terano, A., Razandi, M.,
Sugimoto, T., Harada, T., and Ivey, K. J. (1992)
J. Biol. Chem. 267, 1481214817
534. Deng, H.-X., Hentati, A., Tainer, J. A., Iqbal,
Z., Cayabyab, A., Hung, W.-Y., Getzoff, E. D.,
Hu, P., Herzfeldt, B., Roos, R. P., Warner, C.,
Deng, G., Soriano, E., Smyth, C., Parge, H. E.,
Ahmed, A., Roses, A. D., Hallewell, R. A.,
Pericak-Vance, M. A., and Siddique, T. (1993)
Science 261, 10471051
535. McNamara, J. O., and Fridovich, I. (1993)
Nature (London) 362, 2021
536. Wiedau-Pazos, M., Goto, J. J., Rabizadeh, S.,
Gralla, E. B., Roe, J. A., Lee, M. K., Valentine,
J. S., and Bredesen, D. E. (1996) Science 271,
515518
537. Adman, E. T., Godden, J. W., and Turley, S.
(1995) J. Biol. Chem. 270, 2745827474
538. Suzuki, S., Kohzuma, T., Deligeer, Yamaguchi,
K., Nakamura, N., Shidara, S., Kobayashi, K.,
and Tagawa, S. (1994) J. Am. Chem. Soc. 116,
1114511146
539. Murphy, M. E. P., Turley, S., and Adman, E. T.
(1997) J. Biol. Chem. 272, 2845528460
540. Kukimoto, M., Nishiyama, M., Murphy, M. E.
P., Turley, S., Adman, E. T., Horinouchi, S., and
Beppu, T. (1994) Biochemistry 33, 52465252
540a. Strange, R. W., Murphy, L. M., Dodd, F. E.,
Abraham, Z. H. L., Eady, R. R., Smith, B. E.,
and Hasnain, S. S. (1999) J. Mol. Biol. 287,
1001 1009
541. Howes, B. D., Abraham, Z. H. L., Lowe, D. J.,
Brser, T., Eady, R. R., and Smith, B. E. (1994)
Biochemistry 33, 31713177
542. Murphy, M. E. P., Turley, S., Kukimoto, M.,
Nishiyama, M., Horinouchi, S., Sasaki, H.,
Tanokura, M., and Adman, E. T. (1995)
Biochemistry 34, 1210712117
543. Grossmann, J. G., Abraham, Z. H. L., Adman,
E. T., Neu, M., Eady, R. R., Smith, B. E., and
Hasnain, S. S. (1993) Biochemistry 32, 73607366
544. Peters Libeu, C. A., Kukimoto, M.,
Nishiyama, M., Horinouchi, S., and Adman,
E. T. (1997) Biochemistry 36, 1316013179
544a. Inoue, T., Nishio, N., Suzuki, S., Kataoka, K.,
Kohzuma, T., and Kai, Y. (1999) J. Biol. Chem.
274, 17845 17852
545. Veselov, A., Olesen, K., Sienkiewicz, A.,
Shapleigh, J. P., and Scholes, C. P. (1998)
Biochemistry 37, 60956105
546. Neese, F., Zumft, W. G., Antholine, W. E., and
Kroneck, P. M. H. (1996) J. Am. Chem. Soc. 118,
86928699
546a. Rasmussen, T., Berks, B. C., Sanders-Loehr,
J., Dooley, D. M., Zumft, W. G., and Thomson,
A. J. (2000) Biochemistry 39, 12753 12756
547. Gaykema, W. P. J., Hol, W. G. J., Vereijken, J.
M., Soeter, N. M., Bak, H. J., and Beintema, J.
J. (1984) Nature (London) 309, 2329
548. Cuff, M. E., Miller, K. I., van Holde, K. E., and
Hendrickson, W. A. (1998) J. Mol. Biol. 278,
855870
549. Hazes, B., Magnus, K. A., Bonaventura, C.,
Bonaventura, J., Dauter, Z., Kalk, K. H., and
Hol, W. G. J. (1993) Protein Sci. 2, 597619
550. Solomon, E. I., and Lowery, M. D. (1993)
Science 259, 15751581
551. Kitajima, N., Fujisawa, K., Fujimoto, C.,
Moro-oka, Y., Hashimoto, S., Kitagawa, T.,
Toriumi, K., Tatsumi, K., and Nakamura, A.
(1992) J. Am. Chem. Soc. 114, 12771291
552. Tian, G., and Klinman, J. P. (1993) J. Am. Chem.
Soc. 115, 88918897
553. Gahmberg, C. G. (1976) J. Biol. Chem. 251,
510515
554. Driscoll, J. J., and Kosman, D. J. (1987)
Biochemistry 26, 34293436
555. Clark, K., Penner-Hahn, J. E., Whittaker, M.,
and Whittaker, J. W. (1994) Biochemistry 33,
1255312557
556. Wachter, R. M., and Branchaud, B. P. (1996)
Biochemistry 35, 1442514435
557. Halfen, J. A., Jazdzewski, B. A., Mahapatra, S.,
Berreau, L. M., Wilkinson, E. C., Que, L., Jr.,
and Tolman, W. B. (1997) J. Am. Chem. Soc.
119, 82178227
558. Ito, N., Phillips, S. E. V., Yadav, K. D. S., and
Knowles, P. F. (1994) J. Mol. Biol. 238, 794814
559. Baron, A. J., Stevens, C., Wilmot, C.,
Seneviratne, K. D., Blakeley, V., Dooley, D. M.,
Phillips, S. E. V., Knowles, P. F., and McPherson,
M. J. (1994) J. Biol. Chem. 269, 2509525105
560. Whittaker, M. M., Kersten, P. J., Nakamura,
N., Sanders-Loehr, J., Schweizer, E. S., and
Whittaker, J. W. (1996) J. Biol. Chem. 271, 681
687
560a. Rogers, M. S., Baron, A. J., McPherson, M. J.,
Knowles, P. F., and Dooley, D. M. (2000) J.
Am. Chem. Soc. 122, 990 991
561. Dooley, D. M., Scott, R. A., Knowles, P. F.,
Colangelo, C. M., McGuirl, M. A., and Brown,
D. E. (1998) J. Am. Chem. Soc. 120, 25992605
561a. Itoh, S., Taniguchi, M., Takada, N., Nagatomo,
S., Kitagawa, T., and Fukuzumi, S. (2000) J.
Am. Chem. Soc. 122, 1208712097
562. Scott, R. A., and Dooley, D. M. (1985) J. Am.
Chem. Soc. 107, 43484350
563. Suzuki, S., Sakurai, T., and Nakahara, A. (1986)
Biochemistry 25, 338341
563a. Dove, J. E., Schwartz, B., Williams, N. K., and
Klinman, J. P. (2000) Biochemistry 39, 3690
3698
564. Williamson, P. R., and Kagan, H. M. (1987) J.
Biol. Chem. 262, 81968201
565. Uwajima, T., Shimizu, Y., and Terada, O.
(1984) J. Biol. Chem. 259, 27482753
566. Wachter, R. M., Montague-Smith, M. P., and
Branchaud, B. P. (1997) J. Am. Chem. Soc. 119,
77437749
567. Whittaker, M. M., Ballou, D. P., and Whittaker,
J. W. (1998) Biochemistry 37, 84268436
568. Toussaint, O., and Lerch, K. (1987) Biochemis-
try 26, 85678571
569. Wilcox, D. E., Porras, A. G., Hwang, Y. T.,
Lerch, K., Winkler, M. E., and Solomon, E. I.
(1985) J. Am. Chem. Soc. 107, 40154027
569a. Decker, H., and Tuczek, F. (2000) Trends
Biochem. Sci. 25, 392 397
570. Prigge, S. T., Kolhekar, A. S., Eipper, B. A.,
Mains, R. E., and Amzel, L. M. (1997) Science
278, 13001305
571. Menniti, F. S., Knoth, J., Peterson, D. S., and
Diliberto, E. J., Jr. (1987) J. Biol. Chem. 262,
76517657
572. Tian, G., Berry, J. A., and Klinman, J. P. (1994)
Biochemistry 33, 226234
573. Meyer, T. E., Marchesini, A., Cusanovich, M.
A., and Tollin, G. (1991) Biochemistry 30, 4619
4623
574. Messerschmidt, A., Luecke, H., and Huber, R.
(1993) J. Mol. Biol. 230, 9971014
575. Woolery, G. L., Powers, L., Peisach, J., and
Spiro, T. G. (1984) Biochemistry 23, 34283434
576. Farver, O., Wherland, S., and Pecht, I. (1994) J.
Biol. Chem. 269, 2293322936
577. Palmer, A. E., Randall, D. W., Xu, F., and
Solomon, E. I. (1999) J. Am. Chem. Soc. 121,
71387149
578. Lehn, J.-M., Malmstrm, B. G., Selin, E., and
blad, M. (1986) Trends Biochem. Sci. 11, 228
230
579. Keen, C. L., Lonnerdal, B., and Hurley, L. S.
(1984) in Biochemistry of the Essential Ultratrace
Elements (Frieden, E., ed), pp. 89132,
Plenum, New York
902 Chapter 16. Transition Metals in Catalysis and Electron Transport
References
580. Schramm, V. L., and Wedler, F. C., eds. (1986)
Manganese in Metabolism and Enzyme Function,
Academic Press, Orlando, Florida
581. Bartsevich, V. V., and Pakrasi, H. B. (1996) J.
Biol. Chem. 271, 26057 26061
582. Baker, A. P., Griggs, L. J., Munro, J. R., and
Finkelstein, J. A. (1973) J. Biol. Chem. 248,
800883
583. Goldman, A., Ollis, D. L., and Steitz, T. A.
(1987) J. Mol. Biol. 194, 147153
584. Kanyo, Z. F., Scolnick, L. R., Ash, D. E., and
Christianson, D. W. (1996) Nature (London)
383, 554557
585. Khangulov, S. V., Sossong, T. M., Jr., Ash, D.
E., and Dismukes, G. C. (1998) Biochemistry
37, 85398550
586. Zhou, G. W., Guo, J., Huang, W., Fletterick, R.
J., and Scanlan, T. S. (1994) Science 265, 1059
1064
587. Hirose, K., Longo, D. L., Oppenheim, J. J., and
Matsushima, K. (1993) FASEB J. 7, 361368
588. MacMillan-Crow, L. A., Crow, J. P., and
Thompson, J. A. (1998) Biochemistry 37, 1613
1622
589. Boldt, Y. R., Whiting, A. K., Wagner, M. L.,
Sadowsky, M. J., Que, L., Jr., and Wackett, L.
P. (1997) Biochemistry 36, 21472153
590. Mildvan, A. S. (1974) Ann. Rev. Biochem. 43,
357399
591. Villafranca, J. J., and Wedler, F. C. (1974)
Biochemistry 13, 32863291
592. Goodman, M. F., Keener, S., Guidotti, S., and
Branscomb, E. W. (1983) J. Biol. Chem. 258,
34693475
593. Deuel, T. F., and Prusiner, S. (1974) J. Biol.
Chem. 249, 257264
594. Schwartz, K., and Mertz, W. (1959) Arch.
Biochem. Biophys. 85, 292 295
595. Mertz, W. (1993) J. Nutr. Sci. Vitaminol. 123,
626633
596. Davis, C. M., Sumrall, K. H., and Vincent, J. B.
(1996) Biochemistry 35, 1296312969
597. Saner, G. (1980) Chromium in Nutrition and
Disease, Liss, New York
598. Frieden, E. (1972) Sci. Am. 227(Jul), 5260
599. Anderson, R. A., Bryden, N. A., and Canary, J.
J. (1991) Am. J. Clin. Nutr. 51, 864868
600. Davis, C. M., and Vincent, J. B. (1997) Arch.
Biochem. Biophys. 339, 335 343
601. ODell, B. L., and Campbell, B. J. (1970)
Comprehensive Biochemistry 21, 179226
602. Zhang, L., and Lay, P. A. (1996) J. Am. Chem.
Soc. 118, 1262412637
602a. Thompson, K. H. (1999) BioFactors 10, 4351
603. Zhitkovich, A., Voitkun, V., and Costa, M.
(1996) Biochemistry 35, 72757282
604. Anderson, R. A. (1994) in Risk Assessment of
Essential Elements (Merz, W., Abernathy, C. O.,
and Olin, S. S., eds), pp. 187196, ISLI Press,
Washington,D.C.
605. Conconi, A., Smerdon, M. J., Howe, G. A., and
Ryan, C. A. (1996) Nature (London) 383, 826829
606. Stearns, D. M., Wise, J. P., Sr., Patierno, S. R.,
and Wetterhahn, K. E. (1995) FASEB J. 9,
16431648
607. Stearns, D. M., BelBruno, J. J., and Wetterhahn,
K. E. (1995) FASEB J. 9, 16501657
608. Macara, I. G. (1980) Trends Biochem. Sci. 5,
9294
609. Nechay, B. R., Nanninga, L. B., Nechay, P. S.
E., Post, R. L., Grantham, J. J., Macara, I. G.,
Kubena, L. F., Phillips, T. D., and Nielsen, F.
H. (1986) Fed. Proc. 45, 123132
610. Chasteen, N. D. (1983) Struct. Bonding 53,
104138
611. Boyd, D. W., and Kustin, K. (1985) Adv. Inorg.
Biochem. 6, 311365
612. Tullius, T. D., Gillum, W. O., Carlson, R. M.
K., and Hodgson, K. O. (1980) J. Am. Chem.
Soc. 102, 56705676
613. Ryan, D. E., Grant, K. B., Nakanishi, K.,
Frank, P., and Hodgson, K. O. (1996)
Biochemistry 35, 86518661
614. Orgel, L. E. (1980) Trends Biochem. Sci. 5, X (Aug.)
615. Ryan, D. E., Grant, K. B., and Nakanishi, K.
(1996) Biochemistry 35, 86408650
616. Chasteen, N. D., ed. (1990) Vanadium in
Biological Systems, Kluwer Acad. Publ.,
Dordrecht, Germany
617. Winter, G. E. M., and Butler, A. (1996)
Biochemistry 35, 1180511811
618. Wever, R., and Krenn, B. E. (1990) in Vanadium
in Biological Systems (Chasteen, N. D., ed), pp.
81 97, Kluwer Acad. Publ., Dordrecht,
Germany
618a. Isupov, M. N., Dalby, A. R., Brindley, A. A.,
Izumi, Y., Tanabe, T., Murshudov, G. N., and
Littlechild, J. A. (2000) J. Mol. Biol. 299,
1035 1049
618b. Weyand, M., Hecht, H.-J., Kiess, M., Liaud,
M.-F., Vilter, H., and Schomburg, D. (1999)
J. Mol. Biol. 293, 595 611
619. Haupts, U., Tittor, J., Bamberg, E., and
Oesterhelt, D. (1997) Biochemistry 36, 27
620. Lindquist, Y., Schneider, G., and Vihko, P.
(1994) Eur. J. Biochem. 221, 139142
621. Hemrika, W., Renirie, R., Dekker, H. L.,
Barnett, P., and Wever, R. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 21452149
622. Chatterjee, R., Allen, R. M., Ludden, P. W.,
and Shah, V. K. (1997) J. Biol. Chem. 272,
2160421608
623. Zhang, M., Zhou, M., Van Etten, R. L., and
Stauffacher, C. V. (1997) Biochemistry 36, 1523
624. Elberg, G., Li, J., and Shechter, Y. (1994) J. Biol.
Chem. 269, 95219527
625. Li, J., Elberg, G., Crans, D. C., and Shechter, Y.
(1996) Biochemistry 35, 83148318
626. Heyliger, C. E., Tahiliani, A. G., and McNeill,
J. H. (1985) Science 227, 14741476
627. Johnson, J. L., Rajagopalan, K. V., and Cohen,
H. J. (1974) J. Biol. Chem. 249, 859866
628. Barber, A., Rodrguez-Gil, J. E., and Guinovart,
J. J. (1994) J. Biol. Chem. 269, 2004720053
629. Huyer, G., Liu, S., Kelly, J., Moffat, J., Payette,
P., Kennedy, B., Tsaprailis, G., Gresser, M. J.,
and Ramachandran, C. (1997) J. Biol. Chem.
272, 843851
630. Li, J., Elberg, G., Gefel, D., and Shechter, Y.
(1995) Biochemistry 34, 6218 6225
631. Mikalsen, S.-O., and Kaalhus, O. (1998) J. Biol.
Chem. 273, 1003610045
632. Hille, R. (1996) Chem. Rev. 96, 27572816
633. Romo, M. J., Archer, M., Moura, I., Moura, J.
J. G., LeGall, J., Engh, R., Schneider, M., Hof,
P., and Huber, R. (1995) Science 270, 11701176
634. Hille, R., and Nishino, T. (1995) FASEB J. 9,
9951003
635. Harris, C. M., and Massey, V. (1997) J. Biol.
Chem. 272, 83708379
636. Glatigny, A., Hof, P., Romao, M. J., Huber, R.,
and Scazzocchio, C. (1998) J. Mol. Biol. 278,
431438
637. Xiang, Q., and Edmondson, D. E. (1996)
Biochemistry 35, 54415450
638. Harris, C. M., and Massey, V. (1997) J. Biol.
Chem. 272, 2833528341
639. Glatigny, A., and Scazzocchio, C. (1995) J. Biol.
Chem. 270, 35343550
640. Blse, M., Bruntner, C., Tshisuaka, B., Fetzner,
S., and Lingens, F. (1996) J. Biol. Chem. 271,
2306823079
641. Canne, C., Stephan, I., Finsterbusch, J., Lingens,
F., Kappl, R., Fetzner, S., and Httermann, J.
(1997) Biochemistry 36, 97809790
642. Gladyshev, V. N., Khangulov, S. V., and
Stadtman, T. C. (1996) Biochemistry 35, 212223
643. Huber, R., Hof, P., Duarte, R. O., Moura, J. J.
G., Moura, I., Liu, M.-Y., LeGall, J., Hille, R.,
Archer, M., and Romao, M. J. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 88468851
644. Ori, N., Eshed, Y., Pinto, P., Paran, I., Zamir,
D., and Fluhr, R. (1997) J. Biol. Chem. 272,
10191025
645. Garrett, R. M., Johnson, J. L., Graf, T. N.,
Feigenbaum, A., and Rajagopalan, K. V. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 63946398
646. Johnson, J. L., and Wadman, S. K. (1995) in
The Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2271
2283, McGraw-Hill, New York
646a. Raitsimring, A. M., Pacheco, A., and Enemark,
J. H. (1998) J. Am. Chem. Soc. 120, 1126311278
646b. Brody, M. S., and Hille, R. (1999) Biochemistry
38, 6668 6677
647. Schindelin, H., Kisker, C., Hilton, J., Rajagopalan,
K. V., and Rees, D. C. (1996) Science 272, 1615
1621
648. Schneider, F., Lwe, J., Huber, R., Schindelin,
H., Kisker, C., and Knblein, J. (1996) J. Mol.
Biol. 263, 5369
648a. Stewart, L. J., Bailey, S., Bennett, B., Charnock,
J. M., Garner, C. D., and McAlpine, A. S.
(2000) J. Mol. Biol. 299, 593 600
648b. Rothery, R. A., Trieber, C. A., and Weiner, J.
H. (1999) J. Biol. Chem. 274, 13002 13009
648c. Temple, C. A., George, G. N., Hilton, J. C.,
George, M. J., Prince, R. C., Barber, M. J., and
Rajagopalan, K. V. (2000) Biochemistry 39,
40464052
648d. Li, H.-K., Temple, C., Rajagopalan, K. V., and
Schindelin, H. (2000) J. Am. Chem. Soc. 122,
7673 7680
649. Pollock, V. V., and Barber, M. J. (1997) J. Biol.
Chem. 272, 33553362
650. Rajagopalan, K. V. (1984) in Biochemistry of the
Essential Ultratrace Elements (Freiden, E., ed),
Plenum, New York
651. Kramer, S. P., Johnson, J. L., Ribeiro, A. A.,
Millington, D. S., and Rajagopalan, K. V.
(1987) J. Biol. Chem. 262, 1635716363
651a. Rajagopalan, K. V., and Johnson, J. L. (1992)
J. Biol. Chem. 267, 10199 10202
651b. Luykx, D. M. A. M., Duine, J. A., and de
Vries, S. (1998) Biochemistry 37, 11366 11375
651c. Liu, M. T. W., Wuebbens, M. M.,
Rajagopalan, K. V., and Schindelin, H. (2000)
J. Biol. Chem. 275, 1814 1822
651d. Feng, G., Tintrup, H., Kirsch, J., Nichol, M.
C., Kuhse, J., Betz, H., and Sanes, J. R. (1998)
Science 282, 1321 1324
652. Cramer, S. P., Solomonson, L. P., Adams, M.
W. W., and Mortenson, L. E. (1984) J. Am.
Chem. Soc. 106, 14671471
653. Berg, J. M., and Holm, R. H. (1985) J. Am.
Chem. Soc. 107, 917925
654. Cramer, S. P., Wahl, R., and Rajagopalan, K. V.
(1981) J. Am. Chem. Soc. 103, 77217727
655. Shah, V. K., Ugalde, R. A., Imperial, J., and
Brill, W. J. (1984) Ann. Rev. Biochem. 53, 231257
656. Orme-Johnson, W. H. (1985) Ann. Rev.
Biophys. Biophys. Chem. 14, 419459
657. Howes, B. D., Bray, R. C., Richards, R. L.,
Turner, N. A., Bennett, B., and Lowe, D. J.
(1996) Biochemistry 35, 14321443
657a. Caldeira, J., Belle, V., Asso, M., Guigliarelli, B.,
Moura, I., Moura, J. J. G., and Bertrand, P.
(2000) Biochemistry 39, 27002707
658. Rothery, R. A., Magalon, A., Giordano, G.,
Guigliarelli, B., Blasco, F., and Weiner, J. H.
(1998) J. Biol. Chem. 273, 74627469
903
References
659. Augier, V., Guigliarelli, B., Asso, M., Bertrand,
P., Frixon, C., Giordano, G., Chippaux, M., and
Blasco, F. (1993) Biochemistry 32, 20132023
660. Magalon, A., Asso, M., Guigliarelli, B.,
Rothery, R. A., Bertrand, P., Giordano, G., and
Blasco, F. (1998) Biochemistry 37, 73637370
661. Campbell, W. H., and Kinghorn, J. R. (1990)
Trends Biochem. Sci. 15, 315319
662. Garde, J., Kinghorn, J. R., and Tomsett, A. B.
(1995) J. Biol. Chem. 270, 66446650
663. Ratnam, K., Shiraishi, N., Campbell, W. H.,
and Hille, R. (1995) J. Biol. Chem. 270, 24067
24072
663a. George, G. N., Mertens, J. A., and Campbell,
W. H. (1999) J. Am. Chem. Soc. 121, 9730 9731
664. Stadtman, T. C. (1991) J. Biol. Chem. 266,
1625716260
665. Berg, B. L., Baron, C., and Stewart, V. (1991)
J. Biol. Chem. 266, 2238622391
666. Berg, B. L., Li, J., Heider, J., and Stewart, V.
(1991) J. Biol. Chem. 266, 2238022385
667. Stewart, V. (1988) Microbiol. Rev. 52, 190232
668. Sawers, G., Heider, J., Zehelein, E., and Bck,
A. (1991) J. Bacteriol. 173, 49834993
669. Gladyshev, V. N., Boyington, J. C., Khangulov,
S. V., Grahame, D. A., Stadtman, T. C., and
Sun, P. D. (1996) J. Biol. Chem. 271, 80958100
670. Boyington, J. C., Gladyshev, V. N., Khangulov,
S. V., Stadtman, T. C., and Sun, P. D. (1997)
Science 275, 13051306
Study Questions
1. Describe one or more metabolic functions of ions
or chelate complexes of ions derived from each of
the following metallic elements: Ca, Mg, Fe, Cu,
Ni, Co.
2. If the concentration of Cu, Zn-superoxide dismutase
(SOD) in a yeast cell is 10 M, the total copper
(bound and free) is 70 M, and the dissociation
constant for loss of Cu
+
from SOD is 6 fM, what
will be the concentration of free Cu
+
within the
cell? If the cell volume is 10
14
liter, how many
copper ions will be present in a single cell? See
Roe et al. (1999) Science 284, 805 808.
3. Outline the metabolic pathways that are utilized
by acetic acid-producing bacteria (acetogens) in
the stoichiometric conversion of one molecule of
glucose into three molecules of acetic acid. Indicate
briefly the nature of any unusual coenzymes or
metalloproteins that are required.
671. Khangulov, S. V., Gladyshev, V. N., Dismukes,
G. C., and Stadtman, T. C. (1998) Biochemistry
37, 35183528
672. George, G. N., Colangelo, C. M., Dong, J.,
Scott, R. A., Khangulov, S. V., Gladyshev, V.
N., and Stadtman, T. C. (1998) J. Am. Chem.
Soc. 120, 12671273
673. Shuber, A. P., Orr, E. C., Recny, M. A., Schendel,
P. F., May, H. D., Schauer, N. L., and Ferry, J. G.
(1986) J. Biol. Chem. 261, 1294212947
674. Muller, U., Willnow, P., Ruschig, U., and
Hpner, T. (1978) Eur. J. Biochem. 83, 485498
675. Rohde, M., Mayer, F., and Meyer, O. (1984) J.
Biol. Chem. 259, 1478814792
676. Ribbe, M., Gadkari, D., and Meyer, O. (1997)
J. Biol. Chem. 272, 2662726633
676a. Dobbek, H., Gremer, L., Meyer, O., and
Huber, R. (1999) Proc. Natl. Acad. Sci. U.S.A.
96, 8884 8889
676b. Gremer, L., Kellner, S., Dobbek, H., Huber,
R., and Meyer, O. (2000) J. Biol. Chem. 275,
1864 1872
676c. Hnzelmann, P., Dobbek, H., Gremer, L.,
Huber, R., and Meyer, O. (2000) J. Mol. Biol.
301, 1221 1235
677. Rajagopalan, K. V., and Johnson, J. L. (1992) J.
Biol. Chem. 267, 1019910202
678. Wahl, R. C., Warner, C. K., Finnerty, V., and
Rajagopalan, K. V. (1982) J. Biol. Chem. 257,
39583962
679. Nagahara, N., Okazaki, T., and Nishino, T.
(1995) J. Biol. Chem. 270, 1623016235
680. Nishino, T., Usami, C., and Tsushima, K. (1983)
Proc. Natl. Acad. Sci. U.S.A. 80, 18261829
681. Rech, S., Wolin, C., and Gunsalus, R. P. (1996)
J. Biol. Chem. 271, 25572562
682. Blanchard, C. Z., and Hales, B. J. (1996)
Biochemistry 35, 472478
683. Chan, M. K., Mukund, S., Kletzin, A., Adams,
M. W. W., and Rees, D. C. (1995) Science 267,
14631469
683a. Hu, Y., Faham, S., Roy, R., Adams, M. W. W.,
and Rees, D. C. (1999) J. Mol. Biol. 286, 899
914
684. Koehler, B. P., Mukund, S., Conover, R. C.,
Dhawan, I. K., Roy, R., Adams, M. W. W., and
Johnson, M. K. (1996) J. Am. Chem. Soc. 118,
1239112405
685. Mukund, S., and Adams, M. W. W. (1995) J.
Biol. Chem. 270, 83898392
686. Johnson, M. K., Rees, D. C., and Adams, M.
W. W. (1996) Chem. Rev. 96, 28172839
687. Yadav, J., Das, S. K., and Sarkar, S. (1997) J.
Am. Chem. Soc. 119, 43154316
688. Juszczak, A., Aono, S., and Adams, M. W. W.
(1991) J. Biol. Chem. 266, 1383413841
4. Suppose that you could add a solution containing
micromolar concentrations of Cu
2+
, Mn
2+
,
, Fe
3+
,
Co
2+
, Zn
2+
, and MoO
4
2
and millimolar amounts of
Mg
2+
, Ca
2+
, and K
+
to a solution that contains a
large excess of a mixture of many cellular proteins.
What would be the characteristics of the sites that
would become occupied by each of these metal ions?
How tightly do you think they would be bound?
5. What factors affect the rate of electron transfer
from an electron donor (atom or molecule) to an
acceptor?
6. List some mechanism that cells can use to combat
the toxicity of metal ions?
7. NADH peroxidase (p. 857, Eq. 15-59) does not
contain a transition metal ion. Propose a reasonable
detailed mechanism for its action and compare it
with mechanisms of action of heme peroxidases.
A
ABC transporters 417, 419, 839
Abequose 180s
abl 572, 576
Abortive complexes 466. See also Nonpro-
ductive complexes
Absorption spectra
of N-acetyltyrosine ethyl ester 123
of adenosine 205
of aromatic amino acid derivatives 123
bands at 500 nm 750
of cytidine 205
of cytochrome c 847
of proteins 122, 123
of uridine 205
of vitamin B
6
coenzyme 750
Acarbose 607, 607s
Acatalasemia 852
Acetabularia 21, 22
Acetaldehyde, hydration equilibrium 677
Acetan 179
Acetate
binding constants to metal ions 311
Gibbs energy of oxidation 297
Acetate kinase 661
Acetic acid
pK
a
values of 99, 293
Acetic anhydride, acylation by 124
Acetoacetate decarboxylase 705
Acetoacetyl-CoA 700s
Acetohydroxy acid isomeroreductase 712
Acetohydroxy acid synthase 734
Acetolactate 734s, 735
Acetone 705s
tautomerism 45
Acetyl adenylate 661, 720
Acetylation
coenzyme A for 720
of histones 549
of sulfanilamide 720
Acetylcholine
as neurotransmitter 720
receptor 422
Acetylcholinesterase 609, 634, 636
pseudosubstrate of 636
Acetyl-CoA (acetyl-coenzyme A) 506s, 508,
881
carboxylation of 724
corrinoid-dependent synthesis 876,
877
formation from fatty acids 511
synthesis of 660
reactions of, table 700
Acetyl-CoA carboxylase 725 729
biotin subunit 726
Acetylene hydratase 894
Acetylenic fatty acids 381
Acetyl-enzyme
in citrate lyase 704
from serine proteases 617
N-Acetylglucosamine (GlcNAc) 164, 599
N-Acetyllactosamine 183
N-Acetylmuramic acid (MurNAc) 165s, 599
N-Acetylneuraminic acid (NeuAc) 165s
N-Acetylornithine 742
Acetyl phosphate 799
O-Acetylserine sulfhydrylase 742
S-Acetyltransferase 661
N-Acetyltyrosine amide 617s
Acid(s)
enthalpy change of dissociation, table
293
Gibbs energy of dissociation, table 293
pK
a
values of, table 293
strengths of 9596
Acid base catalysis 469, 486 491
concerted 490
of mutarotation 487
Acid base chemistry
of amino acids 51 55, 304 307
of enzymatic action 486 491, 691 698
of nucleic acids 204 211
Acid catalysis
general 487 491
specific 489
Acid phosphatases 645
Acidity
of benzoic acids 308
of fatty acids 380
of groups in enzymes 486
of ketones 46
of phenols 308
of phenylacetic acids 308
Aconitase 676, 686 688, 861
active site structure 689
proton retention 686
cis-Aconitate 516s, 686s
isomerization of 697
Acrasins 557
Acridine yellow 223s
Acrosomal process 370
Acrylochlorin heme 845, 845s
Acrylonitrile 115
ACTH 54s
adenylate cyclase in action of 556
Actin 406
F-actin structure 338
thin filaments 369
Actinidin 618
-Actinin 370, 405
Actinomycetes 6
Activation
of enzyme(s) 471 478
of enzyme synthesis 536
noncompetitive 473 475
Activation cascade, pancreatic proteases 609
Active transport 379, 410, 417 420
of Na
+
and K
+
424
Activity of a solute 287
Activity coefficients 288
Acyl-ACP, dehydrogenation of 766
Acyl adenylate 507
Acyl carrier protein (ACP) 681, 704, 723
Acyl-CoA 526
Acyl-CoA dehydrogenase 766, 785, 789
Acyl-dihydrolipoyl derivative 736
Acyl-enzyme
in glyceraldehyde phosphate dehydro-
genase 775
intermediates 610 613
Acyl groups table of names 380
Acyl phosphates 507, 509, 660 661, 775s
Acylation 124
of proteins 559
in regulation 543
Acylglycerols 382
Acyltransferases 637
Adair equation 328, 354
Adaptor proteins
Grb2 577
Shc 577
Addition elimination sequence 526
Addition reactions 526, 530, 677 690
of double bonds, adjacent to
carboxylate 683 690
of double bonds, carbon-carbon
681 683
of double bonds, isolated 688
of double bonds, polarized 677 690
of enolate to carbon dioxide 705 710
facilitation by carbonyl group 681
isomerization 690
nucleophilic 123
reversibility of 690
Index to
Volume 1
Page numbers set in boldface refer to major discussions.
The symbol s after a page number refers to a chemical structure.
905
906 Volume 1 Index
stereochemistry 680, 681
Adducin 405
Adenine (Ade, A) 199s, 203
binding constants to metal ions 311
Adenosine (Ado) 203, 778s
absorption spectra of 205
acid base chemistry 204, 205
hypermodified 235
pK
a
value of 293
Adenosine 5'-phosphate. See AMP
Adenosine diphosphate. See ADP
Adenosine monophosphate. See AMP
Adenosine triphosphate. See ATP
Adenosine-diphosphate-ribose. See ADP-
ribose
S-Adenosylhomocysteine 592s, 778s
S-Adenosylhomocysteine hydrolase 778
S-Adenosylmethionine (AdoMet,
SAM) 591, 592s, 813, 875
decarboxylation 753, 754
reaction with catechol
O-methyltransferase 592
Adenosyltransferase, B
12s
870
Adenoviruses 247, 346
Adenylate cyclase 556557, 556s, 657
characterization of 556
toxic 548
Adenylate kinase 303, 654, 655s
fold 658, 659
functions of 655
Adenylate system 302 304
storage of energy 302
5'-Adenylic acid. See AMP
Adenylosuccinate lyase 685
Adenylylation 545
Adenylyl cyclases. See Adenylate cyclase
Adenylyl groups
transfer from ATP 660
Adhesion discs (septate desmosomes) 29
Adhesion proteins 187
Adipocyte 379s
ADP (Adenosine diphosphate) 536
in adenylate system 302 304
complexes with metal ions 296
dissociation as acid 288
intracellular concentration 304
P-31 NMR spectrum 642
pK
a
value of 293
in regulation 535
ADP-ribose (ADPR) 315, 778, 780
ADP-ribosylation 545, 778
ADP-ribosylation factors (ARFs) 559
Adrenaline (epinephrine) 534, 542, 553, 553s
in adrenergic receptor 535
-Adrenergic receptors 553, 558, 563
-Adrenergic receptors 553, 554
in asthma 553
in heart failure 553
receptor kinase 553
structure (proposed) 534, 555
topology 555
Adrenocorticotropin (ACTH) 54s
Adrenodoxin reductase 764, 788
Adsorption chromatography 103
Adsorption isotherm 326
Aequorin 314
Aerobes, obligate 8
Affinity chromatography 104 106
Affinity labeling 127, 477, 623
African sleeping sickness 787
Agammaglobulinemia 572
Agar-agar 170
Agarose 177s
activated 105
double helices in 177
gel networks of 175
Agglutinins (lectins) 427
Aggrecan 181
Aggregation of cells 187
Aglycone 167
Agonists 479
Agrin 437
Agrobactin 838
Alamethicin 414
Alanine (Ala, A) 52s
pK
a
values of 293
L-Alanine (Ala, A) 52, 52s
binding constants to metal ions 311
biosynthesis 517
-Alanine
in coenzyme A 722
D-Alanine 429
L-Alanyl-L-alanine
pK
a
value of 293
D-Alanyl-D-alanyl carboxypeptidase
625
L-Alanyl-L-valyl-L-methionine 57s
Albinism 887
Alcohol(s)
addition to carbonyl 677
dehydrogenation of 765 767
Alcohol dehydrogenase(s) 766
isoenzymes 774
stereospecificity of 479
structure 773
zinc in 680, 772 775
Aldaric acid 164
Aldehyde(s)
covalent hydrates 775
dehydrogenation of 766
oxidation of 775
Aldehyde dehydrogenases 776
Aldehyde oxidases 794, 890
Aldehyde oxido-reductase 891s
Aldimine. 434s See also Schiff bases
external, of pyridoxal phosphate 741
internal, of pyridoxal phosphate 741
from pyridoxal phosphate 742s
Aldo-keto reductases 774
Aldolase(s) 527, 679, 699703
Schiff bases in 732
type I 699
type II 699, 700
Aldolase A
enzyme-substrate complex 699
Aldol cleavage 698, 742
in epimerization 778
Aldol condensation(s) 527, 529s, 699, 700
Aldonic acid 164
Aldose-ketose interconversions 692
Aldose reductase 774
Algae 20 22
Algin 21
Alginates 170, 178
Alkali metal ions 206
Alkaline phosphatases 645
active site of 645
occurrence 645
Alkane(s) 382
Alkenes, hydration of 683
Alkyl shift, rearrangements with 527
Alkyl cobalt 867
Alkyl ethers 382
Alkyl cobalamins
formation 870
synthesis 869 870
Alkyl radicals, table of names 380
Alleles 184
Alliinase 742
Allolactose 168
Allose (All) 163s
Allosteric activation 539
Allosteric constant 476
Allosteric control
of enzymes 539 541
Monod-Wyman-Changeux model 349
Allosteric effectors 357, 475 477, 536
Allosteric inhibition 539
Allosteric properties
of enzymes 539 541
of repressor protein 539
Allosteric site 473
Allostery (allosterism) 475
Alloxan 121
Allylic rearrangement 527, 712
with condensation 527
Allysine 429
Alpha-carbon plot 64
Alpha effect on reaction rates 590
Alpha helix See Helix
Alport disease 438
Alternan 175
Alternative splicing of proteins 538
Altman, Sidney 84
Altrose (Alt) 163s
Aluminum
hydroxide complex, soluble 310
toxicity of 658
Amadori rearrangement 169
Ameba 18
Amebic dystentery 187
Amebocytes 23, 25
Ameloblasts 442
Ameloblastin 442
Amelogenins 442
Amicyanin 817, 883
Amide(s)
displacement reactions of 608
linkages in proteins 55 57
mechanism of formation 661, 662
resonance energy 299
weak basic properties 56
Amidination 124
Amiloride 421s
Amine(s)
addition to carbonyl 677
dehydrogenation of 766
Amine oxidases 766, 782
Amino acid(s) 5155
abbreviations of, table 52, 53
alpha (s) 41
aromatic, light absorption 749
C-terminal 57
configurations 42
dehydrogenation of 775
as dipolar ions 42, 51
essential 52, 53
N-terminal 57
pK
a
values of 55
table 52, 53
residues (definition) 57
separation by ion exchange
chromatography 104
sequences of 57
side chain groups 55
table 52, 53
907 Volume 1 Index
Amino acid decarboxylase 744
Amino acid dehydrogenase 766
Amino acid oxidases 782
D-Amino acid oxidase 478, 790, 791s
of kidney 781
Amino acid sequence 57 59
of aspartate aminotransferase 57, 202
of insulin 118
Amino acid side chains
torsion angles of 61, 62
Amino group(s)
reactions of 123
Aminoacrylate 744s
Aminoacyl-tRNA synthetases 662
p-Aminobenzoate 473s
-Aminobutyric acid (Gaba) 744
Aminocyclopropane carboxylate (ACC) 744s
-Aminoisobutyric acid 69
-Aminolevulinic acid 742, 745s
biosynthesis 742, 745
formation from glutamate-1-
semialdehyde 742
Aminomutases (vitamin B
12
) 872, 874
8-Amino-7-oxo-nonanoate 718, 742
Aminopeptidase(s) 117, 609, 627
-Aminopropionitrile 438
Aminopterin, inhibition of dehydrofolate
reductase 805
Aminotransferase(s) 737, 740 743, 747 753
half reactions 737
Ammonia
binding constants to metal ions 311
pK
a
value of 99, 293
Amoeba proteus 19
AMP (Adenosine 5'-phosphate) 203
in adenylate system 302 304
dissociation as acid 288
pK
a
value of 293
as protein handle 719
Amphipathic character 380, 401
Amplification
of DNA 260
of signals 567
-Amylase(s) 603, 605 608
action patterns of 606
calcium ion in 606
chloride ion in 606
pancreatic, 606s
pH profile of 471
subsites 606
-Amylase(s) 603, 608
Amylopectin 165, 170, 172, 173s
Amylose 165, 170, 172
double-helical form 173
Anaerobes
facultative 8
obligate 8
Anaerobic nucleotide reductases 864
Anaphylaxis 385
Anchors, membrane for proteins 402
diphytanylglyceryl 402
glycosylphosphatidylinositol
(phosphatidylinositol-glycan) 403,
403s, 523
myristoyl 402, 522
palmitoyl 522
polyprenyl 402
Angiosperms 29
Angiotensin II 563
Angiotensin-converting enzyme 625
Anhydrobiotic organisms 168
1,5-Anhydro-D-glucitol 165s
Animal cells, surroundings of 431 443
Animal tissue, composition of 31
Ankyrin 405
Annelida (segmented worms) 24
Annexins 316, 422, 571
Anomeric carbon atom 162
Anomeric effect 166
Antagonists 479
Antamanide 414
Na
+
complex 415s
Anthranilate 747s
Anthrax 548
Anti conformation of nucleosides 211, 212
Antibiotics 414, 479
anthracycline 223
Anticoagulation systems in blood 634
Anticodon 231
Anticodon loop in tRNA
hypermodified base in 231
Anticooperativity 352
in binding 329, 497
to metal ions 307
of protons 330
Antidotes for organic phosphates 636
Antienzymes 539
Antifreeze proteins 191
Antigen(s) 427
O-Antigen(s) 428
of Salmonella 180
Antigenic determinants 427
Antihemophilic factor 632
Antimetabolites 473
Antioxidants 818 820, 822 824, 826
vitamin E 721, 818, 819, 822, 823
Antiparallel structures 62

2
-Antiplasmin 634
Antiport process 410
Antithrombin 634
Antitrypsin 58
AP-1 (Activator Protein) 576, 577
Apoferritin 842
Apoptosis 574, 619
Apple motif of proteins 367
Apurinic acid 250
Apyrimidinic acid 255
Aquaporins 411, 412
Arabidopsis thaliana
genome 12, 29
Arabinan 177
Arabinogalactan 431
L-Arabinose (Ara) 163
binding protein 418
D-Arabinose (Ara) 163s
L-Arabinose isomerase 693
Arachidonic acid 381s, 565
Aragonite 440
Archaea 8
Archaeobacteria 7, 815
Area detector for X-rays 134
Arg-Gly-Asp (RGD) sequence 408
Arginase 888
manganese in 888
Arginine (Arg, R) 53s, 745
Arginine kinase 656
Arginine phosphate 303
Argininosuccinate lyase 685
Aromatic clusters in proteins 75
Aromatic groups, formation by elimination
689
Arrhenius activation energy 483
Arrhenius equation 483
Arrhenius, Svante A. 9, 83
Arrows, drawing 528, 529
Arsenate
phosphorylation, uncoupling of 596
similarity to phosphate 596
Arsenic compounds 387, 596597
toxicity of 596
Arsenicals in treatment of syphilis, 597
Arsenite, reaction with thiols 596
Arseno-ribofuranoside 596s
Arsenolysis 596
of glucose 1-phosphate 595
Arsenophospholipid 387s
Arthropods 24
Ascaris 25
chromosome number of 18
Ascarylose 180s
Ascomycetes 20
Ascorbate oxidase 887
Ascorbate peroxidase 852
Ascorbic acid 721
nutritional requirement 756
Ascus 20
Ash, amount from tissue 31
Asparagine (Asn, N) 53s
Aspartase 526
Aspartate 737s
binding constants to metal ions 311
biosynthesis 711
effect on biotin requirement 724
metabolism of 539
residues 312
Aspartate aminotransferase 57s, 135s, 753
absorption spectra 749
active site structure 744
atomic structure 750
catalytic intermediates, models 752
NMR spectra 149
quinonoid intermediate 750
Ramachandran plot 61
sequence 57
transamination 742
Aspartate ammonia-lyase 685
Aspartate carbamoyltransferase 348s
active sites 348
regulation 540
Aspartate -decarboxylase 753, 755
Aspartate -decarboxylase 746
Aspartate racemase 741
Aspartic acid (Asp, D) 52, 53s
biosynthesis 517
pK
a
value of 293, 487
Aspartic proteases 621625
Aspartyl aminopeptidase 621
-Aspartyl phosphate 539, 540s
Assays of enzyme activity 456
Assembly core of virus shell 365
Assembly pathway
Bacteriophage T4 367
for virus X174 366
Association constant. See Formation constant
Astacin 627
Asthma 26, 385
Asymmetric unit of crystal 134
Asymmetry of molecules 41 43
handedness 41 43
in oligomers 344
Ataxia telangiectasia 566, 574
Atomic force microscope 131
diagram of 131
Atomic force microscopy 131, 179, 392
Atomic structures of macromolecules. See
Three-dimensional structures
908 Volume 1 Index
pyridoxal phosphate enzymes 750
Atoms, table of sizes 41
ATP (Adenosine triphosphate) 536, 719 720
in acetylation of sulfanilamide 720
in adenylate system 302 304
analogs of 655
binding constants to metal ions 311
as coenzymes 719
complexes with metal ions 296
dissociation as acid 288
exchange-inert complex 643
formation during oxidation of
aldehyde, scheme 775
hydrolysis of 507
enthalpy of 296
Gibbs energy of 293, 297
pK
a
values of 293
proteases, dependent 627, 628
in regulation 535
screw-sense isomer 643
synthesis of 303, 507 515
ATPase 638
Ca
2+
424, 425s
gastric H
+
, K
+
- 424
H
+
, K
+
- 424
Na
+
, K
+
- [ (Na
+
+ K
+
)-activated ] 423
P-type 423
traffic 417
vacuolar 425
ATP, biosynthesis of
in fermentation reactions 661
uncoupling 636
ATP-citrate lyase 703
ATP synthase 425, 514, 659
Atrial natriuretic factor 561
Aurocyanin 883
Autocatalytic reaction 470
Autocrine effect 385, 574
Autophosphorylation of receptor 562
Auxilin 427
Avidin 257, 728
Avogadros number 283
Axis (axes) of symmetry 322, 323, 327 338
Axons 11
Azurins 883
B
Bacillus, definition of 6
Bacillus subtilis
genome 12
Bacteria 2. See also Specific genus and species
acetic acid 8
aerobes 10
anaerobic 8
autotrophic 8
binding to cells 186
branched fatty acids of 381
chemoheterotrophic 7, 8
chemolithotrophic 7
classification of 68
coats 431
composition of 31
electron micrograph of 4
flagella 6
gram-negative 6, 7, 180
transporters in 416 420
gram-positive 6, 7, 185, 190, 428
L forms 6
largest 8
methanogens 7
coenzymes of 813815
nutrition of 8
photoautotrophic 8
photosynthetic 7, 9
pili 335
Bacterial dehydratases
Fe-S clusters in 861
Bacterial diseases 7
Bacteriophages
filamentous (fd, f1, M13) 129, 334, 335
helical 244
temperate 248
Bacteriophage X174 244
Bacteriophage P22
tailspike protein 66, 66s
Bacteriophages T-even 244, 363
Bacteriophage T4(s) 363s
assembly pathway 367
lysozyme 599
Bacteriorhodopsin 71
Baculoviruses 247
Baltimore, David 84
Band 3 protein 404
of erythrocytes 420
Barrels
/ 77
, 71
65
Basal body of flagella 15
Basal metabolic rate 283
relationship to surface area 283
Basal stem cells 439
Base(s)
of nucleic acids 199
strengths of 95, 96
Base catalysis, general 487 491
Base composition of DNA and RNA 239
Base pairs 207 211
angle of twist 217
base inclination 217
of DNA 200
Hoogsteen 207, 231
propeller twist 217
strength of 209
tautomerism 211
thermodynamics of 209
Watson-Crick 207, 231
wobble 209, 231
Base quartets 207 211
Base triplets 207 211
Basement membrane 15, 72, 431, 436
Basidiomycetes 20
Basophils 26, 188
Bend 72 74
bcl 572
Bdellovibrio 3
Benzaldehyde, resonance energy 299
Benzene, resonance energy 299
Benzoic acid
acidity of 308
pK
a
value of 309
Benzoylformate decarboxylase 734
Berenil 225s
Beriberi 721
Beryllium, toxicity of 299
BES buffer 99
pK
a
value of 99
Beta cleavage. See Cleavage, beta
Beta condensation. See Condensation, beta
Beta helix See Helix
Beta oxidation 511, 512
of fatty acids 511, 512
Knoops theory 513
pathway 506
Beta propeller 67, 560, 764
Beta ribbons 241
Beta sheet 64
Beta spiral 74
Bicarbonate ion
from carbon dioxide 677
in carboxylation 710
into carboxyl groups 711
rates of incorporation 710
Bicarbonate sensor 557
Bicinchoninic acid 102
BICINE buffer 99
pK
a
value of 99
Biliverdin 852
Binding. See also Association
by macromolecules 327 330
cooperative processes 330 332
equilibria 325 332
of metal ions 307
multiple sites of 327 330
ordered 464
of oxygen. See oxygen (O
2
), binding of
random order 464, 467
Binding constant(s) 325 327. See also
Formation constant
anticooperativity 329, 330
intrinsic 328
for metal complexes, table 311
microscopic 328, 329
of protons to dianions, table 328
Binding curves 326
sigmoidal 331
Binding energy
relation to maximum velocity 617
Binding equilibria 325 332
dimerizing protein 350 352
Binding protein(s) 418
for histidine 419s
for hormones 536
Binding sites, multiple 327
Bioactivation of insecticides 636
Biochemical equilibria 304 316
Biochemical reactions
table of types 526, 527
Biocytin 724, 724s
Biological recognition 29
glycoproteins, in 186
Biomineral precursor 843
Biopolymers, hydrolysis of 292
Biopterin 803, 804s
as hydroxylation coenzyme 803
Biosensors 122
Biosynthesis 507, 515 517
of isoprenoid compounds 689
scheme 506, 509
Biosynthetic pathways
committed step 535
feedback inhibition 535
schemes 506, 509
Biotin 515, 516, 711, 721, 723 730, 723s
biosynthesis of 718, 745
in enzymes, table 724
mechanism of action 725 729
nutritional requirement 756
Biotin-binding proteins 728
Biotin carboxylase 724
Biotin carboxyl carrier protein 724
Biotin holoenzyme synthetase 724
909 Volume 1 Index
Biotinidase 725
BIS-TRIS buffer 99
pK
a
value of 99
1,3-Bisphosphoglycerate 596s
in glyceraldehyde-3-phosphate
dehydrogenase 775
2,3-Bisphosphoglycerate 357s, 361
effect on hemoglobin 357
Bisphosphoglycerate mutase 654
Bisulfite 779
reaction with uridine 251
with thiamin 731
Biuret 102s
Biuret method 102
BK virus 247
Black membrane 393
Bleomycin 225s
Bleomycin hydrolase 619
Blood
carbonic anhydrase 678
cell(s) 26
complement system 58, 414
proteins of 58
Blood-brain barrier 29
Blood clotting 631 634
anticoagulation systems 634
cascade mechanism 632
cofactors 631
inhibition of 622
intrinsic mechanism 633
Blood group determinants 182, 183
ABO system 184, 185
Cad antigen 185
Kell antigens 185
Lewis blood group 184, 185
MN antigens 185
P group 185
Rh antigen 185
Blood plasma
electrolyte concentrations 206
proteins 58
Blood platelets 26, 384
Blood type. See Blood group determinants
Blue-green algae. See Cyanobacteria
Blue multicopper oxidase 887
Bohr effect 357
Boltzmann constant 284
numerical value of 283
Bond(s)
angles 39 40
energies, table 299
lengths 40
Bonding
heterologous 333, 334
isologous 338
nonsymmetric 344
Bone 26, 440, 441
Bone morphogenic factor 443
Bordetella 548
Boric acid, pK
a
value of 99
Borohydride
in carbohydrate analysis 190
reduction by 124, 190
Boronic acids 622
Bradford method 102
Brain
of molluscs, organization 24
transporters in 416, 417, 420
Branch migration in DNA 229
Breast cancers 572
Brine shrimp 168
Bromelain 618
Bromoperoxidase(s) 856
Brnsted equation in basicity 490
Brnsted plot 309, 490
Brown, Michael S. 84
Brownian movement 461
Brucellosis 7
Bryophyta 29
Buchner, Edward 83, 456
Buffer(s) 9798
table 99
volatile 98
Bulge 66, 67s
Burkitts lymphoma 572, 576
Butyrylcholinesterase 634
C
Cacodylic acid, pK
a
value of 99
Cadherin(s) 187, 407
domains 408s
Cadmium 317
toxicity of 680
cADP ribose (cyclic ADP ribose) 564s
Caenorhabditis elegans
apoptosis 619
cuticle 440
genome 12
number of cells 24
Cage effect 462, 463
Caged phosphate 137
Calbindins 313
Calcineurin 646
Calciosomes 314, 563
Calcite 440
Calcitonin 314
Calcium 314 315
oscillations 315
release channel 314
in signaling 563 566
Calcium-binding protein(s) 312
Calcium-binding sites 312
Calcium carbonate
spicules 443
Calcium channels 422
Calcium ion(s) 31, 314, 315
activation of metabolic processes 314
concentration
in tissues 314
in serum 314
effects on equilibria 297
interaction with carboxylate groups 47
Callose 170, 174
Calmodulin 313, 315, 317s
in phosphorylase kinase 544
Calnexin 188, 520
Caloric values of food 282, 283
table 283
Calorie, definition of 282
Calorimeter
bomb 282
differential scanning 285
Calorimetry
animal 283
differential scanning of membranes
394
Calpactin See Annexin
Calpains 619
Calreticulins 313
Calsequestrin 313, 314
Calvin, Melvin 84
Cambium layer 29
Cambrian explosion 9
cAMP 536, 544, 544s, 556, 557
second messenger 557
Cancer 573 575
chemotherapy 224, 812
p53 as suppressor 581
Cancer cells
communicating junctions in 29
lack of contact inhibition 29
promoters 573
Capsid 244
Capsomers 244
Capsule 6
Captopril 624
Carbamate group (See also carbamino group)
80
in ribulose bisphosphate carboxylase
707 709
Carbamates
in proteins 80
Carbamino groups 678 See also Carbamate
group
in hemoglobin 358
in regulation 543
Carbamoylation
by sodium cyanide
Carbamylation See also Carbamoylation
of lysine 707
of ribulose bisphosphate carboxylase
707
by sodium cyanate 124
Carbanion mechanism of flavin dehydroge-
nase 790
Carbinolamine(s) 677
configurations 680
formation of 526
Carbocation(s)
from alkenes 683
oxocarbenium ion 598s
Carbocation mechanism
of fumarate hydratase 684
Carbocationic intermediate 595
Carbodiimide 105s
Carbohydrate(s)
analysis 188
caloric value 283
content, in tissues 31
hydrolysis of 189
methylation of 189
NMR spectra of 190 192
periodate oxidation of 190
Carbohydrate-binding proteins. See lectins
Carbon-14, properties of 110
Carbon-carbon bonds
addition to 681 683
cleavage of 530
reduction of 777
Carbon dioxide 358
carbon units, source of 810
hydration of 677, 710
reduction of 881
reduction, scheme 814
resonance energy 299
stability of 690
Carbon monoxide 553
in hemoglobin 358
oxidation of 881
resonance energy 299
Carbon monoxide dehydrogenase/acetyl-
CoA synthase 881, 882
910 Volume 1 Index
Carbon monoxide oxidase 893
Carbonic acid, pK
a
value of 99
Carbonic anhydrase 443, 676 678, 710
active site structure 679
mechanism 678
turnover number of 458, 678
Carbonium ion. See Carbocation
1,1'-Carbonyl-diimidazole 105s
Carbonyl group
addition of enolate anion 699 704
addition to 677
in displacement reactions 608 637, 698
facilitation of addition or elimination
681
formation 723 730
in metabolic sequences 681
reactivity 608
transient formation of 777, 778
N-1'-Carboxybiocytin
in action of enzyme 725
N-1'-Carboxybiotin 725, 726, 726s
dimethyl ester of 725
intermediate 725
-Carboxyglutamate 821, 821s
binding of calcium ions 80
formation of 820 822
Carboxylation
beta 527, 727 729
biotin-assisted 724
driven by cleavage of ATP 705
Carboxylesterases 635
Carboxyl (carboxylate) groups
in proteins 312
reduction of 776
resonance energy 299
Carboxylic acid
activation of 507
dehydrogenation of 766
Carboxylic acid reductase 893
Carboxyltransferase 724
of propionic acid bacteria 725
Carboxymethylation
of proteins 594
Carboxypeptidase(s) 117, 609, 625
zinc ion in 625
Carboxypeptidase A 64s
active site 626s
pK
a
values of 626
Carboxypeptidase Y 610
Carboxyphosphate 726, 726s
Carcinogenic compounds
adducts with nucleic acids 235
Carcinomas 573
Cardiolipin 384
Caricain 618
Caries 442
-Carotene 10, 390s
Carpal tunnel syndrome 756
Carrageenan(s) 170, 177, 178s
Carrageenin 22
Cartilage 26, 436
articular 182
matrix protein 437
Cascades
blood clotting 632
dephosphorylation 542
MAP kinase 576 578
pancreatic proteases 609
phosphorylation 542
regulatory 566, 567
Casein kinase 544
Caspases 619
Catabolism 536
and ATP synthesis 507 515
pathways of 506, 509
Catalase(s) 844, 852 855, 853s
catalytic cycles, scheme 855
compounds I, II 853 855
function of 852
hereditary deficiency 852
iron properties 853
mechanisms of catalysis 853 855
molecular activity of 852
turnover number of 458
Catalysis (acid, base) 469, 486
Catalysis (enzymatic)
acid and base 469, 486 491
covalent 494
mechanisms of 482 497
microenvironment 495
microscopic reversibility 486
multiple attack 606
nucleophilic 494
proximity factor 495
stereoelectronic effect 495
strain and distortion in 496, 497
tautomeric 490, 491
transition state 482 486
Catalyst, polyfunctional 490
Catalytic cycles. See also Other specific
enzymes and cell cycle
for catalase 855
for coenzyme M reductase 880
for glycine decarboxylase 809
for lysozyme 599
for -oxoacid dehydrogenases 798
for serine protease 613
for vitamin B
12
-dependent isomerases
872
Catalytic dyad 620
Catalytic subunits 348
Catalytic triad 134, 611 614, 619, 635
of serine proteases 611 614
Cataracts 169
Catechol 838
Catecholamine(s) 553
Catechol O-methyltransferase (COMT) 591
Cathepsin(s) 619, 621
G 610
K, in osteoclasts 619
Cations. See also Individual ions
active transport 422
channels 421, 422
Cat scratch disease 7
Cauliflower mosaic virus 247
CD (Cluster of Differentiation) 187, 407
Cech, Thomas R. 84
CED-3 619
Cell(s)
aggregation of 187
columnar epithelium 26
composition of 30 33
doubling time 470
fractionation of 98 100
junctions 28
metal binding sites in 311, 312
sizes of 11
squamus epithelium 26
Cell adhesion molecules (CAMs) 406
Cell constant organisms 24
Cell contacts 28, 29
Cell culture 26
Cell cycle 578 581
figure 580
Cell differentiation antigens (cluster of
differentiation, CD) 187, 407
Cell membranes 3
anions 420, 421
Cellobiohydrolases 602
Cell recognition 29
Cell respiration, inhibition 636
Cell signaling, table of abbreviations 577
Cell structure, compartments 552
Cell types
animal 25
plant 29, 30
Cell wall 5, 10, 15, 427 441
of bacteria 427 431
of plants 5, 177, 179, 443
in micrograph 13
Cellulases 602, 603
structural features 602
Cellulose 170, 171s, 172
content in spinach 31
Cellulose I 171s, 172
Cellulose II 172
Cellulose-binding domains 602
Cellusome 602
Cementum 442
Centipedes 24
Centrifugation 100
in CsCl gradients 249
density gradients 101
zone 101
Centrioles 10, 15
Centromeres, DNA of 228
Centrosome 627
Cephalopods, mesozoa in kidneys of 23
Ceramide 389s
Ceramide aminethyl phosphonates 389s
Cerebral ganglion in Platyhelminthes 23
Cerebrosides 388, 389s
Ceruloplasmin 58, 633, 839, 883, 887
CFTR 417
cGMP 561
Chagas disease 787
Chain elongation process 700
Chain-terminating inhibitors for DNA
sequencing 262
Channeled substrates 552
Chaperone (chaperonin) 339
calnexin 188
DnaK 518
GroEL 518
GroES 518
heat shock proteins 518
Chargaff, Erwin 200
Charge, effect on metal binding 311
Charge-relay system 612
Chelate complexes 310s
Chelate effect 310
Chelate stability 311
Chemical force microscopy 131
Chemical shift 138
hydrogen bonding, effect of 139
prediction 139
shielding of nuclei 138
table of NMR spectra 141
units of 138
Chemoheterotrophic bacteria 8
Chemolithotrophic bacteria 7
Chemoprevention 575
Chemoreceptors 562s
bacterial 561 563
Chemostat 470
Chemotaxis
911 Volume 1 Index
carboxylmethylation in 548, 563
CheA 562, 563
CheB 563
CheR 563
CheW 562, 563
CheY 562, 563
CheZ 563
Chiral acetates 704, 705
nonenzymatic preparation 704
Chiral methyl group 591
Chirality 42
Chitin 19, 170, 175
in exoskeleton of arthropods 24
Chitinases 599 602, 601, 603
Chitosan 175
Chlamydomonas 20, 22
Chloramphenicol 637s
Chloramphenicol acetyltransferase (CAT)
637
Chlorella 17, 22
Chloride channels 420, 421
band 3 protein 420
Chloride ion, activation of -amylases 606
Chlorin(s) 843
-Chloroalanine 739
Chlorobenzoyl-CoA dehalogenase 682, 683
Chlorocruorins 362
Chlorocruoroheme 845s
Chloroperoxidase(s) 855
Chlorophyll(s) 13
chelate effect 310
Chlorophyll a 10, 843
Chlorophyta 20
Chloroplast(s) 10, 14
in micrograph 13
ribulose bisphosphate carboxylase in
706
Cholera 7
Cholera toxin 546 548, 546s
in ADP-ribosylation 546
AMF images 131
Choleragenoid 333s
Cholesterol 390s, 392
elevated 16
as precursor of steroid hormones 696
from reduction of desmosterol 777
Chondroitin sulfate(s) 170, 175, 176s
Chordata 25
Chorismate 485s, 690s
biosynthesis of 689, 690
Chorismate mutase 485
Christian, W. 767
Christmas factor 632
Chromatin 11
Chromatography 102 105. See also specific
types
Chromium 888 889
complex with ATP 643
Chromosome(s) 11, 17
bacterial 3
homologous pair 18
micrograph of 371
numbers of 18
sizes of 18
Chymopapain 618
Chymosin 621
Chymotrypsin 66, 609
inhibition by TPCK 477
specificity 117
turnover number of 457
Chymotrypsinogen 609, 613, 615
activation of 609, 615
hydropathy index 78
Cibachron blue in affinity chromatography
106
Cilia 10, 15. See also flagella
eukaryotic structure 15
Ciliophora 19
Cinnamic acid 756s
Cinnamoyl-CoA thiol esters 682
Circular dichroism (CD)
of bound PLP and PMP 750
Cisplatin 218, 224s
Citrate 480s, 506s, 515, 516s, 686s
binding constants to metal ions 311
prochiral centers 480
Citrate cleaving enzymes 703 704
Citrate synthase(s) 527, 676, 700703, 702s
active site 703
mechanism 700
Citric acid
pK
a
values of 99
synthesis of 700
Citric acid cycle 506, 508, 515
discovery of 516
priming of 711
scheme 516
Citrulline 80
Citryl-CoA 703
Claisen condensation 699
Claisen rearrangement 485
Clathrin 426
Clathrin light chains 313
Cleavage
aldol 698
alpha 730
of amino acid side chains 745
beta 698 711
Clone of cells 26
Cloning vehicle 263
Clostridium acetobutylicum 705
Clostripain, specificity 117
Clp (caseinolytic protease) 628
Cluster of differentiation, CD 187
CMP (Cytidine 5'-phosphate) 203
Cnidaria (Coelenterata) 23
Coated membrane 426s
Coated pits 426
Coated vesicles 426
Coatomer 426
Cobalamin 867. See also Cyanocobalamin,
Vitamin B
12
in ribonucleotide reductase 871
Cobalamin-binding proteins 58
Cobalophilin 869
Cobalt 866 877
complex with ATP 643
Cocarboxylase 721
Coccidia 19
Coccus, definition of 6
Cocoonase 610
CO dehydrogenase/acetyl-CoA synthase
881
Coding strand (non-transcribing) of DNA
204, 237
Codon(s) 4, 236
initiation 236
stop 236
table 236
Coenzyme(s) 511, 719 757, 765 829
ease of dissociation from proteins 719
oxidative 719
Coenzyme A (CoA) 507, 720 723, 722s
functions of 722
Coenzyme A persulfide 790
Coenzyme A transferases 662
Coenzyme F
420
788
Coenzyme M 813s, 814, 815
Coenzyme M reductase
mechanism of 881
scheme 880
Coenzyme Q 818. See Ubiquinones
Cofactor F
430
879 881, 880s
Coiled coil
helical wheel 70
heptad repeat 70
superhelix 70
Colchicine 371, 371s
ColE1 248
Colicins 414, 418
Colipase 635
Colitose 180s
Collagen 15, 72, 181, 409, 432 441
alternative splicing 436
amino acid composition 72
biosynthesis of 433
characteristics, table 434
crosslinking 433, 434
gene location, table 434
genetic defects 438
hydrogen bonds in 72
location, table 434
sequence 72
structure 72, 432, 433
vertebrate types, table 434
Collagen fibril, electron micrograph 73
Collagen, type IV
electron micrograph of 437
structural model of 437
Collagenases 627
Collagenlike peptide 73s
Collenchyma 30
Color reagents 120
Combinatorial libraries 128, 129
Combustion, Gibbs energies of 297, 298
Communication between cells 26 29
Competitive inhibitors 471, 472
slow, tight-binding 472
Complement system
of blood 72, 414
of thioester-containing proteins 630
Complementarity
of amino and carbonyl 679
of enzyme surfaces 478
of substrate 478
Compound I, II
of peroxidases, catalases 853, 855
COMT. See Catechol O-methyltransferase
Concanavalin A 186, 342
Concentration gradient(s) 289, 410
Concerted mechanism 490
Condensation
aldol 699, 700
alpha 730
beta 527, 698 711
Configuration
of amino acids 41 43
entgegen (E) 43
rectus (R) 42
RS notation 42
sinister (S) 42
of sugars 163
zusammen (Z) 43
Confocal micrograph
of a cerebellum 130
of metaphase chromosomes 361
912 Volume 1 Index
Confocal scanning microscopy 129
Conformations of molecules 43 45
anti 43
anti-periplanar 43
boat (B) 166
eclipsed 44
envelope 166
Fisher projection 42
gauche 44
Newman projection 44
of nucleotides 211
of polysaccharide chains 170
quasi-equivalence 348
random coil 69
sickle 44
skew (S) 166
twist (T) 166
5- and 6-membered rings 166
C1 of sugars 166
chair 166
Conformational changes
cooperative 349 353
during dehydrogenase action 773
in enzymes 481
in proteins 81
role of buried hydrogen bonds 81
slow 477
Conformational maps 60
Conformational substates in proteins 496
Conidia 20
Conjugate bases 486
Connective tissue 72, 431
Connexons 29
Consensus sequences in DNA 265
Constitutive enzyme 538
Contact inhibition 573
of cells 29
Control elements of metabolic reactions 536
Controllability coefficient 537
Coomassie brilliant blue 102, 122s
Cooperative binding 352
of protons 331
of substrate 476
Cooperative changes in conformation 349
353
Cooperative processes 330 332, 334
Copper 65, 514, 882 887
as nutritional essential 882, 883
uptake regulation 883
Copper amine oxidases 816 817
mechanisms 817
Copper oxidases 885 887
Copper proteins, electron-transferring 883,
884
Copper sulfide cluster 884s
Coproporphyrin III 843, 845s
Cornified cell envelope 439
Corrin in transmethylation 592
Corrin ring 867, 868
Corrinoid-dependent synthesis
of acetyl-CoA 876, 877
Cosmarium 22
COSY-NOESY diagram 143
Cotransport (symport) process 411, 416, 417
Coulomb 283
definition of 47
Coulombs law 47
Countercurrent distribution 102
Coupled enzyme assay 768
Coupling constants. See also NMR spectros-
copy
Covalent catalysis 494, 591
Covalent modification
in control of metabolism 541 545
by dephosphorylation 541 545
by phosphorylation 541 545
reactions, table 543
Covalent radii 40
Covalent structure of proteins 57
Cow, genome 12
Cozymase 767
Crambin, amide exchange rate 149
Creatine kinase 656
transition state structure 640
Creatine phosphate 303
Crepenynic acid 381s
Cretinism 31
Creutzfeldt-Jakob disease 248
Crick, Francis H. C. 84, 200
Cristae of mitochondria 14
Crossing-over 18
Crosslinking 79
Crotonase. See Enoyl hydratase
Crowfoot Hodgkin, Dorothy M. 84
Cruciform structure in nucleic acids 229
Crustacea 24
Cruzain 619
Cryoenzymology 469
elastase 616
Cryoprotectants 191
Crystallins 169
Crystallography 131 137
electron 131
X-ray 132 137
Crystals, liquid 392 394
Crystal systems 133
Cubic symmetry
of oligomers 342
of pyruvate dehydrogenase 796
Cupric ion, complexes with ammonia 307
Curdlan 170, 174
Cuticle
of invertebrates 440
of plants 15, 30
in micrograph 13
Cutinase 134s, 635
Cyanase 717
Cyanate, carbamoylation by 360
Cyanide ion
adduct with NAD
+
779
complex with hemoglobin 358
reactivity of 590
-Cyanoalanine 438
Cyanobacteria 7, 10
Cyanoborohydride 124
Cyanocobalamin 867s, 869. See also Vitamin
B
12
reduction 869, 870
Cyanogen bromide 117
agarose, activation of 105
Cyclic adenosine 3',5'-monophosphate
as attractant for Dictyostelium 20
Cyclic ADP ribose 778
Cyclic AMP. See cAMP
Cyclic GMP. See cGMP
Cyclic symmetry 333
Cyclins, mitotic 580
cell cycle 524
D1 572
G1 or start 580
Cyclization
PLP-dependent enzymes 741
Cyclodextrins 171
Cyclodextrin glucanotransferase 603, 607
Cycloglutamate inhibitors 739s
Cycloguanil 805
1,2-Cyclohexanedione, reaction with
guanidinium groups, 126
Cyclophilin 488
human 488s
D-Cycloserine 739s
Cyclosporin 488, 488s
Cylinders 65, 66, 686
Cystathionine, 746s
formation 746
Cystathionine lyase 742
Cystathionine -synthase 744
Cystathionine -synthase 743, 746
Cystatins 622, 629
Cysteamine disulfide (cystamine) 549
Cysteine (Cys, C) 52, 53s
microscopic pK
a
values 306
zinc complex 680
Cysteine residues 53
crosslinking of 80, 521, 522, 786, 787
Cysteine proteases 618 619
Cystic fibrosis 16, 421
transmembrane conductance
regulatory 421
Cystine residues
beta elimination 85
Cystinuria 16, 417
Cytidine (Cyd) 203
absorption spectra of 205
reaction with hydrazine 252
Cytidine 5'-phosphate. See CMP
Cytidine diphosphate choline 720
5'-Cytidylic acid. See CMP
Cytoadhesins 409
Cytochalasin B 416s
Cytochrome(s) 845 848
Cytochromes b 847
Cytochrome b
562
847
Cytochrome c 846, 847, 847s
absorption spectra 846
structural studies 846
Cytochrome c oxidase 848, 887
Cytochrome c peroxidase 849s, 852
active site of 853
Cytochrome c
3
847s
Cytochrome oxidase 513
Cytochrome P-450 846
Cytokines 188
Cytokinin 235
Cytomegalovirus
micrograph 246
protease of 614
Cytoplasm, definition of 3
Cytosine (Cyt, C) 199s, 203
acid base chemistry 204, 205
Cytoskeleton 15, 368 372, 405
intermediate filaments 369
microfilaments 369, 370
microtubules 370
Cytosol, definition of 14
D
Daltons 4
Dansyl chloride 118, 118s
Darwin, Charles 9
Daunomycin 222, 223s
intercalation with DNA, figure 223
913 Volume 1 Index
DDT dehydrochlorinase 551
Dead-end complexes 466
DEAE-cellulose 104
DEAE-Sephadex 106
Deamidation
of asparagine 594
of asparaginyl residues 84
5-Deazaflavin
in hydrogenase 879
nonenzymatic oxidation of NADH 789
Deazariboflavin 5'-phosphate 789
Debranching enzyme 606
Debye-Hckel equation 288
Decarboxylase(s) 753 755
biotin-dependent 729
dialkylglycine 745
pyridoxal phosphate-dependent
enzymes 744, 745
pyruvoyl enzymes
Schiff bases in 732
thiamin-dependent 730 733
Decarboxylation 527, 705 710
catalysis by amines 705
catalysis by metal ions 705
of -oxoacids 698, 705, 742
Decarboxylation-transamination reaction
748
Decarboxylative elimination 526
Degree of hydration, effect on metal binding
311
Dehydroalanine 755, 757s
Dehydroalanine residue 754, 756
Dehydroascorbate 787
transport 416
Dehydroascorbic acid 416, 787
Dehydrogenase(s) 514, 765 802
conformational changes 771, 772
3-dimensional structures 768
mechanisms 768775, 770 771
nucleotide-dependent 777
stereospecificity 768 775
Dehydrogenation
of alcohols 765 767
of amines 775
of amino acids 775
reactions of flavoproteins, table 784
types of, table 766
Dematin 405
Denaturation of proteins 82
by guanidinium chloride 82
by sodium dodecyl sulfate 82
by urea 82
Dental plaque 442
Dentin 442
5'-Deoxyadenosyl cobalamin 867, 867s,
872
Deoxyhemerythrin, oxygenation of 862
Deoxyhemoglobin 850, 851
Deoxy-D-mannooctulosonic acid (KDO) 180s
Deoxyribonuclease 652 653
Deoxyribonucleic acid. See DNA
2-Deoxyribose 200s
1-Deoxyxylulose 5-phosphate 735, 736
Dephosphorylation, cascade 542
Depsides 23
Depsipeptide 414
Derepression of enzyme synthesis 536
Dermatan sulfate 170, 175, 176s
Dermatosparaxis 438
Dermis 439
Desaturases 863
Desmid 22
Desmosine 436, 436s
Desmosome 10
Desmosterol 777s
reduction to cholesterol 777
Desulforedoxin 859
Desulfuration of insectides 636
Dextrans 170, 174
Dextrin(s) 606
Diabetes mellitus 567
Diabetic nephropathy 438
Diacylglycerols 563, 564
scheme 565
in signaling 563 566
Dialkylglycine decarboxylase 743, 745
Dialkylnitrosamines 254s
Dialysis 100
1,4-Diaminopropane 745
Diastereoisomers (diastereomers) 43, 162
Diastereotopic atoms or groups 479
Diatomaceous earth 21
Diatoms 21
motion of 21
Dicoumarol 821, 821s
Dictyosome in micrograph 13
Dictyostelium discoideum 20
cAMP as messenger 557
genome 12
Dicyanocobalamin 870
3,6-Dideoxyhexoses, formation of 747
Dielectric constant 47
Diesenhofer, Johan 84
Diethanolamine, pK
a
value of 99
Diethylmalonic acid, pK
a
value of 99
Diethylpyrocarbonate reaction with histidine
126
Difference electron density map 136
Differential scanning calorimetric curves 395
Differentiation
among prokaryotes 9
cluster of (CD) 187, 407
Diffractometer 134
Diffusion 109, 410
exchange 410
facilitated 410
mobile carrier 414
saturation effect 410
through membranes 410, 414
Diffusion constant 461
Diffusion-controlled limit on reaction rates
462
Digestion 505, 507
Digoxigenin 257
Dihedral (D
2
) symmetry 338
D-gluco-Dihydroacarbose 607s
Dihydrofolate reductase 804 808, 810, 811
active site structure 807
inhibitors 805
mechanism 805 808
structure 805 808
Dihydrolipoic acid amide 784s
Dihydrolipoyl dehydrogenase 785, 796
Dihydrolipoyl transacetylase 796s
Dihydropteroic acid 473
Dihydropyridine receptor 314
Dihydroriboflavin 794
Dihydro U loop of tRNA 231
Dihydrouridine 234
Dihydroxyacetone 164s
Dihydroxyacetone phosphate 694s
Dihydroxyacid 697
Dihydroxyacid dehydratase
Fe-S clusters in 861
ortho-Dihydroxybenzene. See also Catechol
3,4-Dihydroxyphenylalanine (dopa) 80, 887,
887s
2,3-Dihydroxyproline 80
Diiron desaturases 863
Diiron oxygenases 863
Diiron proteins 862 864
Diiron-tyrosinate proteins 862 863
Diisopropylfluorophosphate (DFP,
diisopropylphosphofluoridate) 610s
Dimethylallyl diphosphate 712s
Dimethylallyl pyrophosphate, condensation
of 527
Dimethylarsine 876
Dimethylglutaric acid, pK
a
value of 99
Dimethylimidazole 868s
Dimethylsuberimidate 125s
crosslinking of proteins 125
Dimethylsulfoxide reductase 836s
Dimethylsulfoxide reductase family 890
Dinoflagellate 19
Dioldehydratase 871
stereochemistry 872, 874
Dioxetane 820
Dioxygen 850s
Dipeptidyl aminopeptidase 117
Dipeptidyl peptidase 619
Diphtheria toxin, ADP-ribosylation 548
Diphytanyl group 385
Diphytanyl tetraether lipids 387
Diploid cell 17
Diploid phase 18
Dipolar ion 41
Disaccharides 167169
hydrolysis of, Gibbs energies 169
Discoidin I 186
Diseases, bacterial 7
Displacement reactions, nucleophilic 123,
482, 530, 589 670
on carbonyl groups 608 637, 698
multiple 660
on phosphorus atom 637 657 See also
Kinases, phosphatases, transferases
in-line 638
permutational rearrangement 638
proteases 608 637
rates of 589 590
on sulfur atoms 659, 660
Dissipative structures 289
Dissociation constant(s) 304
apparent 293
Disulfide bridges (crosslinkages) 65, 80
cleavage of 115 116
cleavage with performic acid 115
locating 119
in proteins 80
reduction of 115, 785
Dithioerythritol 115
Dithionite ion 779
Dithiothreitol 98, 115, 822
Dityrosine linkages 81
DMSO reductase 890
DNA
A form 213, 214s
within ancient conserved regions 9
B form 213, 214s
hydration of 213
right handed helical structure 213
side-by-side structure 213
base composition of 239
base inclination 216
base roll 216
914 Volume 1 Index
bending of 218
cDNA 257
circular, in trypanosomes 219
cloning of 201
coding capacity 16
coding strand of 204, 237
conformational flexibility 216
consensus sequences 265
content in tissues 31
denaturation of 255
density gradient centrifugation 101
double helical structure 201
electron micrograph of 213
electron transfer in 218
electrophoresis 221, 249
extraction of 249
fingerprinting 259
footprinting 266
gyrase 220
H-DNA, proposed structure 226
helical twist 216
helix parameters 216
Holliday junction 228 230
hydration of 216
hydrolysis of 249, 251
hypersensitive sites 266
interaction with spermidine and
spermine 218
intercalation 221, 223
intrachain NOEs 268
knot, electron micrograph 219
light absorption 209
linking number 220
major groove 201, 213
melting curve 256
minor groove 201, 213
mispaired bases in 218
mitochondrial 14
map 262
nanochemical scale construction 230
nearest neighbor analysis 265
negative strand 244
NMR spectroscopy 266 270
1
H spectrum 269
non-coding (transcribing) strand 237
open reading frame 237
operator 237
overstretched 217
palindromes in 238
plectonemically interwound 220
polymorphism 259
propeller twist 216
R-DNA 226
reassociation curves, figure 256
reassociation kinetics 255
relaxed 221
repetitive 11
replicative form 244
sequence determination 260 266
sequences, understanding 265
sequencing gel, figure 263
single-stranded binding (SSB) proteins
239
solenoidal 220
structures 214, 223, 225, 228, 229,
240 243
supercoiled 219 222
superhelix density 221
tetraplex (quadruplex) structures 227
topological isomers 221
toroidal (solenoidal) 220
triplex 227
typing 259
underwound 220, 221
Watson-Crick structure 200
Z form 214s, 215, 239
enhancers in 216
DNA-binding motifs
in proteins 241
DNA-binding proteins
helix-turn-helix motif 239
DNA, circular 218, 219
linking number 220
writhe 220
DNA gyrase 220
DNA, mitochondrial 14
physical map 262
DNA photolyase 794, 802
DNA polymerase(s) 657
Thermus aquaticus (Taq) 260
zinc in 680
DNA - protein interactions 266
DNA-RNA hybrids 257
DNA sequencing 260 266
chain-terminating inhibitors 262
method of Maxam and Gilbert 264
Sanger dideoxy method. 262
DNA supercoiling estimating amounts 222
DNase. See Deoxyribonuclease
Docking protein for SRP 520
Domains in protein structure 74
Donnan equilibrium 420
Dopamine 744
Dopamine -monooxygenase 887
Double displacement reactions 595
ping-pong mechanism 595
in ribonuclease 647
Double helices 201, 213 218
conformational flexibility 216
hybrid DNA-RNA 230
Double reciprocal plot 460
Drawing arrows 528, 529
Drosophilia melanogaster 24
genome 12
Drosopterins 803, 804s
DSS or Tiers salt 138
Dyad axis 134, 213
Dye, reducible 300
Dynamic equilibrium 289
Dyneins 370
Dysentery, Shigella dysenteriae 7
Dystrophin 405
E
E (entgegen) configuration 43
E3 binding protein 796
Eadie-Hofstee plot 460
E-cadhedrin 574
Echinodermata 25
Ectoderm 23
Ectoenzymes 409
Ectopic proteins 573
Edelman, Gerald M. 84
Edman degradation 118
EDTA. See Ethylenediaminetetraacetic acid
Effector(s) of allosteric enzymes 473 475
EF-hand motif 313, 317
EGF (epithelial growth factor), definition of
577
EGF-like domains 367
Ehlers-Danlos syndrome 438
Eicosanoid 565
Eigen, Manfred 84
Elastase 66, 609, 610, 611s
cryoenzymology 616
-cylinder in 78
Elastic fibers 436
Elastin 15, 72, 436
Electrical double layer 400
Electric field jump methods 468
Electrochemical gradient 410
Electrochemical transference 311
Electrode(s)
calomel 300
reference 300
Electrode potentials 300 302
measurement of 302
observed 301
standard, table 300
Electrogenic 423
Electromotive force of electrochemical cell
300
Electron carriers 512 514, 767 802, 843
850, 883
Electron crystallography 131
Electron micrograph
of bacteria 4
of cell junctions 27
of plant cell 13
of starch granules 172
of viruses 246
Electron microscopy 129 131
Electron microscope tomography 130
Electron-nuclear double resonance (ENDOR)
399
Electron paramagnetic resonance (EPR, ESR)
spectra 398, 399
first derivative plot 398
of putidaredoxin 860
of tetramethylpiperidine-1-oxyl 398
Electron transfer
biological, mechanism of 848 850
coupling 849, 850
gating 849, 850
ionic equilibria, effects of 850
pathways 848
Electron transferring flavoprotein (EFT) 514,
794
Electron transport chain 512 515
of mitochondria 513
Electron-transport proteins 514, 767 802,
819, 843 850
Electronic noses 122
Electropherogram, single erythrocyte 107
Electrophilic center
definition of 589
in enzymes 754 757
Electrophilic reagents
reactions of nucleic acids 253 255
Electrophoresis 58, 106 108
agarose gel 106
capillary 106, 107
diagonal 120
pulsed field 249, 250
SDS-polyacrylamide gel 106, 113
zone 106, 249
Electrophoretic molecular sieving 106
Electrostatic effects
on pK
a
values 330
titration curves of proteins 330
transmission through aromatic rings
330
915 Volume 1 Index
Electrostatic theory 311
Element(s)
essential 33
in human body 31
toxic 33
Elimination reactions 526, 530, 677 690
beta, of cystine residues 85
conjugative 689
decarboxylative 689
facilitation by carbonyl group 681
of substituent 746
of PLP-dependent enzymes 742
reversibility 690
Ellmans reagent 125, 125s
Elongation factor EF-Tu 558
Elongin complex 564
Embden-Myerhof-Parnas pathway 508, 510
Embryos, plant 30
Emphysema 629
treatment of 631
Enamel 442
Enamelins 442
Enamelysin 627
Enamines, 528s
formation of 526
Enantiomers 164
definition of 41
Enantiotopic atoms or groups 479
Endergonic reactions 286
Endo ring conformation 212
Endocytosis 13, 425 427
Endoderm 23
Endoglucanases 602
Endoglycanases 593, 602
Endonexin See Annexin
Endoplasmic reticulum (ER) 10, 13, 14, 521
cisternae of 14
definition of 14
in micrograph 13
rough 14
smooth 14
Endosomes 13, 426
Endosperm 30
Endothelial cell-derived relaxing factor. See
Nitric oxide, NO
Endotoxins 431
cis-Enediol 693s
Enediol; Enediolate 694s, 695s
intermediate 693
Energy (E) 282
solar 289
sources of 507
storage and utilization of 302, 303
Enhancer 576
Enkephalin 54s
Enkephalin convertase. See Carboxypepti-
dase N
Enolase 685
active site 685s
metal ion requirement 685
relaxation of water protons 685
Enolase superfamily 686
Enolate anion 46, 46s, 681, 698 710
formation of 526
as nucleophile 527
from oxosteroid 696
phosphorylation of 706
resonance-stabilized 494
Enolic forms 530
Enolic intermediates 691 699
Enolpyruvoylshikimate-3-phosphate. See
EPSP
Enoyl-acyl carrier protein reductase 777
Enoyl hydratase 681
Enoyl reductase 766
Entamoeba, PEP carboxytransphosphorylase
of 706
Entamoeba histolytica 19
Enterobactin (enterochelin) 838s
Enterokinase. See Enteropeptidase
Enteron 24
Enteropeptidase 609
Enthalpy 282
of combustion 282, 283
Enthalpy change(s) 282, 283
estimation of 297
of dissociation, table 293
Entner-Doudoroff pathway 697
Entropy (S) 284, 285
of gases 284
from heat capacities 285
of racemization 284
relationship to microscopic disorder
284
table 285
units, definition of 284
Environment (thermodynamics), definition
of 282
Enzymatic reactions
catalysis 482 497
classification of 530
enolic intermediates 691 699
first order 457
integrated rate equations 457 460
pH, effect on 469 471
progress curve 456
reversible 458, 464 468
second order 458
stereoelectronic effect 495
velocity of 455 458
Enzyme(s) 1, 2, 455 503
activation of 471 478
active site 491 494
biotin-containing 724, 725
table 724
catalysis by 482 497. See also Catalysis
(enzymatic)
classification 497
conformational changes in 481
constitutive 538
dead-end complexes 466
debranching 606
effectors of 474
equilibrium 467
feedback inhibition, figure 540
history of 456
hydrogen bonding in 491 494
induced fit 481
inducible 538
inhibition of 471 478
competitive 471, 472
irreversible 477, 478
noncompetitive 473 475
inverting 593
isologous dimers 349
kinetic parameters 459
kinetics of action 455 471
mechanism
iso 466
ping-pong 465
reciprocating (flip-flop) 497
modifiers of 474
molecular activity of 457
molybdenum-containing 890 893
Fe-S clusters in 862
multisubstrate, kinetics of 464, 465
oligomeric 475, 497
pacemaker 535 537
processive action 602
prochiral centers 478 481
proofreading 482
pyridoxal phosphate, coenzyme table
743
rate equation for 460
rate of encounter with substrate
461 464
reaction rate 455 458. See Kinetics
regulation. See Enzyme regulation
retaining 593
saturation of 457
selenium-containing 822 827
specific activity of 457
specificity of 478 482
starch-hydrolyzing 605 608
stereospecificity 478 481
substrate binding 463, 464
tetrameric 342
thiamin-dependent, structures 733
thiamin diphosphate-dependent, table
735
Enzyme-activated inhibitors 682, 864
Enzyme assay, coupled 768
Enzyme-inhibitor complex 472
Enzyme regulation 535 537, 539 553, 571
581
allosteric control 539 541
by allosteric effectors 475 477
by covalent modification 541 545
by group transfer modification 545
549
by hormones 553 563
by ions 549
by thiol-disulfide equilibria 549
Enzyme-substrate (ES) complexes
formation and reaction of 458 461
Enzyme synthesis, genetic control of. See also
Protein synthesis
Eosinophils 26, 188
Epidermal growth factor (EGF) 544, 571
Epidermis 439
Epidermolysis bullosa 440
Epimerases 692
Epimerization of sugar, mechanisms 778
Epimers, definition of 163
Epinephrine (adrenaline) 542, 553, 553s
Episomes. See plasmid
Epithelial cells 29
Epitheliocytes 25
Epoxides, alkyation by 254
Epoxide hydrolases 591
EPR (electron paramagnetic resonance)
spectroscopy 398, 399
of glutamate mutase 873
in study of phosphotransferases 639
EPSP (enolpyruvoylshikimate-3-phosphate)
687s
synthase 687
Epstein-Barr virus 247
Equilibrium in carboxylation reactions 725
Equilibrium constant(s) 304
for association 325 332
for binding of metal ions 307
for carboxylation of propionyl CoA
725
changes with temperature 289
from concentrations 288
916 Volume 1 Index
effect of pH 293 297
for hydrolysis 292
prediction by Hammett equation 309
as ratio of rate constants 458
relationship to Gibbs energy change 2
87
stepwise, for complex formation 307
c-erb 571
v-erb 571, 572
Erhlich, Paul 473
ERK 577 579
Erythrocruorins 68, 362
Erythrocytes 26, 404 406
cytoskeleton 405
defects 826
integrin 405, 406
membrane skeleton 406
NADP
+
from 767
size of 11
Erythrocyte membranes
band 3 protein 420
proteins of 403
Erythrose 163s
Erythrulose 164s
Escherichia coli 3
composition of 31
DNA of 4
envelope structure 428
genome 12
Essential trace elements 31, 33, 837, 877 881,
887 894
Ester(s)
displacement reactions of 608
mechanism of formation 661, 662
resonance energy 299
Esterase(s) 526, 634 637
Ethanol 508
Ethanolamine, pK
a
value of 99
Ethanolamine ammonia-lyase 871
Ether lipids 382
Ether phosphatides 383s
Ethidium 223s
Ethidium bromide 221, 222
Ethylenediamine, binding constants to metal
ions 311
Ethylenediaminetetraacetic acid (EDTA) 98,
311s
N-Ethylmorpholine, pK
a
value of 99
ets 576
Eubacterium 3, 6
Euglena 19
Euglenophyta 20
Eukaryote(s), unicellular 18 22
Eukaryotic cells 11 15
definition of 2
iron uptake 839
structure 10
Evolution 9
of eukaryotes 15 18
Exchange diffusion 410
Exchange rates, amide protons 148
Exchange reactions of enzymes 595
Exergonic reactions 285
Exo ring conformation 212
Exocytosis 14, 427
Exoglycanases 593
Exponential growth rate constant 470
Extensins
glycosylation of 181
in plant cell wall 72
Extracellular matrix (ECM) 406, 427 441
F
FAD (flavin adenine dinucleotide) 511, 511s,
765, 780, 781s
FADH
2
529, 529s
Familial adenomatous polyposis 574
Faraday, numerical value of 283
Farnesyl group 402, 559
Fat(s). See also Triacylglycerol (triglyceride)
composition of 380
hydrolysis of 507
Fatty acid(s) 380 382
activation of 512
acyl CoA, derivatives of 507
biosynthesis of 722
branched chain 381
cyclopropane-containing 399
essential 721
in lipids 380
names of, table 380
oxidation 511
pK
a
values of 380
stability of 589
Fatty acyl-CoA derivatives 507
Fatty alcohols 380 382, 382
Favin 64s
Feedback inhibition 536, 539
Feedback repression 536
Ferments 456
Fermentation(s) 8, 508 511, 535
of glucose, Gibbs energy change 298
glyceraldehyde-3-phosphate dehydro-
genase in 775, 776
reactions 298, 510
Ferns 18
gametophyte and sporophyte forms 18
Ferredoxin(s) 793, 857, 858s
from chloroplasts 859
reduction potentials 857
Ferredoxin:NADP+ oxidoreductase 788, 794
Ferrichrome 838s, 839
Ferric uptake regulation protein 839
Ferriheme(s) 844
Ferritin 343s, 841s, 841, 842
Ferrochelatase 843
Ferroelectric crystals 486
Ferroheme(s) 844
Ferrous ion in aconitase 688
Ferroxidase center 843s
in ferritin 842
Ferryl iron in peroxidase and catalase 854
Fetal hemoglobin 362
-Fetoprotein 573
FhuC, D 839
Fiber cells 30
Fibrillin 440
Fibrin 631, 632
stabilizing factor 632
Fibrinogen 58, 409, 578, 632
Fibroblasts 25, 433
micrograph of 370
Fibrocytes 25
Fibronectin 407, 409
domain structure 409
Fibrous dysplasia 576
Fibulin 437
Ficin 618
Field flow fractionation 103
Fifth disease 244
Filaggrin 370, 439
Filamin 370
Filaria worms 24
Fimbriae 6. See also Pili
Fingerprinting. See also Peptide mapping
of DNA 259
of proteins 118, 360
First Law of Thermodynamics 282
First order reactions 457
Fischer, Edmond H. 84
Fischer, Emil H. 42, 83
Fischer projection formula 42
of monosaccharides 163
FK506 488
Flagella
bacterial 3, 6, 562
eukaryotic structure 15
Flatworms 23
Flavin(s)
chromatography of 103
electrophilic centers 790
fluorescence 103
half-reduced 791
isoalloxazine ring 790
metal complexes 794
properties of 793
Flavin adenine dinucleotide. See FAD
Flavin adenine diphosphate. See FAD
Flavin coenzymes 766, 780 795
modified 788, 789
reduced 794
Flavin radicals 792
color of 794
formation constant 794
Flavocytochrome b
2
782, 794, 847
Flavodoxins 793, 799, 800
Flavoprotein(s) 513, 788
dehydrogenation reactions 782 787
table 784
reduction potentials of 782
Flavoprotein dehydrogenases 789
free radicals 793
mechanisms 789 791
Flavoprotein oxygenases
free radicals 793
Flow of materials within cells 289
Flow cytometry 107
Flukes 24
Fluorescamine 120s
Fluorescence
in analysis 120
of flavins 103
recovery after photobleaching 400
of vitamin B
6
coenzyme 750
Fluorescence probe
polarity-dependent 396
Fluoride ion
inhibition of acid phosphatases 645
5-Fluorouracil 812
Fluorographs 103, 252
5-Fluorouridine 574
Flux
across membranes 410
in metabolism (analysis) 536, 537
FMN (Riboflavin 5'-phosphate) 513, 765, 780,
781s
Focal adhesions 405, 406
Fodrin 405
Folate coenzymes, 803, 804. See also Folic acid
Folate receptors 805
3-Fold axis 333
Folding patterns
of polypeptides 76, 77
917 Volume 1 Index
effects of sequence 78
Folic acid (pteroylglutamic acid) 473, 557,
721, 802
deficiency of 802
metabolic functions 802
nutritional requirements 756
in transmethylation 592
Folyl polyglutamates 803
Footprinting
DNA 266
hydroxyl radical 267
Foraminifera 19
Forces
acting between molecules 46 51
electrostatic 47
van der Waals 46
Formaldehyde 802
hydration equilibrium 677
reaction with nucleic acid bases 253
Formate. See also Formic acid
carbon units, source of 810
Formate dehydrogenase 824, 890, 892, 893
molybdopterin 892
Formate dehydrogenase H 893
Formation constants 304. See also Equilibrium
constants
macroscopic 328
of metal complexes 312
for Schiff bases 679
stepwise 327, 328
Formic acid 802
pK
a
value of 99, 293
Formiminoglutamate 810
Formiminoglycine 810
Formyl-THF synthetase 810
Formylglycine in sulfatases 659
Formylmethanofuran 814s
N-Formylmethionine 231
6-Formylpterin 803
10-Formyltetrahydrofolic acid (l0-formyl
THF) 810
Fos 70, 241, 572
definition of 577
fos 572, 576, 579
Fossils of bacteria and blue-green algae 17
Fourier transform infrared spectroscopy
of membranes 396
Fractionation
of cells and tissues 98 100
Fragile X syndrome 228
Frame-shifting 538
Franck-Condon principle 848
Franklin, Rosalind N. 200
Free energy. See Gibbs energy
Free radicals. See also Superoxide anion
radical, nitric oxide (NO)
in action of peroxidase and catalase
855, 856
in coenzyme M reductase 880
flavin 791 794
in metalloenzymes 850 886
in pyruvate formate-lyase 800 801
in pyruvate oxidase 736, 799
in reactions of vitamin B
12
869 876
in ribonucleotide reductases 864, 865
Freeze-fractured membranes, figure 405
Freeze fracturing 130
FreIp metalloreductase 839, 883
Frictional coefficient 109
of sphere 109
Fructofuranose (Fruf) 165s
Fructofuranosyl-enzyme 597s
Fructose (Fru) 162s, 175
Fructose 2,6-bisphosphate (Fru-2,6-P
2
)
in regulation of glycogen metabolism
537
beta form 541s
Fructose bisphosphate aldolases 699
Fructose 6-phosphate 535, 693s
Fructose 6-phosphate kinase 656
Fructose-1,6-bisphosphatase 645
Fructose-2,6-bisphosphatase 646
Fruit fly. See Drosophilia melanogaster
Fucose (Fuc) 165s
L-Fucose isomerase 693
Fucosyltransferase 184
Fucoxanthin 22
Fumarase. See Fumarate hydratase
Fumarase-aspartase family 685
Fumarate 481s, 516s, 683s
Fumarate hydratase (fumarase) 526, 683, 688
acid base catalysis 471
concerted reaction 685
Fumarase A 688
Fumarase B 688
Fumarase C 683
mechanism 683 685
pH dependence 684
rates of substrate exchange 684
turnover number of 683
Fumarate reductase 785
Fumarylpyruvate 690s
Function of state R 476
Fungal infections 20
Fungi 20
cell walls 443
Furanose rings 161, 175. See also Sugars
Furfural 251, 167s
G
G-quartets 208, 227
G
0
phase of cell cycle 579
Gabaculline 738s
Galactan 177
Galactanase 602
Galactolipid 387s
Galactosamine 176
Galactose (Gal) 162, 163s
metabolism of 778
Galactose chemoreceptor protein 186
active site 187s
Galactose oxidase 885
active site 886
mechanism of 887
tyrosine-cysteine cofactor 885, 886
Galactosemia 16, 169
-Galactosidase 602, 603
Galectins 186
Gametes 17
Gametophyte form of fern 18
Gangliosides 388
of cholera toxin 546
Gap junctions 29
Garrod, Archibald 16
Gas constant, numerical value of 283
Gastricsin 621
Gated pores 411
GDP dissociation inhibitors (GDIs) 559
Gel electrophoresis 112
relative molecular mass by 112
Gel filtration 100, 112
oligosaccharides, separation of 100
relative molecular mass by 112
Geminal diamine 751, 751s
Geminiviruses 244
Gene(s)
amplification of 538
average number of nucleotide pairs in
4
human, number of 16
imprinting of 234
sequence for aspartate aminotrans-
ferase 202
structural, definition 1
Gene therapy 16, 203
sickle cell disease 361
Genetic code 1, 236 239
tables 236, 237
variations 237
Genetic diseases 2, 16
Genetic recombination 6, 230
Genomes
bacterial 3 5, 201
changing 17
definition of 4
human 201
sequences 12,
table of 12
of viruses 201
Geometrical isomers 43
Geometry of orbitals
effect on metal binding 311
Geranylgeranyl group 402
Giardia lamblia 12, 19
Gibbs energy (Free energy) 285, 286
of activation (G

) 483
of association 325 332
of dissociation 293
of acetic acid 50
of formation 286, 292 298
for biochemical substances, table
290
of RNA helix, table 210
of hydrolysis 283
table 294
in hydrophobic interactions 50
linear relationships 308
of oxidation
for biochemical substances, table
290
by NAD
+
, table 290
partial molar 287
summing 286
Gibbs energy change(s)
for dilution 287
and equilibrium constant 287
for fermentation 298
for oxidation-reduction reactions 300
for subunit association 333
Gingipains 619
Gla proteins 821
Glial cells 26
Glial filaments 369
Globin, evolution of gene 17
Globular proteins, properties 59
Globulin(s) 101, 58
Glucagon 542
adenylate cyclase in action of 554
receptor 554
-Glucanases 602, 603
Glucans 172 175
-1,3-linked 174
918 Volume 1 Index
Glucarate dehydratase 686
D-Glucitol. See D-Sorbitol
Glucoamylase 603, 607
, barrel structure, 77s
active site structure 608
Glucocerebrosidase 603
Glucokinase 538, 568
Gluconeogenesis 515
pathway 509
regulation of 541
5-Gluconolactone 604
Gluconolactone 604s
Glucosamine (GlcN) 164, 176
Glucose (Glc)
caloric value 283
Glucose (D-glucose, Glc) 161s, 162s
concentration in blood 161
diastereomers 162
equilibrium mixture in water 162
fermentation of 298
ring form 162s
Glucose (L-glucose, L-Glc), 161s
Glucose 1-arsenate as intermediate in
arsenolysis 595
Glucose 1,6-bisphosphate 653, 653s
Glucose dehydrogenase(s) 814
Glucose oxidase 782
Glucose-6-phosphatase 645
Glucose 6-phosphate 506s, 507, 535, 693, 693s
Gibbs energy of hydrolysis 293
pK
a
value of 293
Glucose-6-phosphate dehydrogenase 770,
776
Glucose-6-phosphate isomerase
efficiency of 692
turnover number of 692
Glucose tolerance factor
chromium in 888
Glucose transporters 415, 416
GLUT1, topology diagram 416
GLUT4, response to insulin 416
Glucosidase
alpha 603, 606
alpha 1,6 606
beta 602, 603
Glucosylasparaginase
(aspartylglucosaminidase) 620
Glucuronic acid; glucuronate (GlcA) 164s,
165s, 176
epimerization 177
GLUT1GLUT7. See Glucose transporters
L-Glutamate 737s
biosynthesis 711
Glutamate dehydrogenase 770, 775
inhibition by 2-oxoglutarate 780
Glutamate mutase 871
stereochemistry 874
Glutamate-1-semialdehyde 742
Glutamate synthase 788
Glutamic acid (Glu, E) 52, 53s. See also
Glutamate
pK
a
value of 487
Glutamine (Gln, Q) 53s
Glutamine PRPP amidotransferase 620
Glutamine synthetase 342, 662
Glutaredoxin 522, 549, 786, 787, 864
Glutathione 521, 549, 550 552, 550s
biosynthesis 551
protective functions 551, 786, 826
Glutathione peroxidase 550, 826, 856
selenium in 823
Glutathione reductase 785, 785s
reaction mechanism 791
scheme 792
Glutathione S-transferases 550, 682
Glycans 169 180. See also Polysaccharides
N-linked 183
Glycation 169
Glyceraldehyde 163s
Glyceraldehyde 3-phosphate 506s, 508, 694s
oxidation of 508 510
Glyceraldehyde 3-phosphate dehydrogenase
63s, 766
in fermentation reactions 775, 776
formation of NADH-X 780
in oxidation of aldehydes 775, 776
thiol group of 677
D-Glycerate dehydrogenase, stereochemistry
768
Glycerol 380s
accumulation in fish and insects 191
in lipids 380
Glycerol 1-phosphate, pK
a
value of 293
Glycerol dehydratase 871
Glycerol oxidase 886
sn-Glycerol 3-phosphate 383s
Glycerolteichoic acids 431s
L-Glycero-D-mannoheptose 180s
Glycerophospholipids 383
Glycerylcysteine 428, 428s
Glycine (Gly, G) 52, 52s, 745
binding constants to metal ions 311
cleavage system, scheme 809
decarboxylation of 745
pK
a
value of 99
reductases 753, 755
Glycineamide, binding constant to metal ions
311
Glycine decarboxylase 743, 745, 809
Glycine decarboxylase-synthetase system
808
Glycine N-methyltransferase 591, 593s
Glycine reductase 824
Glycocalyx 6
Glycogen 165, 170, 173, 173s, 603
breakdown of 537
content in rat liver 31
methylation analysis 196
13
C NMR 174
particles in cytoplasm 10, 174
STM image of 174
storage of 537
synthesis 509
Glycogen phosphorylase 541 545, 543s, 604,
605, 746
catalytic site 605s
conformational states 604
control of 541 545
effect of insulin 571
regulation of 539, 604
Glycogen synthase
effect of insulin 571
Glycogenin 174
Glycogenolysis 545
Glycoglycerolipids 387
Glycolate 707
Glycolate oxidase 782
Glycolipids 387, 392
biological recognition 161
Glycolysis 506, 508 511, 699
regulation of 541
Glycophorin A 404
Glycophorin C 405
Glycoproteins 15, 180 188, 404, 405
biological recognition 161
blood group determinants 184
high mannose type 183
linkage of saccharides to 181
O-linked 181, 182
release of oligosaccharides 188
Glycosaminoglycan 181
repeating disaccharide units of 176
Glycosidases
beta 602
endo 605
Glycosides
displacement reactions of 593
N-Glycosides (glycosylamine) 169
Glycosidic acetal linkages 168
Glycosidic linkages 780
Glycosylamine 169
Glycosylation
definition of 169
Glycosyl enzyme intermediates 595
Glycosyl esters 595
Glycosyl exchange reactions 593
Glycosylphosphatidylinositol (GPI) anchors
403
Glycosyltransferase(s) 184, 526, 604, 608
catalytic groups, table 603
Glycyl endopeptidase 618
Glycylglycine
binding constants to metal ions 311
copper complexes 312s
pK
a
value of 99
Glycyl radical 864
Glyoxalase 527, 551, 698
Glyoxalase I, II 698
Glyoxylate 705s
Glyoxylate carboligase 736
Glyoxylic acid
in polysaccharides 179
Glyoxysomes 14
Glyphosate 687s
GMP (Guanosine 5'-phosphate) 203
Gold 318
Goldstein, Joseph L. 84
Golgi apparatus 10, 521
definition of 14
Golgi body in micrograph 13
Golgi, Camillo 83
Gonadatropin-releasing hormone
NMR spectrum 145
Gonorrhea 7
Goodpasture syndrome 438
G protein(s) 534, 557 561
bovine brain G
i
560s
function, scheme 557
monomeric 558
three-dimensional structures 559 561
Gramicidin A 414
Gramicidin S 54s
Gram-negative bacteria 6, 7
outer membrane 428 431
Gram-positive bacteria 6, 7, 185, 190, 428
Gram stain 6
Granulocytes 26
Granzymes 610
Grb2 577, 578, 580s
Green plant, composition of 31
GroEL 339, 518
GroES 339, 518
Ground substance between cells 26
Group transfer potential(s) 292, 293
effect of metal ions 296, 297
of pyrophosphates 636
919 Volume 1 Index
Group translocation 411
Growth, control of 578
Growth factors 571 576, 722
Growth rates of cells 470
Growth regulating hormones 567 571
GTP (guanosine triphosphate)
as coenzyme 719
GTPase activating proteins (GAPs) 559
Guanidination 124
Guanidinium chloride 82s
Guanidinium groups 53, 55s
Guanidinoacetate methyltransferase 591
Guanine (Gua, G) 199s, 203
hydration of 215
quartet 208
Guanine nucleotide binding proteins. See G
proteins
Guanine nucleotide dissociation stimulators
(GDSs) 559
Guanosine (Guo) 203, 234s
absorption spectra of 205
Guanosine 5'-(, -imido) triphosphate
(GppNp) 558s
Guanosine5phosphate (GMP,Guanosine 5'-
guanylic acid) 203
Guanylate cyclase 561, 657
Guanylin 548, 561
Guanylyl cyclase. See Guanylate cyclase
Guard cells 30
Gulose (Gul) 163s
Gustducin 558
Gutta-percha 389
Gymnosperms 29
H
Haemophilus influenzae
genome sequence 12
Hageman factor 632
Hairpin ribozyme 649
Haldane, J. B. S. 9
Haldane relationship 464, 465
Half-life 457
Haloacid dehalogenase(s) 590
mechanism of 590
Haloalkane dehalogenase(s) 591
active site structure 591
Halocyanin 883
Haloperoxidases 855, 889
Hammerhead ribozyme 649, 651s
mechanism of action 651
Hammett equation 308
for dehydrogenation reactions 771
for multisubstituted compounds 309
table of constants 308
Handedness of molecules 41 43
Haploid
definition of 5
phase 18
Haptoglobin 58
Harden, Arthur 83
Haworth structural formulas 164
Hay fever 26
Heart disease 16
Heat 282
of combustion 282
flow of 284
Heat shock proteins (chaperonins) 518
Heisenberg uncertainty principle 138
HeLa strain of human cancer cells
30 S ribosomal subunit 26
Helical structure
polyethylene 44
polyfluoroethylene 44
Helical wheel 69s, 70
Helix (helices). See also Superhelix
3
10
69, 70
amphipathic 79, 401
hydrogen bonds in 68
one-start 334
Pi 69
pitch of 68
primary 334, 337
properties of 69
radial projection 334
in serum albumin 71
stacking of 70
structures of 334
torsion angles of 68 71
Helix (helices) 68s, 69, 78
as helical wheel 69s, 70
Helix (helices)
parallel 65
Helix bundles
four helix 71
Helix-turn-helix motif 76
Hemagglutinin 186
Heme 68, 353, 514, 844, 850, 851
covalent linkage to proteins 719
imidazole group in 851
Heme a 844, 845s
Heme c 845, 845s
Heme d 845, 845s
Heme o 845s
Heme proteins 843 845
reactions with hydrogen peroxide
850 857
reactions with oxygen 850 857
Heme-oxygen complexes 851
Hemerythrin 362, 852, 862
active site, 863s
Hemiacetals 161, 162, 677. See also Glycosidic
linkages
formation of 526
Hemicelluloses 177, 602
Hemiketals 677
formation of 526
Hemimercaptals 677
formation of 526
Hemin 844
Hemochromatosis 840
Hemochrome(s) 844
Hemocyanins 885
Hemoflagellates 19
Hemoglobin(s) 17, 353 359, 844
abnormal 354, 359 362
2,3-bisphosphoglycerate, effect 357,
358
carbamino groups in 358
comparative biochemistry 362
competing ligands 358
fetal 362
fingerprints of 359
folding pattern 354
oxygen binding cooperativity 851
in oxygen transport 354
oxygenation curves 355
pK
a
values of 357
salt bridges 357
saturation curve 355
structural changes during oxygenation
354 357
subunits 353
thermodynamic function for
oxygenation, table 355
Hemoglobin mutants 354, 359 362
F 362
H 354
M 359
S 359, 360
Hemopexin 58, 627
Hemophilia A, B 633
Hemosiderin 842
Henderson-Hasselbalch equation 97
Heparan sulfate 175 177, 176s
Heparin 176s, 181
anticoagulant properties of 177
pentasaccharide segment 177s
Heparin cofactor II 177
Hepaticae 29
Hepatitis A virus 247
Hepatitis B virus 244
Hepatitis delta virus 247
HEPES buffer 99
pK
a
value of 99
Heptad in coiled coil 71
Heptanoyl threonine phosphate (HTP,
coenzyme B) 881
Herbicides 678
Herbicides
glyphosate 687
Herpesviruses 247
Heterocysts 10
Heteroduplex 256
Heterologous bonding interaction 333, 334
Heterologous interactions 342
Heterologous square 352s
Heteropolysaccharide(s)
of animal body 175 177
definition of 169
of plants 177 179
Heterotropic interactions 476
Hexagonal phase
of alkanes 394
of lipid bilayer 394, 395
Hexokinase 467, 518, 654, 656
in regulation 535
Hexoses 162
High performance liquid chromatography
(HPLC) 103
High potential iron proteins (HIPIP) 857
Hill plot 331, 332
Hippuric acid 513s
Hirudin 634
Histamine 745
Histidine (His, H) 53s
binding constants to metal ions 311
catabolism of 810
diethylpyrocarbonate, reaction with
127
pK
a
value of 487
titration curve 97
Histidine ammonia-lyase 755
Histidine-binding protein 419s
Histidine decarboxylase
active site 755
mechanism 754
proenzyme 754
Histidinohydroxymerodesmosine 434, 434s
Histone(s) 204
complex with DNA 221
H2A, ubiquitination of 525
920 Volume 1 Index
HIV (human immunodeficiency virus) 248
HIV-1 protease 624s
catalytic mechanism 625
Hofmeister series 311
Holley, Robert W. 84
Holliday junction 229s
branch migration in 229
in DNA 228 230
immobile 230
Homeostatic mechanisms for metal ions 307
Homoaconitase 688
Homocitrate 700s
L-Homocysteine 746, 746s, 778s
Homogenization 98
Homology, definition of 59
Homopolysaccharide(s) 175
definition of 169
Homotropic interactions 476
Hoogsteen base pair 207
reversed 208
Hookworms 24
Hopkins, Frederick G. 83
Hormone(s). See also Specific hormones
receptors for 479, 536, 553 563
regulation of enzymes 553 563
Hormone-sensitive lipase
effect of insulin 571
Horseradish peroxidase 852, 856
Horseshoe crabs 24
Housing silk 67
HPLC. See High performance liquid
chromatography
HSQC 144
Huber, Robert 84
Human body, elements in 31
Human genome 12, 201
Human glucose transporters 416
figure 416
Human immunodeficiency virus. See HIV
Hyaluronan 170, 175 177, 176s
hydrogen-bonding 176
properties of 175
repeating disaccharide units of 176
Hyaluronate lyase 686
Hyaluronic acid 15, 182. See also Hyaluronan
Hyaluronidase. See Hyaluronate lyase
Hybrid duplexes 256
Hycanthone 223s
Hydra 23, 24
Hydration
of alkenes, mechanism 683
effects on biochemical equilibria 50
of ions 50, 311
of nucleic acid bases 213
of polar molecules 50
Hydration layer 51
Hydride ion
shift of 527
transfer of 765
Hydride-transfer mechanism 789
Hydrocarbon(s) 380 382
-Hydrogen
loss from PLP Schiff bases 741
Hydrogen (H
2
), See also Hydrogenase
formation by bacteria 300
Hydrogenase 861, 878, 879
Hydrogen atoms, direct transfer of 768
Hydrogen bond(s) 47 49, 69. See also
Hydrogen bonding
chains in proteins 56, 64
in collagen 72
competition for 82
in hyaluronan 176
in ion pairs 75
lengths of 48, 493, 494
low-barrier 494
in polypeptides 56, 61
in proteins 75
strengths of 49, 494
strong, in papain 619
Hydrogen-bonded network
in carbonic anhydrase 678
in proteins 75
Hydrogen bonding 491 494. See also
Hydrogen bond
chemical shift, effect on 139
Hydrogen transfer reactions, table 766
Hydrogenase(s) 878, 879, 879s
iron-only 878
Ni containing 879
Hydrogenosomes 15
Hydrolases 589
definition of 498
Hydrolysis 593, 608
of biopolymers 116 118
of carbohydrates 189
of fats 507
Gibbs energies of, table 294
of proteins 116 118
Hydropathy index 78, 79
Hydrophobic effect 50
Hydrophobic interaction chromatography
103
Hydrophobic interactions 50, 51. See also
Hydrophobic bonding
Hydrophobicity 51
Hydroquinone(s) 815 822
Hydroxamate 838
Hydroxide ion, binding constants to metal
ions 311
Hydroxocobalamin 869
S-3-Hydroxy-3-methylglutaryl-CoA (HMG-
CoA) 700s
4-Hydroxy-l-proline 432s
erythro--Hydroxyaspartate
reaction with aminotransferase 750
Hydroxyl radicals 856
Hydroxylamine 251
reactivity of 590
Hydroxylapatite 103, 441s
Hydroxylysine 432s
5-Hydroxymethylcytosine 234, 810
Hydroxymethylfurfural 167s
3-Hydroxy-3-methylglutaryl-CoA 699, 701s
lyase 699
reductase 776
synthesis of 699
5-Hydroxymethyluracil 234
3-D-Hydroxymyristic acid 429
4-Hydroxynonenal 682
Hydroxyproline 52s, 72
2()-Hydroxypyridine 491s
as catalyst 490, 491
tautomerism 45
Hydroxypyridine crosslink in collagen 434
D-10-Hydroxystearate from hydration of oleic
acid 688
Hyperfine structure of epr signal 398
Hyperpolarizability 70
Hyphae 20
Hypochromic effect 209, 255
Hypoxanthine
tautomerization of 203
I
ICAM-1
diagram 408
Ice
hydrogen bonds in 49
melting of 284
reaction rates in 492
structure of 49
Ice-nucleation proteins 191
Ichthyoses 439
Icosadeltahedra 346
Icosahedral viruses 345
Icosahedron 344
Idose (Ido) 163s
Iduronate, iduronic acid 176
conformation 166
Image reconstruction 130
Imidazole
binding constants to metal ions 311
pK
a
value of 99, 487
Imidazole group(s) 46s, 53
hydrogen bonding 48
numbering of ring atoms 46
as a polarizable base 590
in proteins 312
protonation of 46
tautomerism of 46
Imidazolium cation 46, 46s
Imine(s), 679. See also Schiff bases
in enzymes 699, 740 746, 750
Immunoglobulin(s) 58, 74
repeat 367
superfamily 187
Immunosuppresant 488, 805
Indole, resonance energy 299
Indole-3-acetate 734
Induced fit 481
model 351
theory 349
Inducer(s) 539
of gene expression 539
Inducible enzyme 538
Induction of enzyme synthesis 538, 539
Inflammation 384
Inhibition
classical noncompetitive 474
of enzyme synthesis 536
of enzymes 471 478
noncompetitive 473 475
uncompetitive 475
Inhibition constant 472
Inhibitors
competitive 471
enzyme-activated 478, 864
in study of enzyme mechanisms 475
transition state 484
Initiation codon 236
Inorganic deposits by cells 15
Inosine 203
Inosinic acid 203
myo-Inositol 387s
Inositol 1,3,4,5-tetrakisphosphate (InsP
4
) 564
Inositol 1,3,4,5,6-pentakisphosphate (InsP
5
)
564
Inositol 1,4,5-trisphosphate (InsP
3
) 563
receptors 564
Inositol hexakisphosphate (InsP
6
) 564
Inositol phosphates, scheme 565
Inositol polyphosphates
921 Volume 1 Index
in signaling 563 566
Inositol trisphosphate 542
Insecta 24
Insecticides 636
Instantaneous velocity 456
Insulin 346s, 544, 567 571
effects on acetyl-CoA carboxylase 568
effects on ATP citrate-lyase 568
effects on GLUT4 568
effects on ribosomal subunit S6 568
glucose uptake 570
half-life 571
hexameric, 347s
metabolic effects of 568
phosphorylation 571
response element 578
second messenger 569 571
Insulin receptor 568, 569
autophosphorylation 569
down-regulation 571
mutant forms 569
protein kinase of 544
structure (schematic) 569
substrates 559
substrate-1 (IRS-1) 570
tyrosine kinase domain 569, 570s
Integral proteins of membrane 391, 401, 402
Integrated rate equations 457 461
Integrin(s) 187, 405, 406
Interaction constant 352
Intercalation in DNA 222, 223
Interleukin-1-converting enzyme 619
Internal energy 282
Internal equilibrium constants 640
International system of units 2
International unit of enzyme activity 457
Intervening sequences. See Intron
Intestinal mucosa
alkaline phosphatase in 645
Intrinsic barrier 493, 691
Intrinsic factor in vitamin B
12
uptake 869
pernicious anemia 869
Introns 202
Inulin 170, 175
Inversion of configuration
as criterion of mechanism 590, 591
by glycosyltransferases 593
Involucrin 439
Iodacetamide 115
Iodide-starch complex 174s
Iodoacetate 115
Iodothyronine deiodinase 824
Ion(s)
hydration of 50
transport of 420 425
electrogenic 421
Ion channels 412 414, 420 425
Ion exchange chromatography 103, 104
Ion exchange resins 104
Ion pairs 55
hydrogen bonded 75
in proteins 75
Ion pumps 422 425
Ion
Ionic atmosphere of membranes 400
Ionic radius
effect on metal binding 311
for ions, table 310
Ionic strength 288, 297
Ionophores 414
Iron 837 866
binding of O
2
850
proximal histidine in heme 851
in hemoglobin 850
storage of 841 843
uptake by cells 837 841
non-transferrin pathway 840, 841
transferrin pathway 840, 841
Iron regulatory factor 688
Iron regulatory protein 1 862
Iron-sulfur cluster(s) 514, 857, 857s
3Fe-4S 859
4Fe-4S 857
reduction of 859
in flavoproteins 861
membrane proton pumps 860
Mossbauer X-ray absorption spectra
860
oxidation states 857
properties of 859, 860
radical generator 862
reduction and oxidation 862
single-electron transfer 861
Iron sulfur enzymes
functions of 860 862
Iron sulfur proteins 686688, 857 862
electron paramagnetic resonance
spectrum 860
Iron superoxide dismutase 866s
Iso-mechanisms 466
L-Isoaspartyl / D-aspartyl methyltransferase
594
Isoaspartyl group 84
Isobutyraldehyde, tautomerism 45
Isobutyryl-CoA mutase 872
Isocitrate 516s, 686s, 704s
oxidation to 2-oxoglutarate 705
Isocitrate dehydrogenase
phosphorylation of 545
stereochemistry 768
Isocitrate lyase 704
Isocoumarins 623
Isodesmosine 436, 436s
Isoelectric focusing 108, 111
Isoelectric point 101, 106
of proteins 83
Isoenzymes (isozymes) 536, 538
Isoionic point 106
Isolation of compounds 98 108
Isoleucine (Ile, I) 52s, 539
biosynthesis 540
branched fatty acids from 381
configuration 43
Isologous interactions 337 353
in oligomers 349 353
square 352s
Isomerases 498, 530, 692 697
aldose-ketose interconversions 692
cis-trans, glutathione-dependent 690
hydride shift mechanism 695
3-oxosteroid 696, 697
ring opening by 693
triose phosphate 693 697
xylose 695
Isomerization reactions 526, 527, 530, 690,
712
of isopentenyl diphosphate 712
stereochemistry 874
vitamin B
12
-dependent 873
table 871
Isomers 41 43
Isomorphous replacement method 133
Isonicotinyl hydrazide 738
Isopentenyl diphosphate (prenyl pyrophos-
phate) 527, 689, 690s, 712s
Isopentenyladenosine 234s
Isopeptidases 525
Isopeptide linkage
in proteins 80
to ubiquitin 524
Isoprenoid alcohol phosphokinase 409
Isoprenoid compounds. See also Polyprenyl
compounds
biosynthesis 689, 700
Isoprenoid lipids 389
Isoprenoid quinones, structures of 819
Isoprenylation. See Prenylation
Isopropylmalate 700s
Isopropylmalate isomerase 689
Isoproterenol 553, 553s
Isospartyl residues in proteins 594
Isotachophoresis 108
Isotope(s)
properties of 110
radioactive 110
stable 110
Isotope effect(s)
for alcohol dehydrogenases 771
in displacement reactions
kinetic 111, 486, 592, 593
Isotope exchange at equilibrium 467, 468
Isotopic labels (tracers) 110, 679
Isotopomer(s) 111
Isotopomer analysis 111
J
Joule 282
definition of 2, 283
Jun 70, 241, 572
definition of 577
jun 572, 576, 579
K
K
+
channel 413s
Karplus equation 139
Katal 457
KDEL receptor 554
KDO (ketodeoxyoctonate) 180s
Kelps 22
Kendrew, John C. 84
Keratan(s)
Keratan sulfate 175 177, 176s
disulfide bridges 115
N-linked 185
terminal units 181
Keratin 70, 369
Keratinocytes 439
Keshan disease 822
Kethoxal 253
Ketimine 121, 744s. See also Schiff base
from pyridoxal phosphate 742
as electron acceptor 746, 747
-Ketoacid. See 2-Oxoacid
Ketoamine 434s
Ketodeoxyoctonate. See KDO
Ketone(s), acidity of 46
Khorana, H. Gobind 84
Kidney cells, alkaline phosphatase in 645
922 Volume 1 Index
Kidney tubules 28
Kilocalorie 282
Kinases 638, 654 657
cyclin-dependent 580
metal ions in 654
Kinesin 370
Kinetic(s)
high enzyme concentrations 461
of enzyme action 455 497
of rapid reactions 468, 469
Kinetic constants 455
Kinetic equations for complex mechanisms
466, 467
Kinetic isotope effect (KIE) 76, 111, 486, 592,
593, 600, 789
Kinetic parameters 459
Kinetic theory of gases 462, 463
Kinetisome 15
DNA of 15
Klug, Aaron 84
Knobs-into-holes bonding 71, 334
in coiled coil 71
Knots
in peptide chains 74
in DNA 219
Kojibiose 431
Kornberg, Arthur 84
Koshland
induced fit theory 349
model 352
Krebs, Hans A. 83
Kringles 367, 368s
K systems 475
Kuru 248
Kynurenine 747s
conversion to alanine 746
L
Lac repressor 240
Laccase 887
Lactacystin 620, 620s
Lactate 506s
binding constants to metal ions 311
Lactate dehydrogenase 342, 467, 768, 771
active site of 769
inhibition by pyruvate 780
isoenzymes of 538
Lactate monooxygenase 799
Lactic acid 508, 510
pK
a
value of 293
Lactobacillic acid 381s
Lactoferrin 840, 840s
Lactoperoxidase 853, 856
Lactose (Lac) 167
-Lactose 168s
sweetness of 168
-Lactose 168
Lactose (Lac) permease 417
Lactosyl ceramide sulfate 389s
O-Lactyl-GlcNAc. See Muramic acid
Lactylthiamin 733s
Lag phase of growth curve 470
Lamellar (smectic) phase 392
Laminin 71, 409, 437
Lamins 369
Landsteiner, blood groups 184
Langmuir trough 394
Langmuir-Adam film balance 394
Langmuir-Blodgett layers 392, 394
Large calorie, numerical value of 283
Lateral phase separation 395
Lathyrism 438
Lavoisier 281
Lead 31
Leader peptidase 620
Leadzyme 649
Leaving group
definition of 589
effect on reaction rates 590
Lecithin. See Phosphatidylcholine
Lectin(s) 64s, 186, 186 188
domains in proteins 186
Lederberg, Joshua 83
Leghemoglobin 362
Legionnaires disease 7
Leprosy 7
Leucine (Leu, L) 52s
branched fatty acids from 381
biosynthesis of 527, 712
L-Leucine 2,3-aminomutase 875
Leucine aminopeptidase 627
Leucine-zipper 69, 71, 241, 576
Leucopterin 803, 804s
Leucovorin 810
Leukemias 573
Leukocytes 26
adhesion 188
Leupeptin 615, 622, 622s
Lewis acid, Zn
2+
680
Lewis blood group 184
Lewis x antigen 188
Lichen(s) 23
Life, origin 9
Ligase(s) 498
definition of 498
Light absorption 122, 123. See also Absorp-
tion spectra
of DNA 209
Lily, genome 12
Lineweaver-Burk plot 460
Link protein 181
Linkage between genes 18
Linoleic acid 381
Lipase(s) 634 637
hormone-sensitive 635
lipoprotein 635
Lipid(s) 379 436
bilayer 391s, 394
melting temperature 395
building blocks of 380 389
caloric value 283
content in tissues 31
in membranes 392
physical properties of 394
neutral 382
Lipid A 429, 430s
Lipmann, Fritz A. 83, 720
Lipoamide dehydrogenase
reaction mechanism, scheme 792
Lipoarabinomannans 431
Lipocortins See Annexin(s)
Lipoic acid 795 802
absorption maximum 795
arsenite, reaction with 596
chemical reactions 795
functions of 796 798, 798
proteins, attachment to 795s
reduction of 785
ring strain in 795
Lipopolysaccharide 429
O-antigen of 180
structures of 430
Lipoprotein(s) 58
bacterial 428
Liposomes 392, 392 394
NMR spectra 396
Liquid crystals 392 394
Liquid crystalline phases 392
Lithium salts, in treatment of manic-
depressive illness 564
Lithostatine 443
Liverworts 29
Lobry de Bruyn-Alberda van Ekenstein
transformation 693
Lock and key theory 478
Log phase of growth 470
Lon protease 628
Loricin 439
Lou Gehrigs disease 884
Low barrier hydrogen bond 614
Lowry method 102
Luciferase 795
Lungfish, genome 12
Luteinizing hormone releasing factor (LRF)
54s
Lutropin receptor 554
Lyases 530
definition of 498
Lymphocytes 25, 26
Lymphomas 573
Lynen, Feodor 84, 725
Lyotropic series. See Hofmeister series
Lysine (Lys, K) 700, 745
biosynthesis 53s , 540
pK
a
value of 487
D-Lysine 5,6-aminomutase 874
D--Lysine mutase 871, 874
L--Lysine mutase 871, 874
Lysine tyrosylquinone (LTQ) 817, 817s
Lysophosphatidylcholine 384
Lysosome 10, 13, 98
Lysozyme(s) 599 602, 599s, 603
active site structure 600
catalytic side chain groups 599
kinetic isotope effect 600, 601
neighboring group assistance 601, 602
substrate distortion 601
transition state 601
HSQC NMR spectrum 146
titration curve 98
Lysyl oxidase 434, 886
-Lytic protease 610
Lyxose (Lyx) 163s
M
M-phase of cell cycle 579
Macrodipole of helix 70
Macroglobulin(s) (IgM) 58, 629

2
630
Macromolecular structures
self-assembly pathways 362 368
Macromolecules. See also Polymers
association of 325
binding by 327
packing of 332 348
synthesis and turnover of 517 527
Macrophages 26
Macroscopic dissociation constants 305 307
923 Volume 1 Index
Magnesium complex
in kinase action 655
Magnesium ion(s) 31
in adenylate system 302 304
concentrations in tissues 299
effect on equilibria 296
enzymes dependent upon 299
intracellular concentration 299
Magnetic iron oxide 842
Magnetic resonance study
of phosphorus atom displacement
639 642
Magnetite in organisms 842
Maize, genome 12
Major groove of DNA 201, 213
Malaria 19
L-Malate, 516s, 683s, 705s See also Malic acid
cleavage to acetyl-CoA 703
Malate dehydrogenase 497, 768
Malathion 636
Maleylpyruvate 690s
Malic enzyme 705
Malolactic enzyme 777
Malonate decarboxylase 723
Maltoporin (LamB) 411
Maltose 81, 167s, 606
-Maltose from -amylase action 608
Mandelate dehydrogenase 784
Mandelate racemase 691
active site 690
Manganese 887, 888
Manganese ion(s)
effect on equilibria 297
in enolase 685
Mannan(s) 170, 175
Mannitol 165, 191
Mannoprotein of yeast cell walls 185
Mannose (Man) 161s, 163s
Mannose-6-phosphate 185
Mannose-6-phosphate isomerase 693
MAP kinase cascade 576 578
figure 579
in insulin signaling 570
MAPK 578
definition of 577
MAPKK, definition of 577
Marcus theory 771, 848
of proton transfer 493
Marfans syndrome 440
Markovnikov rule 777
Mass action ratio 535
Mass spectrometry
electrospray 114
MALDI 112 114
in sequencing 119
Mass spectrum
of Cu/Zn superoxide dismutase 114
of myoglobin, apo 115
Mast cells 427
Mastigophora 19
Matrix of mitochondria 14
Matrix metalloproteases 627
McClintock, Barbara 84
Mechanocytes 25
Medusa form 23
Megakaryocytes 26
Meiosis 18
MEK 578, 579
Melanin 439, 887
Melanocyte(s) 439
drawing 439
Melanophores 439
Melanosomes 439
Mellitin 414
Melting curve
for DNA 256
for a diacylglycerol 395
Membrane(s) 5, 390 410
active transport 410
anchors. See Anchors, membrane
artificial 392 394, 393
basement 10
cation channels 421
chemical compositions, table 393
coated, 426s
communication 427
concentration gradient 206
Davsen-Danielli model 393
differential scanning calorimetry 394
double, of gram-negative bacteria 5
electrical properties 400
fluid mosaic model 390
fluidity 397
freeze fractured 405
inner side composition 401
ionic atmosphere 400
ionic gradients across 400
lateral diffusion of lipids 399
lateral phase separation 395
lateral proton conduction 401
lipid bilayer structure 390, 393
lipid composition 392
lipids, physical properties of 394
NMR spectra 396
outer side composition 401
peripheral proteins 407
permeability to neutral molecules 399
pretransition temperature 394
resting potential 400
structure of 390 401
transport through 410 427
transporters 411 427
Membrane metabolism
enzymes of 409
Membrane proteins 401 409
anchors 402
glycoproteins 404
integral 401
positive-inside rule 401
Menadione 818
Menaquinones (vitamin K
2
) 818, 821
Mengo virus 247
Menkes disease 883
Mercaptans 624
Mercaptides 550
Mercaptoethanol 98
-Mercaptoethylamine in coenzyme A 722s
7-Mercaptoheptanoylthreonine phosphate
814s
Mercapturic acid 550s
Mercuric reductase 787
Mercury 317
methylation of 876
Mercury ions
binding to proteins 125
reduction of
Merrifield procedure, peptide synthesis 127,
128
Merrifield, R. Bruce 84
MES buffer 99
pK
a
value of 99
Mesosome 3
Mesozoa 23
Metabolic diseases. See Genetic diseases
Metabolic pathways 505, 505 507
scheme 506
Metabolic regulation 535 581
control elements of 536
sensitivity coefficient 537
Metabolism. See also Specific compounds
activation 507, 508
beta oxidation 511, 512
control by covalent modification
reactions
table 543
control by substrate cycles 567
definition of 1, 2
effects of insulin 568
fermentation 508 511
glycolysis 508 511
introduction 505 527
oxidative phosphorylation 512 515
regulation of. See Metabolic regulation
study with isotopic tracers 110
Metabolite, activation 507, 508
Metal-binding proteins, chelate effect 310
Metal-binding sites 312
Metal complexes
formation constants, table 311
stability of, transition metal 311
Metal ion(s) 296
binding of 307
charge 311
geometry of orbitals 311
hydration of 311
ionic radii 311
table 310
toxicity 33
Metalloenzymes 680
Metalloflavoproteins 794
Metalloproteases 625 627
Metalloproteins 312
Metallothioneins 317, 317s, 680
Metals, transition 837 895
Metaphosphate ion 639
Metastasis 573
Metazoa 23 29
Methane formation 814
Methanofuran 814s
Methanogens 7, 813 815, 879 881
Methanol dehydrogenase 816s
Methanopterin 803, 804s
Methemerythrin 862
Methemoglobin 358
Methemoglobin reductase system 359
Methionine (Met, M) 53s, 539, 875
biosynthesis of 540, 746
sulfoxides 85
Methionine repressor 243
operator complex 243s
Methionine sulfoxide 631s
Methionine synthase 813, 874s, 875
Method(s) 95 150
isolating compounds 98 108
of peptide synthesis 127 129
of carbohydrate analysis 188 192
Methotrexate 574, 805
5-Methylarginine 881
3-Methylaspartate lyase 685
Methylation
of arsenic 876
of carbohydrates 189, 190
of carboxyl groups 592
of mercury 876
of nucleic acids 591
permethylation 189
924 Volume 1 Index
of protein side chains 79
in regulation 543
of uracil in tRNA 591
Methylcellulose 172
Methylcobalamin 867, 867s, 875
Methyl-coenzyme M reductase 879 881
Methyl corrinoids 876
-Methylcrotonyl-CoA, carboxylation of 724
2-Methylcysteine 881
5-Methylcytosine 234
oxo-Methylene analog of transition state 626,
627s
-Methyleneglutarate mutase 871
Methylene-THF reductase 813
Methylesterase 563
N-Methylglutamate synthase 789
2-Methylglutamine 881
Methylglyoxal 695, 695s
metabolism of 697
Methylglyoxal synthase 697
Methyl group(s) 802
synthesis of 813
transfer reactions of 875
N-Methylhistidine 881
4-Methylidine-imidazole-5-one (MIO) 757
S-Methylmalonyl-CoA
formation from propionyl-CoA 725
equilibrium constant 725
Methylmalonyl-CoA mutase 871
mechanism 873
stereochemistry 874
three-dimensional structure 874
Methylotrophs 815
Methyltetrahydrofolic acid 875
Methyltransferases 526, 563, 591 593
Mevalonic acid-5-diphosphate (pyrophos-
phate) 689, 690s
Micelles 390
Michael addition 681
Michaelis complex 751, 751s, 752
Michaelis constant 464
definition of 459
Michaelis-Menten equation 459
Michaelis pH functions 96, 296
Michel, Hartmut 84
Microbodies. See Peroxisomes
Microfilaments 369, 369, 370
behind desmosomes 29
Micropinocytosis 426
Microscopic binding constants 328, 329
Microscopic dissociation constants 305
of pyridoxine 305
Microscopic reversibility, principle of 486
Microscopy
atomic force 131
chemical force 131
confocal scanning optical microscopy
129
electron 130
near-field scanning optical 130
negative contrast 130
scanning tunneling 131
STM image, glycogen 174
Microsome, definition of 14
Microtomes 130
Microtubules 15, 369, 370
associated proteins 372
growth of 373
labile 334, 370
organizing centers 372
stable 370
Microvilli 10, 12
Miescher, Friedrich 199
Millipedes 24
Minor groove of DNA 201, 213
Mitchell, Peter 84
Mites 24
Mitochondria 14, 15
inner membrane 512
matrix of 512
definition of 14
origin from bacteria 15
of plant cell
in micrograph 13
Mitochondrial DNA. See DNA, mitochon-
drial
Mitochondrial processing peptidase 625
Mitochondrion 10
Mitogen activated protein kinase (MAP
kinase) cascade 576 578
figure 579
Mitogenic activity 576
Mitomycin 225s
Mitosis 18, 371
Mobile defects in proteins 496
Modification of enzyme
by group transfer 545 549
by phosphorylation 541 544
Modifiers of enzymes 473
Molal solution, definition of 287
Molecular activity (of enzyme) 457
Molecular biology, definition of 2
Molecular dynamics simulations of mem-
branes 396
Molecular mass
in daltons 4
relative 4
Molecular size, as basis of separations 100
Molecular weight 4 See also Relative
molecular mass
Molecules, properties of
chiral 42
conformations of 43 45
cyclic symmetry 332, 333
packing of 41
structural principles 39 46
Molluscs 24
Molybdenum 890 893
coenzyme forms 890 892
nutritional need 893
oxo-transfer reactions 892
Molybdenum-iron cofactor (FeMo-co) 892
Molybdoenzymes
active site structures 892
dimethylsulfoxide reductase family
890
nitrogenase family 890
sulfite oxidase family 890
xanthine oxidase family 890
Molybdopterin 803, 804s, 825
in formate dehydrogenases 892
Molybdopterin cytosine dinucleotide 891s
Monensin 414
K
+
complex 415s
synthesis 872
Monocytes 26, 188
Monod, Jacques 84
Monod-Wyman-Changeux (MWC) model
350, 476
allosteric regulation 349
Monosaccharide(s) 161 167
characteristic reactions 167
13
C-NMR spectrum of 162
reduction of 167
Morels 20
Morgan, Thomas H. 83
Mssbauer X-ray absorption spectra
of iron-sulfur clusters 860
Mosses 29
Motion, in proteins 81
Mouse, genome 12
mRNA 5, 230, 257, 536
splicing of 11
Mucins 181, 182
cis,cis-Muconate lactonizing enzyme 692
Mucopolysaccharidoses 659
Multicatalytic protease 339, 620. See also
Proteasome
Multidrug resistance protein 417
Multilamellar vesicles (liposomes) 392
Multiple attack concept 606
Multisubstrate enzymes, kinetics of 464
Muramic acid (Mur) 165s
Murein 170, 428, 429s. See also Peptidoglycan
Musci 29
Muscle(s)
cardiac 26
smooth 26
striated 26
thin filaments 336
electrolyte concentrations 206
Muscular dystrophy 822
nutritional 822
Mutagens 222, 253, 254
Mutarotase 486, 526
Mutarotation 486
Mutases 637
vitamin B
12
dependent 653, 654, 872
Mutation(s) 2
addition 256
autosomal recessive 16
conservative 59
deletion 256
harmful dominant 16
heteroduplex formation 256
lethal 16
rate of 16
myb 576
myc 572, 576, 577, 579
Mycelium 20
Mycobacteria 431
Mycobacterium tuberculosis 431
Mycolic acids 431
Mycoplasma(s) 2, 3
Mycoplasma genitalium 4
genome 12
Mycorrhizae 20
Myelin 390, 399
Myeloperoxidase 856
Myoglobin 17, 68s, 70, 353 359, 354s, 844
bonding of dioxygen 851
folding pattern 354
mass spectrum 113
mass spectrum of apo form 115
NMR spectrum 148
oxygenation curves 355
packing of side chains 75
Myohemerythrin 71s
Myosin 369
coiled coil structure of 71
diffusion constant of 461
Myotonia congenita 420
Myriopoda 24
N-Myristoyl group 559
Myxococcus xanthis, genome 12
Myxomycetes 20
925 Volume 1 Index
N
NAADP
+
315, 779
Na
+
, K
+
-ATPase 423 425
model 424
Nacrein 678
NAD
+
(NAD) 63s, 507s, 535, 765 771, 767s,
778
absorption spectrum of 768
addition to 779
containing
2
H 768
determination of 779
NAD
2
H 771
oxidation by 298
NAD
+
/ NADH ratio, redox potential 767
NADH (NADH + H
+
; NADH
2
) 765 771
absorption spectrum of 768
modification in acid 780
NADH-X 780
NADH-methemoglobin reductase 826
NADH:ubiquinone oxidoreductase 788
oxidation by ferricyanide 780
NADP
+
(NADP) 507, 765 771, 767s, 779
in catalase 853
isolation of 767
NADPH (NADPH + H
+
; NADPH
2
) 507, 765
771
NADPH/ NADP
+
ratio 776
Naphthalene, resonance energy 299
Nathans, Daniel 84
Natural killer cells 185
Near attack conformation 485
Near-field scanning microscope 130
Neighboring group assistance in displace-
ment reactions 601, 602
Nekoflavin 783
Nematocyst 427
Nematodes 24, 25
Neocarzinostatin 218, 224s
Neopterin 803
Nephrogenic diabetes insipidus 412
Neprilysin 625
Nernst equation 400
Netropsin 225s
binding to DNA 225
Neu (protein) 576
neu (oncogene) 572
Neuraminic acid anion (Neu) 165s
Neuraminidase 186
Neurofibromatosis 574
Neurofilaments 369
Neuroglia 25
Neuron(s) 25. See also Axon
of squid 24
Neurospora crassa 20
figure 21
Neurotransmitter(s) 29, 553
receptors for 479
Neutron diffraction 137
Neutrophils 26, 188
Nick translation 257
Nickel 877 882
dietary essential 877
in plants 877
Nickeloplasmin 877
Nicotinamide 507, 721, 766, 767, 769s, 780.
See also NAD
+
, NADH; NADP
+
, NADPH
daily requirements 756, 769
Nicotinamide adenine dinucleotide. See
NAD
+
, NADH
Nicotinamide ring 770, 779
Nicotine 769
oxidation to nicotinic acid 769
Nicotinic acid. 769s See also Nicotinamide
Nicotinic acid adenine dinucleotide
(NAADP
+
) 778
Nicotinic acid adenine dinucleotide
phosphate (NAA 315
Nicotinic acid hydroxylase 825
Nidogen 437
Ninhydrin 120, 121s
Nitrate reductase(s)
assimilatory 890
dissimilatory 890
Nitric oxide, NO 553, 561
binding to hemoglobin 358, 359
Nitrite reductase(s) 836s, 861
copper in 884, 885
cytochrome cd
1
type 885
Nitroform 126
Nitrogen mustards 574
Nitrogenase 890
Nitromethane, inhibition of ammonia-lyases
756
p-Nitrophenolate anion, as a leaving group
636
p-Nitrophenyl acetate 610, 610s
Nitrous acid, reaction with nucleic acids 253
Nitrous oxide reductase(s)
copper in 884, 885
NMR spectra
of ADP,
31
P 642
of aspartate aminotransferase 149
of carbohydrates 190 192
of honey,
13
C 162
of tRNA,
19
F 270
of glycyl-L-histidylglycine 147
of gonadatropin-releasing hormone
145
of DNA,
1
H 269
of lysozyme 146
of myoglobin 147
of muscle,
31
P 304
of phospholipid phases,
31
P 397
of phosphatidylcholine,
1
H 396
of pyridoxal 5'-phosphate,
1
H 138
pyruvate kinase equilibria 641
of serine proteases,
1
H 613
of tRNA 268
NMR spectroscopy 137 149, 494
assignment of resonances 145
band widths 138
basic principles 137
carbon-13 140
chemical shift 138
table 141
correlation time,
c
142
coupling constants 138, 139, 146
deshielded 139
deuterium 140
distance constraints 145
downfield 139
DSS or Tiers salt 138
exchangable protons 148
fluorine-19 140
Fourier transform 141
free induction decay 141
Heisenberg uncertainty principle 138
HMQC (15N-multiple quantum
coherence) 144
HSQC (15N-multiple quantum
coherence)
lysozyme 146
isotope-edited spectra 144, 145
J coupling 139
liposomes 396
membranes 396
multidimensional
mixing time 143
NOESY 142
nitrogen-15 140
NOE 140
NOESY spectra 146
nuclear Overhauser effect 140
phosphorus-31 140, 295
of adenylate system 303
of muscle 304
of phosphotransferases 640 643
positional isotope exchange 641
protein dynamics 145
relaxation times 141
residence time,
m
142
ring current shifts 139
saturation-transfer 641
scalar coupling 139
solid-state 149
spin decoupling 139
spin-lattice relaxation time, T
1
142
spin-spin interaction 139
spin-spin relaxation time T
2
142
stable isotopes, use of 110, 111
study of phosphotransferases 639
tetramethylsilane (TMS) 138
titrations 147
transferred NOE 145
two-dimensional 141
correlation spectroscopy (COSY)
144
TOCSY 144
of various nuclei 141
of vicinal protons 139
Nobel prizes, list 83
Nodes of Ranvier 29
Nojirimycin tetrazole 605, 605s
Non-coding (transcribing) strand in DNA
237
Noncompetitive inhibition 476, 477
Nonheme iron proteins. See Iron-sulfur and
diiron proteins
Nonlinear equations 460
Nonmetallic ions, ionic radii, table 310
Nonproductive complexes 475
Norepinephrine (noradrenaline) 553, 553s
in receptor 555s
Nuclear envelope 11
Nuclear magnetic resonance spectroscopy.
See NMR spectroscopy
Nuclear Overhauser effect (NOE) 140. See
also NMR spectroscopy
Nuclear pore 11
Nuclear relaxation
by chromium ions 639
by manganese ion 639
Nucleases 250
micrococcal, active site 653
Nucleic acid(s) 199 213, 218
abbreviated structures 204
absorption of light 205
acid base chemistry 204, 205
amount in cells 31
bases, aromatic character of 204
stacking of 209
catenated 218
cruciform structure in 229
926 Volume 1 Index
dynamics 218
electrophilic reagents 253 255
homology of 256
hydrolysis of 249
interactions with proteins 239 244
isolation of 249
knots in 218, 219
melting curves of 255
methods of study 249 270
pK
a
values of 204
separation of 249
tautomerism 204, 205
topology 218 230
turnover of 527
Nucleic acid bases
alkylation 253
aromatic character 207
chemical reactions of 207
epoxides 254
halogenation 253
reactions 251 255
reactions with formaldehyde 253
reactions with kethoxal 253
reactions with nitrous acid 253
Nucleoid 4
Nucleolus 10, 11
Nucleophilic catalysis 494. See also Covalent
catalysis
Nucleophilic character and bonding to metal
ions 307
Nucleophilic displacement reactions 526,
589 662
carbocationic intermediates 598
cellulases 602
chitinase 599 602
glycogen phosphorylase 604, 605
-glycosidases 602
glycosyltransferases 593 598
inversion 590, 591
kinetic isotope effects 592, 593
lysozymes 599 602
starch-hydrolyzing enzymes 605 608
transmethylation 591, 592
Nucleophilic group, definition of 589
Nucleophilic reactivity (nucleophilicity) 589
alpha effect 590
effect of polarizability on 590
of sulfur compounds 590
Nucleoside(s)
conformations of 211, 212
names and abbreviations, table 203
torsion angles of 211
Nucleoside diphosphate kinase 655
Nucleoside Q (Queosine) 234s
Nucleoside trialcohols, fluorographs 252
Nucleoside triphosphates 302
Nucleoside Y 234s
Nucleosomes 239
DNA in 221
model 222
Nucleotide(s) 199 213, 200s
names and abbreviations, table 203
chemistry of 203 213
conformations of 211 213
modified 234
pairs 3
structures of 203 213
Nucleotide binding domain 76
glyceraldehyde phosphate
dehydrogenase 63s
Nucleotide handles
of coenzymes 719
and specificity of metabolism 720
Nucleotide kinases 655
Nucleotide sequence
of aspartate aminotransferse 202
of DNA and RNA chains 200
of genes and genomes 12, 201
of mitochondrial DNA 201
of viruses 201
Nucleotidyl transferases 657
Nucleotidylation 545
Nucleus 10, 11
of plant cell, micrograph 13
O
O-Antigen 180, 428 430
Obelia 24
Occlusion zones 29
Octahedral geometry 311
Octopus, intelligence of 24
Oils, composition of 380
Okadaic acid 545
Old yellow enzyme 783
Oleate hydratase 526, 688
Oleic acid 381
Olfactory responses 558
Oligomer(s)
asymmetry 344 347
cubic symmetry 342 344
with dyad axes 337 348
heterologous tetramer 352
isologous tetramer 352
quasi-equivalence 344 347
Oligomeric enzymes 475
Oligonucleotides, 200 205
synthesis of 258
probe 119
Oligosaccharide(s) 167 170. See also Specific
names
asparagine-linked (N-linked) 180 185
biological recognition of 161
N-linked 182 185, 183, 192s
release from glycoproteins 188
separation of 100
Omega. See Oxidation
OmpA protein 411, 428
OmpC (osmoporin) 411
OmpF 411, 412s
Oncogenes 242, 544, 571 576
in human tumors 571
table 572
Oncogenic viruses 248
One-start helix 334
Oparin, I. V. 9
Open systems 289
Operator sequence of DNA
1
H NMR spectrum 269
Operons 240
Opsin(s) 553
Optical rotation 42
Optimum rate for enzymes 469
d Orbitals, participation in covalent bond
formation 311
Orcinol 251
Ordered binding 464
Ordered sequential mechanism 475
Organelle(s) 11
separation 101
Organic mechanisms, use of arrows 528
Ornithine decarboxylase 342, 342s
Ornithine mutases 871, 874
Oscillatoria 22
Osmate ester 393s
Osmotic shock 417
Osteoarthritis 438
Osteoblasts 26, 441
alkaline phosphatase in 645
Osteocalcin 821
Osteoclasts 441
acid phosphatase in 645
Osteocytes 441
alkaline phosphatase in 645
Osteogenesis imperfecta 438
Osteonectin 313
Osteopetrosis 572, 678
Osteopontin 409, 441, 572
Osteoporosis 443, 619
Ouabain 422
Ovomucoids 183, 629
Ovotransferrin 840
Oxalate 699s
Oxaloacetate 699s
Oxaloacetate 506s, 515, 516s, 737s
acetylhydrolase 698
decarboxylases 527, 706
4-Oxalocrotonate decarboxylase 705
4-Oxalocrotonate tautomerase 697
Oxalosuccinate 516s, 705
Oxazoline 601s
Oxidation
of biochemical substances, table 290
linked 705
by NAD
+
, table 290
Oxidation pathway 506, 511
Oxidation-reduction, by dehydration 697
Oxidation states
of iron-sulfur clustsers 857
Oxidation-reduction potentials. See also
Reduction Potential
of pyridine nucleotide system 766
table 301
Oxidation-reduction reactions
coenzymes of 765 827
Oxidative decarboxylation of -oxoacids
511, 736, 796 802
by hydrogen peroxide 799
mechanisms 799 801
with thiamin diphosphate 800
Oxidative metabolism 535
Oxidative phosphorylation 512
Oxidizing power of redox couples 300
Oxidoreductases 497
(2)-Oxoacid(s) (-keto acids). See also
specific acids
cleavage of 802
decarboxylation 730
oxidative decarboxylation 796 802
-Oxoacid(s)
decarboxylation 705
(2)-Oxoacid dehydrogenases 348
sequence of reactions, diagram 798
3-Oxoacyl-CoA transferase 527
-Oxo alcohol, reaction with thiamin 732
Oxocarbenium ion 598s, 604
(2)-Oxoglutarate (-ketoglutarate) 516s,
737s
dehydrogenases 796
synthase 799
2-Oxoisovalerate 700s
Oxonium ion. See Oxocarbenium ion
2-Oxopantoate 810
927 Volume 1 Index
Oxophenarsine 597s
5-Oxoproline, 551s, 662s
Oxosteroid isomerase 526, 696
molecular activity 696

5
-3-Oxosteroid isomerase
turnover number of 458
Oxyanion 613
Oxyanion hole 614, 615s
formation of in trypsinogen 615
of serine proteases 614
Oxygen (O
2
)
binding of, to heme proteins 353 358,
850 852
to hemocyanin 885, 886
paramagnetism of 850
reactivity of 850
Oxygenases 863
diiron 863
Oxyhemoglobin 851
Oxyperoxidase 854
Oxytocin 54s
Oysters, glycogen content 31
P
P Loop 425, 648, 658
Packing
of macromolecules 332 348
of molecules 41
Pagets disease 443
Palindromes
in DNA 238
H- (mirror-repeats) 238
Pallidin 405
Palmitic acid 381
Palmitoyl group 559
Pancreatic trypsin inhibitor 629
Pantetheine 722
Pantetheine 4'-phosphate 722
in acyl carrier proteins 723
Pantoic acid 722, 723
in coenzyme A 722
Pantothenic acid 721, 722, 722s
nutritional requirement of 756
Papain 618
active site 618
Paper chromatography 103
Papovaviruses 244
Parabactin 838, 838s
Parallel structure 62
Paramagnetic ions, effects on NMR signal
640
Paramecium 17, 20
Paramylon 170, 174
Parathion 636s
Parathyroid hormone 314
Paratose 180s
Partition coefficient 410
Parvalbumin 312 316
Parvoviruses 244
Patterson map 135
Pauling, Linus C. 83, 84
Paxillin 406
p 53 cancer suppressor 574, 581
Pectate lyase 65, 686
Pectate lyase C 686, 686s
Pectin(s) 15, 170, 177
Pellagra 721, 769
L-Penicillamine 739
Penicillin 20
Penicillin acylase 620
Penicillopepsin 621
Pentose phosphate pathway 700
reductive 517
Pentoses 162
PEP. See Phosphoenolpyruvate
PEP carboxykinase. See Phosphoenolpyru-
vate carboxykinase
PEP carboxylase. See Phosphoenolpyruvate
carboxylase
PEP carboxytransphosphorylase 706
PEP mutase 711
Pepsin 456, 621
specificities 117
Pepsinogen 625
Pepstatin 623
Peptidase(s) 526, 609
leader 620
procollagen 433
signal 620
ripeptidyl 140
Peptide(s) 51 59
conformation of 60
knotted chains 74
torsion angles 60
Peptide deformylase 627
Peptide hormone(s). See also Chapter 30
amidation of 522
Peptide linkage 51, 55s
cis 56s
dimensions 55
resonance stabilization of 55
trans 56
Peptide mapping 118
Peptide structure
torsion angles of, table 61
Peptide synthesis 127 129
solid-phase 127, 128
Peptide unit
hydrogen bonds 56
partial double bond character 55
planarity of 55
properties of 55, 56
structure 55, 56
Peptidoglycan(s) 179, 428, 429s
Peptidyl transfer 234
Peptidyl transferases 637
Peptidyl-prolyl-(cis-trans) isomerases 488
Peptidylglycine -hydroxylating
monoxygenase 887
Performic acid 115
Perfusion chromatography 100, 101
Perinuclear space 11
Periodate oxidation
of carbohydrates 190
Periodental disease 442
Periplasm 427, 428
Permeability coefficient 410
for D-glucose 415
Permease 411
for lactose 417
periplasmic 417 419
Pernicious anemia
vitamin B
12
treatment 868, 869
Peroxidase(s) 844, 845, 852 855
ascorbate 852
bromo 856
catalytic cycles, scheme 855
chloro 855
compounds I, II 853 855
cytochrome c 852
glutathione 856
halo 855, 889
heme linkage 853
horseradish 852, 856
iron properties 853
lacto 853, 856
mammalian 853
mechanisms of catalysis 853 855
myelo 856
structure 853
thyroid 856
Peroxisomes (microbodies) 10, 14
Perutz, Max F. 84
PEST regions 527
pH 95 98
of cytoplasm 549
effects on enzymatic action 469
effects on equilibria 293 297, 305
effects on metal binding 310
Michaelis pH functions 296
within a cell 295
pH dependence
of chymotrypsin 616
PH (plekstrin homology) domain 102, 367,
566, 571
Phaeophyta 22
Phage head 363
Phage lambda
repressors 241
Phage tail 363
Phagocytosis 13
Phagosomes 426
Phantom atoms 42
Phase transition 284
Phase transitions
of lipid bilayer 394
Phenol(s)
acidity of 308
pK
a
value of 293, 309
resonance energy 299
Phenolsulfonic acid
pK
a
value of 99
Phenylacetic acid(s)
acidity of 308
pK
a
value of 309
Phenylaceturic acid 513s
Phenylalanine (Phe, F) 52, 52s
absorption spectrum of derivative 123
Phenylalanine ammonia-lyase 755
Phenylephrine 563s
Phenylisothiocyanate 118
Phenylketonuria 16
Phenylpyruvate 689
Phenylpyruvate tautomerase 692
Phenylthiohydantoin 118
Pheromones 382
Phloem 30
Phorbol esters 564
Phosducin 558
Phosphagen 303
Phosphatase(s) 515, 526, 637, 645 647
Phosphate anhydride 303
Phosphate, inorganic (P
i
)
in adenylate system 302, 304
Phosphatides 383, 384
structures of 383
Phosphatidic acid 383s
Phosphatidyglycerol 383s, 384
Phosphatidylcholine 383, 383s, 391s
in bilayer 391
packing 391
Phosphatidylethanolamine 383, 383s
928 Volume 1 Index
bilayer, AFM image of 392
Phosphatidylinositol
383s
Phosphatidylinositol (PtdIns) 563, 565, 566s
in signalling 563 566
Phosphatidylinositol 3-kinase 565
Phosphatidylinositol 4,5-bisphosphate 563
Phosphatidylserine 383s, 564
decarboxylation of 753
Phosphatidylserine decarboxylase 409, 755
3'-Phosphoadenosine 5'-phosphosulfate
(PAPS) 659
Phosphoadenylation
of protein side chains 79
Phosphodiesterase 556, 638
for cGMP 558
Phosphoenolpyruvate (PEP) 510, 510s, 685s
key metabolic intermediate 705, 706
as stabilized enol 697
Phosphoenolpyruvate carboxykinase 549
Phosphoenolpyruvate carboxykinase
(PEPCK) 706
Phosphoenolpyruvate carboxylase 527, 711
Phosphoenzyme 653, 703
Phosphofructokinase
in regulation 535
Phosphofructokinase-2 541
Phosphoglucomutase 653
6-Phosphogluconate ion 164s
6-Phosphogluconate dehydratase 697
6-Phosphogluconate dehydrogenase 770
3-Phosphoglyceraldehyde dehydrogenase
770
2-Phosphoglycerate 510s, 685s
dehydration 685
dehydration to PEP 705
3-Phosphoglycerate 506s, 707s
in chloroplasts 706
Phosphoglycerate kinase 654, 656
Phosphoglycerate mutase 653
Phospho (phosphoryl) group 292s
chirality of 644
as a handle 507
transfer from ATP 660
transfer of 292
transfer of, by kinases 654
O-Phosphohomoserine 746
Phosphoketolase 736
Phosphokinases 526
Phospholipases 635
A
1
566
A
2
566, 635
C 563, 566
D 566
Phospholipid(s) 379, 380, 382 387, 392. See
also Specific substances
bilayer diagram 391
head groups, functions of 396
inverted hexagonal phase 397
31P NMR 397
non-bilayer structures 397
Phosphomannomutase 654
Phosphomutases 526
Phosphonamidate 626s
as metalloprotease inhibitor 626
Phosphonate esters 626s
Phosphonates
synthesis of 711
Phosphono group 711
Phosphonoethylamine 385s
Phosphonolipids 385
4'-Phosphopantetheine 723
4' - Phosphopantothenylcysteine
decarboxylases 753, 755
Phosphophoryns 442
Phosphoporin (PhoE) 411, 412s
Phosphoric acid
in lipids 380
pK
a
value of 99
Phosphorolysis, 593
Phosphorothioate
phosphotransferases, study of 642
Phosphorus-32
properties of 110
Phosphorylase a 541
Phosphorylase kinase 541, 544
Phosphorylation 541
in bacteria 545
cascade reactions 542
oxidataive 303
photosynthetic 303, see also
Photosynthetic phosphorylation
of protein side chains 79
substrate level 775, 800
Phosphorylation, photosynthetic. See
Photosynthetic phosphorylation
Phosphorylation reactions 303
Phosphorylation state ratio
definition of 303
O-Phosphoserine 610s
Phosphoserine 545
Phosphothreonine 545
Phosphotransferase system
bacterial 419, 420
Phosphotransferases 637
in-line displacement 638
P31 NMR of 642, 643
pentacovalent intermediate 638
stereochemistry 642, 643
Phosphotriesterase 646
mechanism 646
Phosphotyrosine 545
Phosvitin 80
Photoautotrophic bacteria 8
Photofootprinting 266
Photorespiration 707
Photosynthesis 506, 517, 705
Photosynthetic bacteria 7
Photosynthetic phosphorylation 303, 517
Photosynthetic reaction centers 71
Phthaldialdehyde 120s
Phycoerythrin 22
Phycomycetes 20
Phylloquinones (vitamin K1) 818, 821
Phylogenetic approach
to RNA structure 232
Phylogenetic tree 7, 8
Physarum polycephalum 20
Phytanyl group 385
Phytate. See Inositol hexakisphospate (InsP
6
)
Phytochelins 318
Phytocyanin 883
Phytoglycolipids 388, 389s
Phytosphingosine 388, 388s
Picornaviruses 247
Pili 3, 6, 335, 336, 364. See also F pili
Pimeloyl-Coenzyme A
conversion to biotin 719, 723
Pinacol 712s
Pinacolone 712s
Pinacol-pinacolone rearrangement 712
Ping-pong mechanism 465, 475
rate equation for 466
succinyl-CoA-acetoacetate CoA
transferase 662
Pinocytosis 13
Piperazine, pK
a
value of 99
Piperidine, pK
a
value of 99
PIPES, pK
a
value of 99
Pitch
of a helix 68
of B-DNA 213
Pituitary hormones 54
pK
a
values 95 98. See also Specific compounds
of acids, table 293
of amino acid side chains 55
of buffers, table 99
of buried groups 55
of dicarboxylic acids, table 328
effect on separation 104
in hemoglobin 357
of hydrogen-bonded protons 494
in hemoglobin 357
succinic acid 330
of terminal COOH 55
of terminal NH3
+
55
thiamin 330
Plague 7
Planarians 23
Plankton 22
Plant cell, electron micrograph 13
Plant cells and tissues 29, 30
fiber 11
Plasma amine oxidase 886
Plasma antithrombin III 177
Plasma membrane 12, 379
Plasmalemma. See Plasma membrane
Plasmalogens 383s, 384
Plasmids 5, 248249
ColE1 248
drug resistance 248
Plasmin 634
Plasminogen 634
Plasmodesmata 10
in micrograph 13
Plasmodium 19, 20
Plastids 14, 15
Plastochromanols 818
Plastocyanin 65s, 883
Plastoquinone(s) 818, 818s
Platelet(s) 26
cyclic AMP in 557
Platelet activating factor 384, 384s, 385
Platelet-derived growth factor (PDGF) 544,
571
Platyhelminthes 23
Pleated sheet 62
structure 62
P loop 425, 648
consensus 559
of Ras 559
PLP. See Pyridoxal phosphate (PLP)
PLP-dependent reactions. See Pyridoxal
phosphate-dependent reactions
PNA, 227s
Poison(s). See also Insecticides, Antibiotics,
Inhibitors
cyanide 590
hydroxylamine 590
Polar membrane 5
Polar molecules 50
definition of 48
hydration of 50
Polarizability 590
929 Volume 1 Index
Polarization
of water 47
Poliovirus(es) 247
Poly(ADP-ribose) synthase 545
Polyadenylation site
in RNA 202
Polyamines 222
Polycarboxylate synthases, table 700
Polycarboxylic acid synthases 700 703
Polyethylene
helical structure of 45
Polyethyleneglycol (PEG) 102
Polyfluorethylene (Teflon)
helical structure of 45
Polyglutamates 803
Polyglycine II 72, 73s
Polyhistidine tag 106
Poly--hydroxybutyric acid 3
Polymerase chain reaction (PCR) 260
figure 261
Polymetaphosphate 3
Polymorphonuclear leukocytes 26
Polynucleotide(s) 199s, 226 228. See also
Nucleic acids, RNA, DNA
hybridization 255
hydrazine reactions 254
interactions with ions 218
melting of 255
probes 255
rotational movements of bases, figure
217
triple-helical 226
Polynucleotide kinase 258
Polynucleotide polymerases 638
Polyomavirus 247
Polypeptide(s) 56 59. See also Proteins
antibiotics 66
chemical synthesis 85
conformation of 59 61, 78
definition of 51
torsion angles 59 62
Polypeptides. See also Peptides
Polyphosphates 302, 303
Polyprenyl compounds
polymerization of 712
Polyproline 72, 73
Polyproteins 522, 628
Polysaccharide(s) 169 180
alternating, list of 170
of bacterial surfaces 179, 180
chains, conformation of 170
list of, table 170
main linkages, table 170
structureal features of, table 170
Polysialic acid 187
Polyubiquitin 524
Porifera. See Sponges
Porins 65, 402, 411, 428
FepA 839
FhaA 839
Porphin(s) 843
Porphobilinogen 843s
Porphyrin(s) 843
biosynthesis 742
chelate effect 310
tautomerism of 845
Posttranslational modification 402
of proteins 521 523
Potassium 206
Potassium ion(s) 31
activation of enzymes by 206
binding to tetraplex DNA 206
concentration in cells 206
effect on equilibria 297
replacement by proteins 206
replacement by rubidium 206
in tissues 299
Potentiometer 300
Poxvirus 247
PQQ See Pyrroloquinoline quinone
Preproteins 519 522. See also Proenzyme(s)
peroxisomal 521
Precursor activation 540
Prenatal diagnosis 26
Prenyl diphosphate (pyrophosphate) 390s
Prenylation 402
of proteins 559
Prenyltransferases 712
Prephenate 485, 690s
conjugative elimination 689
Prepro-opiomelanocortin 522
Preproinsulin 519
Pressure jump method 468
Primary kinetic isotope effect
for glucose-6-phosphate isomerase
592, 593, 693
Primary hydration sphere 311
Primary structure
of nucleic acids 200 202
of protein 56
Priming reaction in metabolism 507
Prion 248
Priority sequence 42
Procapsid 366
Processive mechanism
of -amylase 606
Prochiral centers 478, 768
trigonal 480
Prochlorophytes 10
Procollagen 432, 432s
Procollagen peptidases 433
Product inhibition 464, 475
Proenzyme(s), 519, 609. See also Zymogen
of histidine decarboxylase 754
Proflavine 223s
Progesterone 696
Programmed cell death. See Apoptosis
Progress curve for enzyme-catalyzed
reaction 455
Proinsulin 519
Projection formula
Fisher 42
Newman 44
Prokaryotic cells, definition of 2
Proline (Pro, P) 52s
in helices 69
reductases 753, 755
Proline rings, torsion angles 62
Prontosil 473s
Proofreading
by enzymes 482
1,2-Propanediol 874s
Propeller 67, 560, 764
Propionic acid, pK
a
value of 293
Propionic acid bacteria 725
PEP carboxytransphosphorylase 706
Propionyl-CoA, carboxylation of 724
Propranolol 553s
Prostacyclin 557
Prostaglandin(s) 565
Prostaglandin PGE
2
381s
Prostanoids 381
Prostate gland, zinc content 680
Prosthetic groups 312, 719
Protamines 204, 239
Protease (proteinase) 609. See also
Proteosome
aspartic 621 625
ATP-dependent 627, 628
in blood clotting 631 634
cysteine 618, 619
in defense systems 628
E. coli 628
functions of 628
metallo- 625 627
multicatalytic 339, 525, 620
pH dependence of 616
serine. See Serine proteases
specificities, table 117
subsites of, nomenclature 617
substrate specificities 616 618
V-8, specificity 117
stereoelectronic effect 616
Protease inhibitors 622, 629 631
alpha 1 629, 631
Proteasome 523
Protection mapping. See Footprinting
Protein(s)
amino acid composition 59, 115 123
anchors for 402
aromatic clusters 75
caloric value 283
charged groups 101
chemical modification 79 81
classification of residues 79
compositions of specialized 59
content
from nitrogen analysis 31
in tissues 31
covalent structure 57
crosslinking 79 81
crossover connections 74s
degradation of 628
disulfide crosslinkages 80
domains 74
engineering of 85
families 76
globular 59
hydration of 50
hydrogen bonding of side chains 75
hydrogen bonds in 75
hydrolysis 116 118
idealized shapes 60
induction of synthesis 538, 539
intracellular degradation 523
isoelectric point 83
maturation 519
methylation of 79
molecular mass 59
multienzyme complexes 348
nucleotide-binding domain 76
oxidation of 85
oxygen carrying 850 853
packing density of 75
pH, effects of 83
phosphoadenylation of 79
phosphorylation of 79
primary structure 56
processing of 519
repression of genes 538, 539
salting in 101
separation with ammonium sulfate
101
selenium-containing, table 825
side chain groups, reactions of
123 127
930 Volume 1 Index
solubility 101
solvent, effects of 83
sulfate esters of 79
thermostability 85
transport of 519 521
turnover 538, 539
water molecules in 76
Protein(s), association
binding equilibria 350 352
dimerizing 350 352
interfaces 74
quaternary structure 332
regulatory subunits 348
subunits of 74, 332
tertiary structure 59
unsymmetrical dimerization 344
Protein(s), conformational states
compact denatured state 82
conformational changes in 81
conformational substates 496
denaturation of 82
dynamic properties 81 85
local unfolding 81
mobile defects in 496
packing defects 81
Protein(s), folding patterns 76, 77
/ barrel 77
/ barrel 77
architecture of folded 59 76
folding intermediates 82
folding of 81 83, 518, 519
molten globule 82
motifs 367
sandwich 64
secondary structure 63
seven-bladed propeller 561
seven-helix 553 556
symmetry in 77
winged helix 243
Protein C 634
Protein coats of viruses 334
Protein concentrations, estimation of 102
Protein data bank 149
Protein disulfide isomerases 83, 659, 787
Protein kinase 541, 656, 659
cGMP-activated 544
Protein kinase C 544, 564
Protein kinase, cAMP-dependent active site
657
Protein phosphatases 544, 646
Protein S 634
Protein sequenators 118
Protein sequences from genes 119
Protein synthesis 3, 538, 539
Protein tyrosine kinases 544
Protein-disulfide isomerase 83
Protein-DNA interactions 266
Proteinase. See Protease
Proteoglycan(s) 181, 182. See also Glycosami-
noglycans
aggregates of cartilage 182
molecular architecture 182
terminal unit of polysaccharides 181
Proteolipid protein of myelin 401
Proteome 116
Proteosome 620, 627
20S 339, 341s
26S 340
electron micrograph 340
Prothrombin 632, 821
Prothrombinase 631
Protista 18 22
Protoheme(s) 844, 845s
Proton(s)
conduction, lateral 401
dissociation, diffusion-controlled 493
intramolecular transfer 693
inventory technique 493, 618
Proton transfer
coupled 493
Marcus theory 493
rates 492
ultra-fast 491
Proto-oncogenes 571 576, 574
Protoplast(s) 6
Protozoa 18 20
Provirus 248
Proximity factor in catalysis 495
Pseudo first order reactions 457
Pseudoazurin 884
Pseudoknot in RNA 231
Pseudomonad(s) 8, 688
Pseudorotation 212, 638
Pseudosubstrate of acetylcholinesterase 636
Pseudouridine 234s
Pseudo vitamin B
12
867
Psicose 164s
Psoriasin 313
Psoriasis 316, 440
PTEN 566
Pteridine 803
ring system 802 804
Pteridophyta 29
Pterin(s) 802
coenzymes 802 813
structures 802 804
Puffballs 20
Pufferfish, genome 12
Pullulan 170, 175
Purine 199, 199s
catabolism of 810
Purine bases
absorption spectra of 205
hydrogen-bonding possibilities 207
names of 203
pK
a
values of 204, 205
Purine hydroxylase 890
Purple acid phosphatases 658, 862, 863
Purple Cu
A
center 883, 884
Putidaredoxin 859
EPR spectrum of 860
Putrescine 363
Pycnodysostosis 619
Pyochelin 838s
Pyranose rings 161, 162, 175. See also Sugars
Pyrenoid 20
Pyridine
binding constants to metal ions 311
pK
a
value of 293
resonance energy 299
Pyridine nucleotide(s) 765
chemistry 779, 780
coenzymes 767 780
6-Pyridone(s) 780s
Pyridoxal 738
Pyridoxal phosphate (PLP) 121, 679, 737
753, 740s. See also PLP-dependent
reactions
absorption spectrum 749
as active site label 124
amino acid reactions 740
in aspartate aminotransferase 57
as coenzyme, table 743
dependent enzymes, cyclization 741
enzymes, atomic structures 750
in glycogen phosphorylase 604
mechanism of action 740, 741, 751 753
NMR spectrum of 138
optical properties 749, 750
Schiff bases, reactions of 699, 705, 742
transimination 741
Pyridoxal phosphate-dependent enzymes,
list 743
Pyridoxal phosphate-dependent reactions
741 747
beta replacement 742 744
cyclization 741
decarboxylation 745
elimination 742
ketimine intermediate 745, 746
nonenzymatic models 737
racemization 741
side chain cleavage 745
stereochemistry of 749
transamination 741
Pyridoxamine 738
orange product with ninhydrin 121
Pyridoxamine phosphate (PMP) 741, 744s,
747
Pyridoxamine:pyruvate aminotransferase,
stereochemistry of 748
Pyridoxine (vitamin B
6
) 305s, 738
microscopic dissociation constants 305
pK
a
values of 305
tautomerism 45
-Pyridoxyllysine 750s
Pyrimethamine 805s
Pyrimidine 199s
Pyrimidine bases
absorption spectra of 205
hydrogen-bonding possibilities 207
names of 203
pK
a
values of 204, 205
Pyrithiamin 731, 736
Pyroglutamate aminopeptidase 619
Pyroglutamyl group as N-terminus
in proteins 54, 522
Pyrophosphatases 515, 646
Pyrophosphate (PP
i
) 508, 515
in adenylate system 302 304
Pyrophospho group
transfer from ATP 660
Pyrophosphoric acid, pK
a
value of 99, 293
Pyrrole, resonance energy 299
Pyrroloquinoline quinone (PQQ) 764, 815
Pyrrophyta 20
Pyruvaldehyde. See Methylglyoxal
Pyruvate 506s, 510
anaerobic cleavage 800
decarboxylation, reversible 799
Pyruvate carboxylase 711, 724
control mechanisms 729
Pyruvate decarboxylase 718, 734
Pyruvate dehydrogenase 511, 796 798, 797s
effect of insulin 571
Pyruvate:ferredoxin oxidoreductase 799
Pyruvate formate-lyase 875
reaction 800, 801
Pyruvate kinase 656, 706
equilibria by NMR 641
Pyruvate oxidase 799
active site 734
Pyruvic acid, pK
a
value of 293
Pyruvoyl groups in enzymes, 753 755
table 753
931 Volume 1 Index
Q
Quanine quartet 208s
Quantum mechanical tunneling 494, 771, 848
Quasi-equivalence
in oligomers 344
in virus coats 346
Queosine (nucleoside Q) 234s
Quinone(s) 815 822
as electron carriers 819
Quinonoid-carbanionic intermediate 741, 744s
from pyridoxal phosphate 743, 744s
Quinoproteins 815 818
copper 815
R
Rab protein 559
Rabies 247
Racemase(s) 284
amino acid 692
aspartate 741
Racemization 741
of amino acids 284
of lens proteins 85
PLP-dependent enzymes 741
Rac protein 559
Radicals, free. See Free radicals
Radioautogram (radioautograph, autoradio-
graph) 111
Radioautography 110
Radioimmunoassays 110
Radioisotope 258
Radiolaria 18
Radius
collision 462
covalent 40, 41
van der Waals 40
Raf 578, 579
Raffinose 169
Ramachandran plot 60, 61
Random order of binding 464
Random coil conformation 69
Rapamycin 488
Rapid equilibrium assumption 467
Rapid photometric methods 468
Ras 577 579
human c-H ras 560s
P loop 559
Ras oncogenes (ras) 572, 576
Ras protein superfamily 558
Rat liver, composition of 31
Rates
of approach to equilibrium 458
of displacement reactions 589, 590
of enzymatic reactions 455 497. See
also Kinetics
of substrate binding 463
Rate constants 455
apparent first order 458
bimolecular 458, 462
prediction by Hammett equation 309
Rate equation for enzymes 455
Reaction coordinate 482
Reaction rates, diffusion controlled limit 462
Reaction types in metabolism, 530
table of 526, 527
Reading frames in nucleic acids 236, 237
Rearrangement reactions 527, 530, 712
recA protein 219
Receptor(s) 1, 553 563
acetylcholine 422
alpha adrenergic. See -Adrenergic
receptors
autophosphorylation of 562
bacterial, for aspartate 561
beta adrenergic. See -Adrenergic
receptors
on cell surfaces 479
down regulation of 571
for hormones 553 563
human adrenergic 555
for surface proteins 407
seven-helix structures 555
Receptor theory 479
Reciprocal plots
for kinetics of enzymes 460, 465, 472
Recognition domains in proteins 367
Rectus configuration 42
Red blood cell. See Erythrocytes
Redox potential
within cells 767
of flavoproteins 767
Reduced flavins, reactions with oxygen 794
Reducing equivalents from citric acid cycle
515
Reducing powers of redox couples 300
Reduction potential(s)
definition of 300
table, 301
Reference electrode 300
Refinement of X-ray structure 136
Refolding of proteins 82
Regulation
of activity of enzymes 539 553
of aspartate carbamoyltransferase 540
control elements in 535 537
of enzymatic activity 535 581
pacemaker enzymes 535 537
Regulators of G-protein signaling (RGS) 559
Regulatory cascade(s) 566, 567
monocyclic (scheme) 567
Regulatory subunits 348, 540, 541
Relative molecular mass (M
r
)
determination of 108 115
estimation by gel filtration 109
estimation by PAGE 109
Relaxation methods 468
Relaxation time
approach to equilibrium 458, 468
first order reactions 457
Renin 621
Reoviruses 248. See also front cover
Repair systems 16
Replication cycle. See Cell cycle
Repression of enzyme synthesis 536, 538, 539
Repressor(s) 76, 239
of gene expression 539
Resolvase 229
Tn3 219
Resonance 45, 46, 46
energy values, table 299
in esters and amides 608
Respiration 300
Respiratory chain 783
Respiratory distress syndrome 386
Resting potential of cell membrane 400
Restriction endonuclease(s) 249, 653
Restriction maps 260
Resurrection plant 168
Retention signal 521
Retinoblastoma protein (Rb) 574
Retinol-binding protein 58
Retrieval signal KDEL 521
Retroviruses 248
Reverse transcriptase 248, 257, 657
Reversed phase columns 103
Reversible chemical reactions 284
kinetics of 458
Rhabdoviruses 247
Rhamnogalacturonan 177
Rhamnose (Rha) 165s, 180
Rheumatoid arthritis 627
Rhinovirus 247
icosahedral 344s
Rhizopoda 18
Rho protein 558
Rhodophyta 22
Ribbon drawing 64, 65, 71, 240, 243, 336, 338,
343, 347, 348, 372, 409, 412, 413, 419, 488,
560, 580, 599, 606, 614, 633, 708, 786, 807,
840, 841
Ribisco activase 707
Ribitol, conformation 44
Ribitolteichoic acids 431s
Riboflavin 721, 766, 783s
absorption spectrum 781
fluorescence 783
light sensitivity 783
nutritional requirement 756, 783
properties 783
Riboflavin 5'-phosphate (flavin mononucleo-
tide, FMN) 513, 765, 780, 781s
Ribonuclease 264, 647, 648
fungal 648
pancreatic, diffusion constant of 461
refolding of 82
Ribonuclease A
mechanism of catalysis 647
NMR spectra of 647
pK
a
values of 647
reaction mechanism 648
sequence of 647
X-ray structural studies 647
in-line mechanisms 647
Ribonuclease P 649 652
Ribonuclease S 647
Ribonucleoprotein domain 244
Ribonucleotide reductase 863 865
active site 865
cobalamin dependent 871
enzyme-activated inhibitors 864
Ribose 1, 5-bisphosphate
in brain 545
Ribose 5-phosphate isomerase 693
D-Ribose (Rib) 162, 163s, 200s
Ribosome(s) 3, 5, 11, 233 235
structure of 233, 234
Ribosylthymidine (Thd) 203
Ribosylthymidine 5'-phosphate (Thd-5P) 203
Ribothymidine 234
of tRNA 231
Ribothymidylic acid residue 813
Ribozyme(s) 649 652, 239
artificial 652
hairpin 649, 651s
hammerhead 649, 651
leadzyme, 651s
Ribulose 164s
Ribulose bisphosphate 707s
Ribulose bisphosphate carboxylase (Rubisco)
932 Volume 1 Index
549, 706 710, 708s
carbamoylation of 707
mechanism, scheme 709
spinach, active site 708
Rice, genome 12
Ricin 186
Ricinoleic acid 381, 381s
Rickets 421, 721
X-linked hypophosphatemic 625
Rickettsias 7
Rieske iron-sulfur protein 860
Ring conformations 332 337
boat 598
half-chair 598
Ring current 139
RNA 5, 230 236
antisense 237
aptamers 235
base composition of 239
binding proteins 243
content in tissues 31
density gradient centrifugation 101
electrophoresis 249
extraction of 249
forms of, table 232
Gibbs energy of formation 210
hydrolysis of 249, 251
loops and turns 210s, 230
pseudoknot 230
figure 231
self-splicing 649
sequence determination 260 266
RNA polymerase 657
zinc in 680
RNase T1 648
RNAworld 235
Rocky Mountain spotted fever 7
Rods (visual receptor cells) 390
Root hairs, dimensions of 30
Roseoflavin 788, 789s
Rossmann fold. See Nucleotide-binding
domain
Rotamases 488
Rotary diffusion constant 463
Rotation of molecules 462, 463
Rotational barrier 44
Rotifers 24, 25
Roundworms (Nematoda) 24
Rp (phosphorylation state ratio) 303
rRNA (ribosomal RNA) 5, 230, 233s
RS notation
for configuration 42
for trigonal centers 480
Rubella 247
Rubidium 31
Rubisco See Ribulose bisphosphate
carboxylase
Rubredoxin(s) 858[s ], 859
Fe-S cluster 857s
Ruhemanns purple 121s
Rusticyanin 883
Rusts 20
Ryanodine receptor 314, 422, 564
S
S-phase of cell cycle 579
Saccharomyces cerevisiae
figure 20
genome 12, 20
Salmonella 7
PEP carboxylase in 711
serotypes 180
Salt bridges 47
of hemoglobin 357
Salting in/out of proteins 101
Salvarsan 597s
SAM. See S-adenosylmethionine (Adomet)
Samuelsson, Bergt I. 84
Sandwich 64
Sanger, Frederick 83, 84, 118
Sarcodina 18
Sarcomas 573
Satellite tobacco necrosis virus 247
Saturation in binding 326
Saturation curve(s) 326 331, 458 461, 475
477
hyperbolic 326
sigmoidal 326
Saturation fraction
definition of 326
for cooperative processes 331
Scanning mutagenesis 402
Scanning tunneling microscope 131
Scatchard plot 327
Scenedesmus 22
Schiff base(s) 121, 752
in aminotransferases 750
of pyridoxal phosphate 742
of pyridoxal phosphate, 740s
reduction of
by borohydride 124, 679
Schistosoma 24
Schistosomes
protease inhibitors 622
Schistosomiasis 24, 222
Schizophyllan 170, 174
Schwann cells 390
Sclerenchyma 30
Scorpions 24
Scrapie 248
Screw-sense isomer of ATP complexes 643
Scurvy 721
SDS electrophoresis 111
SDS-PAGE 106
of erythrocyte ghosts, figure 404
SDS-polyacrylamide gel electrophoresis. See
SDS-PAGE
Sea cucumbers 25
Sea squirts. See tunicates
Sea urchin 25
genome 12
Second Law of Thermodynamics 284285
Second messengers 553
calcium ions 555
cyclic ADP-ribose 555
cyclic AMP 555
cyclic GMP 555
diadenosine 5' tetraphosphate 555
guanosine 5' diphosphate, 3-diphos-
phate (ppGpp) 555
inositol-1,4,5 trisphosphate 555
for insulin 569 571
Mn
2+
555
table of 555
Second order reactions 458
Secondary kinetic isotope effect 592, 600
on fumarate hydratase 684
Secondary plots for kinetics of multisubstrate
enzymes 465
Secondary structure 63
of a protein 63
Secretion gene (Se) 185
Secretion granules 10, 14
Sedimentation constant 100, 109
of nucleic acids 112
of proteins 112
Sedimentation equilibrium 108
Sedimentation velocity 108
Sedoheptulose 164s
Seeds 30
S-Selanylcysteine 893
Selectins 187, 188
Selenium
in enzymes 822 827
metabolism of 827
in proteins, table 825
proteins, incorporation into 827
Selenocysteine 237, 823, 824
in hydrogenases 878
Selenocysteine lyase 746
Selenoprotein P 824
Selenoprotein W 824
Self-assembly of macromolecules 332
Seliwanoffs reagent 167
Semiquinone radicals of flavins 792, 794
Sensitivity coefficient 537
Sensor kinase/response regulator systems
545
Separations of compounds 98 108
by chromatography 100 106
by partition 102 103
Sephadex, gel networks of 175
Sepiapterin 803, 804s
Septate desmosomes 29
Septum 5
Sequence(s)
of amino acids 57
of nucleic acids
Sequencing primers 260
L-Serine (Ser, S) 52, 53s, 808
as nucleophile 609, 610
pK
a
value of 487
Serine carboxypeptidases 610
Serine dehydratases 688, 742
Serine hydroxymethyltransferase 745
quinonoid intermediate 750
Serine proteases 609 618
acyl-enzyme intermediates 610
carboxypeptidase 609, 625
catalytic cycle 613
catalytic triad 614
chymotrypsin 66, 457, 477, 609
oxyanion hole 614, 615
papain 618
pepsin 456, 621
specificities 117
thrombin 632
trypsin 66, 116, 457, 609
Serine O-sulfate 739
Serotonin(5-hydroxytryptamine) 744
Serpins 629
Serum albumin 58
helices in 70
Serum response factor 578
Sex factors 249
SH2 and SH3 domains 367, 580
Sharp, Phillip A. 84
Shc 579
as adaptor protein 570, 577
definition of 577
Sheet, properties of 64
Shells 440, 441
933 Volume 1 Index
Shigella 7
Shikimate 3-phosphate 687s
SH2 / phosphopeptide complex 368s
Sialic acid 165s, 183, 546
Sialidase 186
Sickle cell disease 16, 359, 360
Side chains packing 75
Siderophilins 840
Siderophore(s) 838, 838s
chelate effect 310
receptors 839
Sieve plates 30
Signal peptidase 522, 620
Signal recognition particle (SRP) 520
Signal sequences 519
Signaling 1
Silica gel, use in separation 103
Silica skeleton
of radiolarian 178
Silicon
in biological cross-linking 178
biological functions 178
Silicon dioxide in shells of diatoms 21
Silk 67
structure in 63
fibroin, composition of 59
Simian virus 40 (SV40) 244
Single carbon compounds
in metabolism 808 813
table, according to oxidation state 808
Singlet oxygen 856
Sinister configuration 42
Siroheme 794, 845, 845s, 861
sis 576
v-sis 571, 572
Sizes of atoms, table 41
Skin 439, 440
S-layers of bacteria 431
Slime molds. See Dictyostelium discoideum
Smith degradation of carbohydrates 190
Smoluchowski equation 461, 462
SNAPS 521
SNARES 521
Snell, Esmond 737
Sodium 206
Sodium arsanilate 597s
Sodium borohydride
action on aldoses and ketoses 167
radioactive 679
Sodium borotritide 190
Sodium channels 421
Sodium dithionite 767
Sodium dodecyl sulfate 82s
Sodium ion 31
concentration in cells 206
Sodium-potassium pump 423
Solubility
of proteins 101
separation by 101
Sonication (sonic oscillation) 98
Sorbitol 164, 196, 774
Sorbose 164s
Sos 579
definition of 577
as exchange protein 578
Southern blots 260
Soybean lectin 186
Space groups 133, 332
Specific activity of an enzyme 457
Specific hydroxyl ion catalysis 487
Specificity of enzymatic action 478 482
Spectrin 313, 405
Sperm receptors 187
Spermidine 204, 363
interaction with DNA 218
Spermine
interaction with DNA 218
Spherocytosis 405
Spheroplast 6
Sphinganine 388s
Sphingoglycolipids 388
Sphingolipids 388, 389
structures of 389
Sphingomyelins 389s
Sphingophospholipids 383, 388
Sphingosine (sphingenine) 388s
biosynthesis of 745
Sphingosine 1-phosphate 578
Sphingosylphosphocholine 422
Spicules 23
Spiders 24, 67
Spin decoupling. See NMR spectroscopy
Spin labels 398, 399
Spin-spin interaction. See NMR spectroscopy
Spinach (Spinacia oleracea)
composition of 31
ribulose bisphosphate carboxylase in
706
Spin-orbit coupling 398
Spiral 74
Spirillum, definition of 6
Spirochetes 7
Spirogyra 21
Sponges 23
species-specific reaggregation 187
Spongin 23
Spore(s), bacterial 8
Sporophyte form of fern 18
Sporozoa 19
Squid 24
v-src, src 572
Src, definition of 577
SRP receptor 520
Stachyose 169
Standard electrode potential 300
Standard states 286
Starch 172, 173s
Starch granules 10
electron micrographs 172
layered structure 173
Starch-hydrolyzing enzymes 605 608
Starch-iodine complex 174s
helical structure 173
Starfish 25
State, thermodynamic 281
Statherin 442
Statine (Sta) 623s
Stationary phase of growth curve 470
Statistical effects
in binding 328, 329
Steady state 1, 289
approximation 459
Stearic acid 380s, 381
Stearoyl ACP desaturase 863, 863s
Stellacyanin 883
Stem, section of, figure 31
Stereochemical numbering system 480
Stereochemical studies
using chiral acetates 704
Stereochemistry
of chorismate formation 689
of enzymatic reactions 111
of phosphotransferases 642, 643
of pyridoxal phosphate-requiring
enzymes 747
Stereocilia 370
Stereoelectronic control 598
Stereoelectronic effect
in enzymatic catalysis 495
in proteases 616
Stereospecificity
of dehydrogenases 768 775
prochiral centers 478
Steroid hormones 536
Sterol(s) 389
Stigma 20
STM image of glycogen 174
Stoichiometric dissociation constant 305
Stokes-Einstein equation 461
Stokes radius 109
Stomach ulcer 187
Stomata 30
Stop codons 236
Stopped flow technique 468
Stratum corneum 439
Strecker degradation 121
Strength of binding. See also Formation
constants, Dissociation constants, Gibbs
in metal complexes 307
Streptavidin 728
Stromelysins 627
Strontium 19
Structural domains, table 367
Structure 78
hydrogen bonds in 61
of proteins 61 68
in small peptides 66
twist in 63, 64
Stuart factor 632
Styrene, resonance energy 299
Substituent constants
in Hammett equation 308
table 308
Substitution reactions. See Displacement
reactions
Substrate
definition of 332
Substrate binding
rate of 463
unequal 349, 350
Substrate cycles 567
Substrate saturation
of an enzyme 457
Substrate specificity
of chymotrpsin 616
of serine proteases 616
of trypsin 617
Substrate surrogate 463
Subtilisin 610
active site 612
Subunits in protein structures 74
Succinate 478s
binding constants to metal ions 311
Succinate dehydrogenase (succinic dehydro-
genase) 478, 766, 785
Succinic acid. See also Succinate
pK
a
values of 99, 330
Succinimide
racemization 85
Succinimidyl group 84
Succinyl-CoA acetoacetate CoA transferase
ping-pong mechanism 662
Succinyl-CoA synthetase 661
Succinyl-CoA 516s
934 Volume 1 Index
O-Succinylhomoserine 746s
conversion to cystathionine 746
Sucrose 167, 168s
Sucrose-fructose exchange 595
Sucrose phosphorylase 595
Sugar(s) 161167. See also Monosaccharides,
Oligosaccharides
configurations of 163
covalent hydrate 162
in lipids 380
Sugar isomerases 526
Sugar phosphates
interconversions 508
Sugar transporters
fructose 416
glucose 416
SGLT1-Na
+
dependent 417
Sulfanilamide 473s, 720s
acetylation of 720
Sulfatase(s) 526
Sulfate esters
in proteins 79
Sulfatides 388
Sulfite oxidase 847
family 890
Sulfite reductase(s) 794, 861, 861s
Sulfoquinovose 165s
Sulfogalactosylglycerolipid 387s
Sulfometuron methyl 735s
Sulfonamides 473, 473s
Sulfonolipids 388
Sulfonylureas 421
receptor 421
Sulfotransferases 526, 659
Sulfur displacements 659
Sulfur-35
properties of 110
Summer, James B. 83
Supercoiling
energy of 221
Supercritical fluid chromatography 103
Superhelix 70
density of DNA 221
Superhelix density
effect of intercalation 223
Superoxide anion radical 794, 826, 850, 851
Superoxide dismutase(s) 866, 884
copper, zinc 884
-cylinder in 78
mass spectrum 113, 114
zinc in 680
Superoxide radical
reaction with FADH
2
529
Surface plasmon resonance 122
Surfactant system of the lungs 386
Surfactin 54s
Sutherland, Earl W., Jr. 84
Suzukacillin 414
Svedberg equation 109
Svedberg units 100
Svedburg, Theodor 83
Symbiotic relationship 17
Symmetry 133, 134, 338 348
cubic 342 344
dihedral 338
space groups 133
Syn conformation of nucleosides 211, 212
Syn transfer of protons 696
Synapse(s) 29
electrotonic 29
Synchrotron radiation 137
as X-ray source 134
Synemin 370
Synthetic probe
for nucleic acids 257
System, thermodynamic, definition of 281
T
Taft equation 309
table of constants 308
Tagatose, 164s
Tailspike protein
of bacteriophage 66s
Talin 406
Talose (Tal) 163s
Tamoxifen 575
Tapetum 680
zinc in 680
Tapeworms 24
Taq polymerase 260
Tartaric acid 43s
configuration of 43
Tautomerases 692
Tautomeric catalysis 490, 491
Tautomeric ratios 45, 305
Tautomerism 45, 46
in amides 45
in base pairing 211
of cysteine 306
equilibrium constant 45
in imidazole group 46
in 1-methyluracil 307
of nucleic acid bases 211
of nucleic acids 204, 205, 211
oxo-enol 45
of papain 306
of porphyrins 845
of pyridoxine 305
of pyrimidines 205
Tautomerization
microscopic dissociation constants 305
Taxol 371s
Teeth 440, 441, 442
diagram of 442
Teichoic acids
in cell walls 431
Teichuronic acids
in cell walls 431
Telomeres 208
DNA in 227
Temperature 283, 284
units of 283
Temperature jump method 468
Tenascin 409
Tensin 407
Tenuin 407
Terminal bars 29
N-Terminal nucleophile hydrolases
autoactivation of 621
Termites, protozoa in 19
Tertiary structure of a protein 59
TES buffer 99
pK
a
value of 99
Testosterone 696
1,3,4,6-Tetrachloro-3,6-diphenylglycouril
126s
Tetrahedral intermediate
in displacement reactions 609
stereochemistry of formation 616
Tetrahedron 342
Tetrahydrofolic acid (THF) 745, 802 813,
803s,
derivatives, scheme 806
Tetrahymena 19
Tetrahymena ribozyme 650s
Tetrameric enzyme 342
2,2,6,6-Tetramethylpiperidine-1-oxyl 398s
EPR spectrum 398
Tetramethylsilane (TMS) 138
Tetranitromethane 126
Tetrodotoxin 736
Theileriosis 544
Thermochemistry 282, 283
standard states 286289
Thermodynamics 281 289
base-pairing 209
biochemical standard, new 297
first law of 282, 283
irreversible 289
of life processes 289
nonequilibrium 289
second law of 284, 285
system 281
Thermodynamic barrier 492
Thermodynamic temperature scale 284, 285
Thermolysin 625
specificity 117
Thermostability
of enzymes 85
Thiamin (vitamin B
1
) See also Thiamin
diphosphate
acid base chemistry (scheme) 331
adducts, 733s
analogs 733
biosynthesis of 736
chemical properties of 730, 731
cleavage by bisulfite 731
cooperative binding of protons 331
degradation by base 731
nutritional requirement 756
pK
a
values of 330
polyphosphates 736
tricyclic form 730, 737
yellow anion of 331
thiol form 730
Thiamin coenzymes 730. See also Thiamin
diphosphate
in nerve action 736
in transport of sodium 736
Thiamin diphosphate (Pyrophosphate) 511,
718, 730 737, 767
cleavage 730, 730s
catalytic mechanisms 731 733
enzymes, table 735
enzymatic reactions requiring 734 736
half-reactions, scheme 735
minor tautomer 733
pyrimidine, role of 733
X-ray diffraction 733
Thiazolium dipolar ion (ylid) 732, 733, 733s
Thin layer chromatography 103
of maltooligosaccharides 189
O-methylated glucoses 190
Thin-filaments. See Actin
Thiochrome 730, 730s
Thioesters 559, 775
displacement reactions of 608
formation from glyceraldehyde
3-phosphate 677
function of 700
mechanism of formation 661, 662
Thioether 559
935 Volume 1 Index
Thioglycine 881
Thiohemiacetal in glyceraldehyde phosphate
dehydro 775
Thiol(s) 115
addition to carbonyl group 677
adduct, dehydrogenation of 766
binding of mercury ions 125
Thiol disulfide equilibria 549
Thiol disulfide exchange 659
Thiol:disulfide oxidoreductases 786
Thiolases 699, 827
biosynthetic 699
Thiolate anions 779
Thioltransferases 549, 659, 786
Thioredoxin 522, 549, 659, 786s, 787, 864
disulfide loop 786
functions of 786
Thioredoxin reductase 785, 786, 787, 824
mechanism of 791
Thiosulfate: cyanide sulfurtransferase
symmetry in 78
Thiouridine 234
Three-dimensional structures of
aconitase 689
adenylate kinase 655
aldehyde oxido-reductase 891
D-amino acid oxidase 791
-amylase, pancreatic 607
aspartate aminotransferase 57, 135
catalytic intermediates 752
aspartate carbamyltransferase 348
aspartate chemoreceptor 562
bacteriophage P22 66
cadherin 408
calmodulin 317
carbonic acid anhydrase I 679
carboxypeptidase A 64
catalase 853
cholera toxin 333, 546
chymotrypsin 611
citrate synthase 702, 703
cutinase 134
cyclosporin 488
cytochrome c 847
cytochrome c peroxidase 849
dihydrofolate reductase 807
DNA 214, 223, 228, 229, 241
DNA complex
with methionine repressor 243
with netropsin 225
with Trp repressor protein 240
with zinc finger protein 242
elastase 611
enolase 686
F-actin 338
favin 64
ferredoxin 858
ferritin 343, 841
glucoamylase 77, 608
glutathione reductase 785
glyceraldehyde phosphate
dehydrogenase 63
glycine N-methyltransferase 593
glycogen phosphorylase 605
Grb2 580
HIV-1 protease 624
hydrogenase 879
insulin 347
insulin receptor 570
iron superoxide dismutase 867
K
+
channel 413
kringle 2 motif 368
lactoferrin 840
lysozyme 599
methanol dehydrogenase 816
methionine synthase 874
methylmalonyl-CoA mutase 874
myohemerythrin 71
P pilus 337
pectate lyase C 686
plastocyanin 65
pyruvate dehydrogenase 797
pyruvate oxidase 734
rhinovirus 344
ribulose bisphosphate carboxylase 708
rRNA 233
rubisco 708
rubredoxin 858
stearoyl-acyl carrier protein
desaturase 863
subtilisin 612
sulfite reductase 861
tissue factor 633
trimethylamine dehydrogenase 784
triose phosphate isomerase 77, 694
tRNA 233
trypsin 611
trypsinogen 615
tubulin 372
virus fd protein sheath 335
virus X174 365
L-allo-threonine 53s
Threonine (Thr, T) 53s, 539
biosynthesis 540
cleavage to acetaldehyde 745
configuration 43
Threonine dehydratase 688, 742
Threonine synthase 746
Threose 163s
Thrombin 610, 632
Thrombocytes 26
Thrombomodulin 634
Thrombospondin 409
Thymidine (dThd) 203
Thymidylate synthase 810, 811
mechanism of, scheme 811
Thymine (Thy) 199s, 203
formation 810
Thymineless death 812
Thyroid peroxidase 856
Thyrotropin (thyrotropic hormone)-releasing
factor 54s, 523
receptor 554
Tight junctions 10, 28
Tissue(s)
adipose 26
animal 25
collenchyma 29
composition of 31
connective 26
epidermal of plants 30
epithelial 26
fractionation of 98 100
meristematic 29
muscle 26
parenchyma 29
sclerenchyma 29
supporting 26
Tissue factor 631, 633s
pathway 632
Tissue plasminogen activator 634
Titin 74
Titration curves 96, 97
of proteins 330
Tobacco mosaic virus 247, 334s, 336s
Tocopherols 392, 815 822, 818
as antioxidants 822
-Tocopherol (vitamin E) 819s
Tocopherolquinone 818, 819, 819s
Tocotrienols 818
Togavirus 247
Toluenesulfonyl-amido-2-phenylethyl
chloromethyl ketone (TPCK)
623s
Tomaymycin 224s
TonA 839
TonB 839
Tonegawa, Susumu 84
Tonoplast 11
Topaquinone (TPQ) 816, 817s
Topoisomerases 219, 575, 638, 657, 659
Topological theory of graphs 466
Topologies
of nucleotide binding proteins 76
Topology of proteins 76
diagram 76
Torsion angles 44
definition of 59
of nucleosides 212
of peptides 63, 139
of polynucleotides 211
of side chain groups 61
of sugar rings 212
Toxins
channel-forming 414
Toxopyrimidine 739
TPQ See Topaquinone
Tracheids 30
Tracheophyta 29
Trachoma 7
Traffic ATPases 417
Transacylation 608
Transaldolase 700
Transaminases. See Aminotransferases
Transamination 737
by PLP-dependent enzymes 741
Transcobalamin 869
Transcortin 58
Transcription
definition of 5
of genetic information 5
regulation of 536
Transcription factor
osteoblast-specific 441
Transcription factor(s) 76, 539
AP-1 576
binding to enhancer site 576
GCN4 70, 241
NF-B, 243s
Max 241, 70s
proto-oncogenes as 576
Sp1 202
Transcripts. See mRNA
Transducin 558
Transferases 498, 589 662
aminotransferases 737, 740 743,
747 753
definition of 498
Transferrins 58, 840, 841
Transglutaminase 80, 633
Transimination
in PLP-dependent enzymes 741, 751
Transition metals 837 894
Transition state 482 486
diagram 482
inhibitors 484
936 Volume 1 Index
theory 483
Transketolase 733, 736
Translation of genetic information, 5. See also
Protein synthesis
definition of 5
nick 257
regulation of 536
Translocation
of proteins 519 521
Transmembrane proteins 391
Transmethylation 591, 592
kinetic isotope effects on 592
Transmission coefficient 483
Transport, See also specific substrates
of ions 420 425
through membranes 400 415
via pores and channels 401 403
Transporters
12-helix 415 417
membrane 411 427
sugar 415, 416
Transposable genetic elements (transposons)
249
Tn3 219
Transsialidase 187
Treadmilling
of microtubules 372
Trehalose 167, 168s
in fungi 168
in insects 168
Triacylglycerol 381, 382s
Trialkyl lock 495s
Trichina 24
Tricholomic acid 739
Trichonympha 19
TRICINE buffer 99
pK
a
value of 99
Triclosan 777s, 777
Triethanolamine buffer 99
pK
a
value of 99
Trigonal bipyramid 638
as transition state in pseudorotation
638
Trigonal carbon atoms 680, 681
Trigonal prochiral centers 480, 481
Triiodothyronine 572
Trimethoprim 805s
inhibition of 805
Trimethylamine dehydrogenase 782, 784s
Trimethylarsonium lactic acid, 387s
Trimethyllysine
in ubiquitin 525
Tripeptidyl peptidase 140, 610
Triose phosphate isomerase 693, 694s
barrel structure 77s
high catalytic activity 693
target for antitrypanosomal drugs 693
Triphosphopyridine nucleotide (TPN
+
,
NADP
+
) 767. See also NADP
+
, NADPH
TRIS buffer
pK
a
value of 99
Tritium
properties of 110
Triton X 403s
tRNA 230, 231, 231s, 233s
19
F NMR spectrum of 270
1
H NMR spectrum of 268
wobble position 231
Tropical macrocytic anemia 802
Tropoelastin 436
Tropomodulin 406
Tropomyosin 370, 406
coiled coil structure of 71
Troponin C 313, 314
Trp repressor protein 239, 240s
Truffles 20
Tryosine kinases 657
Trypanosome 19
DNA circles in 219
mitochondria of 14
Trypanothione 552
Trypanothione reductase 785
Tryparsamide 597s
Trypsin 66, 116, 609. See also Chymotrypsin
hydrogen-bonding network, structure
612
specificity 117
turnover number of 457
Trypsinogen 609, 615s
oxyanion hole, structure 615
Tryptases 610
Tryptic peptides 117
Tryptophan (Trp, W) 52s
absorption spectrum of derivative 123
C

-hydroxylated 853
nicotinamide activity 769
Tryptophan indole-lyase 742
quinonoid intermediate 750
Tryptophan synthase 742
Tryptophan tryptophanylquinone (TTQ)
817, 817s
T system of tubules 12
TTQ See Tryptophan tryptophanylquinone
Tuberculosis 7
Tubular myelin 386
Tubulin 370, 372s
Tumor suppressor gene 407, 574
Tungsten 893
Tunicates
vanadium in 25
Tunichlorin(s) 880s, 881
Tunneling
quantum mechanical 494, 771, 848
vibration-assisted 494
Turnover numbers of an enzyme 457
Turns
beta 78
in protein structures 72, 74
Twisted sheets 63
Two-fold (dyad) axes 134
in oligomers 337 348
Typhoid fever
S. typhi 7
Tyrosinase 886
Tyrosine (Tyr, Y) 52s
absorption spectrum of derivative 123
iodination 126
Tyrosine decarboxylase 737
Tyrosine phenol-lyase 742
Tyrosine protein kinases (MEK) 578
Tyrosine-O-sulfate 548
Tyrosyl radical 864
Tyvelose 180s
U
Ubichromanol 818, 819s
Ubiquinones (coenzyme Q) 392, 514, 818,
819s
Ubiquitin 81, 524s
activating enzyme 524, 525
carboxyl-terminal hydrolases 525
conjugating enzymes 524
genes 525
protein ligase 524
UDP-galactose
conversion to UDP glucose 778
UDP-galactose 4-epimerase 778
Ulothrix 21
Ultracentrifugation 108, 109
Ultracentrifuge 100
analytical 108
optical system, figure 109
Ultrafiltration 100
Ultrasensitive responses 567
Ultrasonic sound 468
UMP (Uridine 5'-phosphate) 200, 200s, 203
Unimolecular processes 457
Uniporters 414
Units, International System 2
Unsatisfied ends of hydrogen bonded chains
64
Uracil (Ura, U) 199s, 203
tautomerism of 45
Urate oxidase 886
Urea 82s, 478s
Urea carboxylase 730
Urease 478, 877, 878
active site of, 877s
mechanism of 877
Ureido anion 726s
Uric acid 203s
formation of 890
Uridine (Urd) 203, 234s
absorption spectra of 205
tautomer, minor 205s
Uridine 5'- phosphate. See UMP
Uridine diphosphate glucose (UDP-Glc) 515s,
720
Uridylate kinase 655
5'-Uridylic acid. See UMP
Urocanase reaction 778
Urocanic acid 755, 756s
Urokinase 634
Uronic acid 164
Uroporphyrin(s) 843
Uroporphyrin I, 845s
Urothione 804s, 891
Urticaria 385
Usher protein 364
V
Vacuole(s) 10, 11, 12
definition of 3
Valine (Val, V) 52s
biosynthesis of 527, 712
branched fatty acids from 381
Valinomycin 414, 415s
effect on potassium transport 414
van der Waals contact surfaces
of purines and pyrimidines 207
van der Waals forces 46
van der Waals radii 40, 41
Vanadate
as inhibitor of ATPases 889
insulinlike action 889
Vanadium 856, 889
Vanadocytes 889
Vanadoproteins 889
937 Volume 1 Index
vant Hoff equation 289
Vapor phase chromatography 103
Vascular plants 29
Vasopressin 54s, 542, 563
receptor 554
Vesicle(s), coated 426
VHL (van Hippel-Lindau cancer syndrome)
564
Vibrio 7
definition of 6
Vimentin 369
Vinblastine 371
Vincristine 371
Vinculin 406
Vinylpyridine 116s
Viral oncogenes 573
Virion 244
Viroids 247
Virus(es) 244 249. See also Bacteriophage
adeno 247
baculo 247
binding to cells 186
BK 247
cauliflower mosaic 247
characteristics, table 245
coat 345
DNA, table 245
dsRNA 244, 244 247
electron micrographs of 246
Epstein-Barr 247
hepatitis A 247
hepatitis delta 247
herpes 247
human immunodeficiency virus (HIV)
248
life cycles of 248
Mengo 247
oncogenic 248
papova 244
picorna 247
plant 346
polio 247
polyhedral structures 344
polyoma 247
pox 247
protein coat 334
rhabdo 247
rhino 247
RNA containing 247, 248
table 245
satellite tobacco mosaic 343s
satellite tobacco necrosis 247, 343s, 344
simian (SV40) 244
ssDNA 244
tobacco mosaic 247
toga 247
Virus fd protein sheath 335s
Virus X174
assembly of virion structure 365
Vitamin(s) 719. See also Individual vitamins
deficiency diseases 721
discovery of 721
Vitamin B complex 721
nutritional requirements 756
Vitamin B
1
331. See also Thiamin
Vitamin B
12
721, 866 877. See also Cobalamin,
Cyanocobalamin
adenosyltransferase 870
blood levels 869
cobalt in 866 877
coenzyme forms 867
nutritional requirements 756
Vitamin B
12
coenzyme 864. See also Cobalamin
dependent reactions, table 871
enzymatic functions 870 877
isomerization reactions 872
nonenzymatic cleavage 870
ribonucleotide reductase 871
Vitamin B
6
family 721, 738
nutritional requirements 756
Vitamin C. See Ascorbic acid (ascorbate)
Vitamin D 721
deposition of calcium in bones 314
Vitamin E 721. See also Tocopherols
nutritional requirement 822
Vitamin K 721, 818, 819s, 820 822
in blood clotting 821
dihydro 820
epoxide 820
phylloquinone 821
Vitronectin 409
Voltage-gated K
+
channel 412
Volvox 21
von Szent-Gyrgyi, Albert 83
von Willebrand factor 409, 633
V system 475
W
Wald, George 84
Warburg, Otto H. 83, 767
Warfarin 821, 822s
Water
addition to carbonyl 677
clusters of molecules 49
content in cells 30
content in tissues 31
diffusion constant of 461
hydrogen bonding of 48
properties of 49 51
structure of 49, 50
Watson -Crick structure
of DNA 200
Watson, James D. 84, 200
Watson-Crick base pair 207, 208, 231
Waxes 382, 382s
WD repeat 67
Wheat, genome 12
Whooping cough 548
Wilkins, Maurice F. 200
Wilsons disease 883
Winding number. See DNA, circular, linking
number
Wobble pairing 209
Wood-Ljundahl pathway 881
Work 282
chemical 282
to concentrate 1 mol of a substance
283
electrical 282, 302
electrochemical 300
mechanical 282
to raise 1 kg 1 m 283
to remove two charges 47
World Wide Web
protein sequences/ structures 148
Wortmannin 566s
Wounds, healing of 29
X
X-ray diffraction 132 137
difference electron density map 136
electron density map 135
isomorphous replacement method 133
MAD phasing 135
refinement 136
space groups 133
X-rays
anomolous scattering 135
Xanthan gum 179
Xanthine 203s
Xanthine dehydrogenases 794, 825, 890
molybdenum in 890
Xanthine oxidase 890, 892
Xanthopterin 803, 804s
Xenobiotics 550
X-ray diffraction 611
Xylanases 602, 603
Xylans 165, 170, 175
Xylem 30
Xyloglucan 177
-D-Xylopyranose 695s
Xylose (Xyl) 163s, 165s, 175
Xylose isomerase 527, 693, 695
D-Xylose (Xyl) 162
Xylosidases 602
Xylulose 164s
Y
Yeasts 20
Yellow fever 247
Ylid 733s
Z
Z (zusammen) configuration 43
Zinc 317, 680
in alcohol dehydrogenases 772 775
content of human 680
Zinc finger protein 241, 242s, 243s, 680
Zinc ion(s)
chelation by imidazole groups 625
in enzymes 773
replacements 680
Zinc proteases 625 627
FtsH 628
Zwitterion 41
Zygote 17
Zymogen 519, 609. See also Proenzyme;
Proenzyme(s)
Zymogen granules 609
Zyxin 406
938 Chapter 17. The Organization of Metabolism
Contents
939
939
941
941
942
942
942
944
944
945
947
947
950
950
950
952
952
953
957
958
958
958
960
960
960
962
962
963
964
965
965
966
966
966
966
967
968
970
971
972
972
973
973
973
974
976
977
977
A. The Oxidation of Fatty Acids
1. Beta Oxidation
Peroxisomal beta oxidation
Unsaturated fatty acids
Branched-chain fatty acids
Oxidation of saturated hydrocarbons
Alpha oxidation and omega oxidation
2. Carnitine and Mitochondrial Permeability
3. Human Disorders of Fatty Acid Oxidation
4. Ketone Bodies
B. Catabolism of Propionyl Coenzyme A and Propionate
1. The Malonic Semialdehyde Pathways
2. The Methylmalonyl-CoA Pathway of Propionate
Utilization
C. The Citric Acid Cycle
1. A Clever Way to Cleave a Reluctant Bond
2. Synthesis of the Regenerating Substrate
Oxaloacetate
3. Common Features of Catalytic Cycles
4. Control of the Cycle
5. Catabolism of Intermediates of the Citric
Acid Cycle
D. Oxidative Pathways Related to the Citric Acid Cycle
1. The -Aminobutyrate Cycle
2. The Dicarboxylic Acid Cycle
E. Catabolism of Sugars
1. The Glycolysis Pathway
Formation of pyruvate
The further metabolism of pyruvate
2. Generation of ATP by Substrate Oxidation
3. The Pentose Phosphate Pathways
An oxidative pentose phosphate cycle
Nonoxidative pentose phosphate pathways
4. The Entner Doudoroff Pathway
F. Fermentation: Life without Oxygen
1. Fermentations Based on the EmbdenMeyerhof
Pathway
Homolactic and alcoholic fermentations
Energy relationships
Variations of the alcoholic and homolactic
fermentations
2. The Mixed Acid Fermentation
3. The Propionic Acid Fermentation
4. Butyric Acid and Butanol-Forming Fermentations
5. Fermentations Based on the Phosphogluconate
and Pentose Phosphate Pathways
G. Biosynthesis
1. Metabolic Loops and Biosynthetic Families
2. Key Intermediates and Biosynthetic Families
H. Harnessing the Energy of ATP for Biosynthesis
1. Group Activation
2. Hydrolysis of Pyrophosphate
3. Coupling by Phosphorylation and Subsequent
Cleavage by a Phosphatase
4. Carboxylation and Decarboxylation: Synthesis of
Fatty Acids
978
980
980
981
982
982
982
985
987
989
990
990
992
992
992
993
993
994
995
995
995
996
997
997
999
999
1000
1000
1000
1002
1003
1006
1010
I. Reducing Agents for Biosynthesis
1. Reversing an Oxidative Step with a Strong
Reducing Agent
2. Regulation of the State of Reduction of the NAD
and NADP Systems
3. Reduced Ferredoxin in Reductive Biosynthesis
J. Constructing the Monomer Units
1. Carbonyl Groups in Chain Formation and Cleavage
2. Starting with CO
2
3. Biosynthesis from Other Single-Carbon
Compounds
4. The Glyoxylate Pathways
5. Biosynthesis of Glucose from Three-Carbon
Compounds
6. Building Hydrocarbon Chains with Two-Carbon
Units
7. The Oxoacid Chain Elongation Process
8. Decarboxylation as a Driving Force in Biosynthesis
9. Stabilization and Termination of Chain Growth by
Ring Formation
10. Branched Carbon Chains
K. Biosynthesis and Modification of Polymers
1. Peptides and Proteins
2. Polysaccharides
3. Nucleic Acids
4. Phospholipids and PhosphateSugar Alcohol
Polymers
5. Irreversible Modification and Catabolism of
Polymers
L. Regulation of Biosynthesis
1. Glycogen and Blood Glucose
Insulin
Glucagon
2. Phosphofructo-1-Kinase in the Regulation of
Glycolysis
3. Gluconeogenesis
4. Substrate Cycles
5. Nuclear Magnetic Resonance, Isotopomer
Analysis, and Modeling of Metabolism
6. The Fasting State
7. Lipogenesis
References
Study Questions
Boxes
Box 17-A Refsum Disease
Box 17-B Methylmalonic Aciduria
Box 17-C Use of Isotopic Tracers in Study of the
Tricarboxylic Acid Cycle
Box 17-D Fluoroacetate and Lethal Synthesis
Box 17-E
14
C and the CalvinBenson Cycle
Box 17-F Lactic Acidemia and Other Deficiencies
in Carbohydrate Metabolism
Box 17-G Diabetes Mellitus
Tables
Table 17-1 Products of the Mixed Acid Fermentation
by E. coli at Low and High Values of pH
Table 17-2 Activated Groups Used in Biosynthesis
Table 17-3 Some Effects of Insulin on Enzymes
943
949
954
957
985
1002
1003
968
975
998
OH
C
C
H
H
H2C
COO
COO
COO
Citrate
C
CO2
CO2
C
H
H2C
COO
COO
COO
H
C
C
HO
H
H2C
COO
COO
COO
H
C
C
O
H2C
COO
COO
COO
C
CH2
O
H2C
COO
COO
C
CoA
CH2
O
H2C
S
COO
CH2
H2C
OOC
COO C
C
H
H COO
OOC
C
C
OH
H
H
H
COO
OOC
Oxidation
Electron
Transport
Chain
O
C
C
H
H
COO
OOC
H2O
O
C
S
CH3
CoA
Acetyl-CoA
Pyruvate
O C
CH3
COO
Fatty acids
Glucose
NADH
CoA-SH
H2O
ATP
NADH
GTP
H2O
ADP + Pi
CO2
NADH
FADH2
NADH
O2
2H2O
FADH2 NADH
HCO3

Citric Acid Cycle


Oxidation of
fatty acids (above)
and reactions of
citric acid cycle (right)
occur in mitochondria;
conversion of glucose
to pyruvate takes place
in cytosol of animal cells
Metabolism, a complex network of chemical reactions, occurs in several different
compartments in eukaryotic cells. Fatty acids, a major source of energy for many human
cells, are oxidized in the mitochondria via oxidation and the citric acid cycle. Glucose, a
primary source of energy, is converted to pyruvate in the cytosol. Biosynthetic reactions
occurring in both compartments form proteins, nucleic acids, storage polymers such as
glycogen, and sparingly soluble lipid materials which aggregate to form membranes.
Hydrophobic groups in proteins and other polymers also promote self-assembly of the
cell. At the same time, oxidative processes, initiated by O
2
, increase the water solubility
of molecules, leading to metabolic turnover. Micrograph courtesy of Kenneth Moore.
Ch 17IND page - E - 2/14/03, 11:14 AM 938
939
The Organization of Metabolism
17
Metabolism involves a bewildering array of chem-
ical reactions, many of them organized as complex
cycles which may appear difficult to understand. Yet,
there is logic and orderliness. With few exceptions,
metabolic pathways can be regarded as sequences of
the reactions considered in Chapters 12 16 (and sum-
marized in the table inside the back cover) which are
organized to accomplish specific chemical goals. In
this chapter we will examine the chemical logic of the
major pathways of catabolism of foods and of cell
constituents as well as some reactions of biosynthesis
(anabolism). A few of the sequences have already
been discussed briefly in Chapter 10.
A. The Oxidation of Fatty Acids
Hydrocarbons yield more energy upon combus-
tion than do most other organic compounds, and it is,
therefore, not surprising that one important type of
food reserve, the fats, is essentially hydrocarbon in
nature. In terms of energy content the component
fatty acids are the most important. Most aerobic cells
can oxidize fatty acids completely to CO
2
and water,
a process that takes place within many bacteria, in the
matrix space of animal mitochondria, in the peroxi-
somes of most eukaryotic cells, and to a lesser extent
in the endoplasmic reticulum.
The carboxyl group of a fatty acid provides a point
for chemical attack. The first step is a priming reaction
in which the fatty acid is converted to a water-soluble
acyl-CoA derivative in which the hydrogens of the
fatty acyl radicals are activated (step a, Fig. 17-1).
This synthetic reaction is catalyzed by acyl-CoA
synthetases (fatty acid:CoA ligases). It is driven by
the hydrolysis of ATP to AMP and two inorganic
phosphate ions using the sequence shown in Eq. 10-1
(p. 508). There are isoenzymes that act on short-,
medium-, and long-chain fatty acids. Yeast contains
at least five of these.
1
In every case the acyl group is
activated through formation of an intermediate acyl
adenylate; hydrolysis of the released pyrophosphate
helps to carry the reaction to completion (see discus-
sion in Section H).
1. Beta Oxidation
The reaction steps in the oxidation of long-chain
acyl-CoA molecules to acetyl-CoA were outlined in
Fig. 10-4. Because of the great importance of this
oxidation sequence in metabolism the steps are shown
again in Fig. 17-1 (steps be). The chemical logic
becomes clear if we examine the structure of the acyl-
CoA molecule and consider the types of biochemical
reactions available. If the direct use of O
2
is to be
avoided, the only reasonable mode of attack on an
acyl-CoA molecule is dehydrogenation. Removal of
the hydrogen as a proton is made possible by the
activating effect of the carbonyl group of the thioester.
The hydrogen can be transferred from the inter-
mediate enolate, as a hydride ion, to the bound FAD
present in the acyl-CoA dehydrogenases that cata-
lyze this reaction
25
(step b, Fig. 17-1; see also Eq.
15-23). These enzymes contain FAD, and the reduced
coenzyme FADH
2
that is formed is reoxidized by an
electron transferring flavoprotein (Chapter 15),
which also contains FAD. This protein carries the
electrons abstracted in the oxidation process to the
inner membrane of the mitochondrion where they
enter the mitochondrial electron transport system,
5a
as depicted in Fig. 10-5 and as discussed in detail in
OH
C
C
H
H
H2C
COO

COO

COO

Citrate
C
CO
2
CO
2
C
H
H2C
COO

COO

COO

H
C
C
HO
H
H2C
COO

COO

COO

H
C
C
O
H2C
COO

COO

COO

C
CH2
O
H2C
COO

COO

C
CoA
CH2
O
H2C
S
COO

CH2
H2C
OOC

COO
C
C
H
H COO

OOC

C
C
OH
H
H
H
COO

OOC

Oxidation
Electron
Transport
Chain
O
C
C
H
H
COO

OOC

H
2
O
O
C
S
CH3
CoA
Acetyl-CoA
Pyruvate
O C
CH3
COO

Fatty acids
Glucose
NADH
CoA-SH
H
2
O
ATP
NADH
GTP
H
2
O
ADP + P
i
CO
2
NADH
FADH
2
NADH
O
2
2H
2
O
FADH2 NADH
HCO
3

Citric Acid Cycle


Oxidation of
fatty acids (above)
and reactions of
citric acid cycle (right)
occur in mitochondria;
conversion of glucose
to pyruvate takes place
in cytosol of animal cells
Ch 17IND page - E - 2/14/03, 11:15 AM 939
940 Chapter 17. The Organization of Metabolism
Chapter 18.
The product of step b is always
a trans-
2
-enoyl-CoA. One of the
few possible reactions of this unsat-
urated compound is nucleophilic
addition at the position. The
reacting nucleophile is an HO

ion
from water. This reaction step (step
c, Fig. 17-1) is completed by addi-
tion of H
+
at the position. The
resulting -hydroxyacyl-CoA
(3-hydroxyacyl-CoA) is dehydro-
genated to a ketone by NAD
+
(step
d).
5b
This series of three reactions is
the oxidation sequence.
At the end of this sequence,
the -oxoacyl-CoA derivative is
cleaved (Fig. 17-1, step e) by a
thiolase (see also Eq. 13-35). One
of the products is acetyl-CoA, which
can be catabolized to CO
2
through
the citric acid cycle. The other
product of the thiolytic cleavage is
an acyl-CoA derivative that is two
carbon atoms shorter than the original
acyl-CoA. This molecule is recycled
through the oxidation process,
a two-carbon acetyl unit being
removed as acetyl-CoA during each
turn of the cycle (Fig. 17-1). The
process continues until the fatty
acid chain is completely degraded.
If the original fatty acid contained
an even number of carbon atoms in
a straight chain, acetyl-CoA is the
only product. However, if the
original fatty acid contained an
odd number of carbon atoms,
propionyl-CoA is formed at
the end.
For every step of the oxidation
sequence there is a small family of
enzymes with differing chain length
preferences.
6,7
For example, in
liver mitochondria one acyl-CoA
dehydrogenase acts most rapidly
on n-butyryl and other short-chain
acyl-CoA; a second prefers a sub-
strate of medium chain length such
as n-octanoyl-CoA; a third prefers
long-chain substrates such as pal-
mitoyl-CoA; and a fourth, substrates
with 2-methyl branches. A fifth
enzyme acts specifically on isovaleryl-
CoA. Similar preferences exist for
the other enzymes of the oxida-
tion pathway. In Escherichia coli
CH
3
C
O
S CoA
R
C
CH
2
C
S
O
CoA
H OH
Shortened acyl-CoA
is recycled, a 2 C fragment
being cut off each time
c
H
2
O
-Hydroxyacyl-CoA
R
C
C
C
S
H
H
O
CoA
Acyl-CoA
Enoyl-CoA
b
FAD
FADH
2
R
C
C
C
S
O
H H
H H
CoA
Free fatty acids
ATP
see Eq. 10-1, p. 508
a
e
Oxidation through
citric acid cycle
Acetyl-CoA
When chain degradation is complete,
a short terminal piece remains:
Acetyl-CoA if R' = CH
3
Propionyl-CoA if R' = CH
2
CH
3
(CH
2
)
2
C
O
S CoA
R 2CH
2
R' =
R'
d
NAD
+
NADH + H
+
R
C
CH
2
C
S
O
H
+
O
CoA
CoA S

-Oxoacyl-CoA
Acyl-CoA dehydrogenase
Enoyl-CoA hydratase
-Hydroxyacyl-CoA dehydrogenase
Thiolase
The
Oxidation
Sequence
Figure 17-1 The oxidation cycle for fatty acids. Fatty acids are converted to
acyl-CoA derivatives from which 2-carbon atoms are cut off as acetyl-CoA to
give a shortened chain which is repeatedly sent back through the cycle until
only a 2- or 3-carbon acyl-CoA remains. The sequence of steps b, c, and d also
occurs in many other places in metabolism.
Ch 17IND page - E - 2/14/03, 11:15 AM 940
941
most of these enzymes are present as a complex of multi-
functional proteins
8
while the mitochondrial enzymes
may be organized as a multiprotein complex.
9,10
Peroxisomal beta oxidation. In animal cells
oxidation is primarily a mitochondrial process,
5
but
it also takes place to a limited extent within peroxi-
somes and within the endoplasmic reticulum.
1114
This division of labor is still not understood well.
Straight-chain fatty acids up to 18 carbons in length
appear to be metabolized primarily in mitochondria,
but in the liver fatty acids with very long chains are
processed largely in peroxisomes.
13
There, a very
long-chain acyl-CoA synthetase acts on fatty acids
that contain 22 or more carbon atoms.
15
In yeast all
oxidation takes place in peroxisomes,
15,16
and in most
organisms, including green plants,
1718a
the peroxi-
somes are the most active sites of fatty acid oxidation.
However, animal peroxisomes cannot oxidize short-
chain acyl-CoA molecules; they must be returned to
the mitochondria.
16
The activity of peroxisomes in
oxidation is greatly increased by the presence of a
variety of compounds known as peroxisome prolif-
erators. Among them are drugs such as aspirin and
clofibrate and environmental xenobiotics such as
the plasticizer bis-(2-ethyl-hexyl)-
phthalate. They may induce as much
as a tenfold increase in peroxisomal
oxidation.
11,12,19,19a
Several other features also distin-
guish oxidation in peroxisomes.
The peroxisomal flavoproteins that
catalyze the dehydrogenation of
acyl-CoA molecules to unsaturated
enoyl-CoAs (step b of Fig. 17-1) are
oxidases in which the FADH
2
that is
formed is reoxidized by O
2
to form
H
2
O
2
.
13,20
In peroxisomes the enoyl-
hydratase and the NAD
+
-dependent
dehydrogenase catalyzing steps c and
d of Fig. 17-1 are present together
with an enoyl-CoA isomerase (next
section) as a trifunctional enzyme
consisting of a single polypeptide
chain.
21
As in mitochondrial oxida-
tion the 3-hydroxyacyl-CoA inter-
mediates formed in both animal
peroxisomes and plant peroxisomes
(glyoxysomes) have the L configura-
tion. However, in fungal peroxi-
somes as well as in E. coli they have
the D configuration.
22,23
Further
metabolism in these organisms
requires an epimerase that converts
the D-hydroxyacyl-CoA molecules
to L.
24
In the past it has often been
assumed that peroxisomal membranes
are freely permeable to NAD
+
, NADH, and acyl-CoA
molecules. However, genetic experiments with yeast
and other recent evidence indicate that they are imper-
meable in vivo and that carrier and shuttle mechanisms
similar to those in mitochondria may be required.
14,25
Unsaturated fatty acids. Mitochondrial oxi-
dation of such unsaturated acids as the
9
-oleic acid
begins with removal of two molecules of acetyl-CoA
to form a
5
-acyl-CoA. However, further metabolism
is slow. Two pathways have been identified (Eq.
17-1).
2629b
The first step for both is a normal dehydro-
genation to a 2-trans-5-cis-dienoyl-CoA. In pathway I
this intermediate reacts slowly by the normal oxida-
tion sequence to form a 3-cis-enoyl-CoA intermediate
which must then be acted upon by an auxiliary enzyme,
a cis-
3
-trans-
2
-enoyl-CoA isomerase (Eq. 17-1, step c),
before oxidation can continue.
The alternative reductase pathway (II in Eq. 17-1)
is often faster. It makes use of an additional isomerase
which converts 3-trans, 5-cis-dienoyl-CoA into the
2-trans, 4-trans isomer in which the double bonds are
conjugated with the carbonyl group.
29
This permits
removal of one double bond by reduction with NADPH
as shown (Eq. 17-1, step f ).
29a,29b
The peroxisomal
R
O
5
S
R
O
5
2
CoA
S
R
O
3
CoA
S
O
2
CoA R
c
a Acyl-CoA dehydrogenase
Oxidation continued
Acetyl-CoA
Complete oxidation
I

3
,
2
-Enoyl-CoA
isomerase
b
S
R
O
5 3
CoA
S
O
CoA R
3
S
O
CoA R
2
3-trans-5-cis-Dienoyl-CoA
S
O
4
CoA R
H
+
From
NADPH H

e
f 2,4-Dienoyl-CoA reductase
g Enoyl-CoA isomerase
d
II
Enoyl-CoA
isomerase
(17-1)
A. The Oxidation of Fatty Acids
Ch 17IND page - E - 2/14/03, 11:15 AM 941
942 Chapter 17. The Organization of Metabolism
pathway is similar.
21
However, the intermediate
formed in step e of Eq. 17-1 may sometimes have the 2-
trans, 4-cis configuration.
17
The NADH for the reduc-
tive step f may be supplied by an NADP-dependent
isocitrate dehydrogenase.
29c
Repetition of steps a, d,
e, and f of Eq. 17-1 will lead to oxidation of the
entire chain of polyunsaturated fatty acids such as
linoleoyl-CoA or arachidonoyl-CoA. Important addi-
tional metabolic routes for polyunsaturated fatty acid
derivatives are described in Chapter 21.
Branched-chain fatty acids. Most of the fatty
acids in animal and plant fats have straight unbranched
chains. However, branches, usually consisting of
methyl groups, are present in lipids of some micro-
organisms, in waxes of plant surfaces, and also in
polyprenyl chains. As long as there are not too many
branches and if they occur only in the even-numbered
positions (i.e., on carbons 2, 4, etc.) oxidation proceeds
normally. Propionyl-CoA is formed in addition to
acetyl-CoA as a product of the chain degradation. On
the other hand, if methyl groups occur in positions 3,
5, etc., oxidation is blocked at step d of Fig. 17-1. A
striking example of the effect of such blockage was
provided by the synthetic detergents in common use
until about 1966. These detergents contained a hydro-
carbon chain with methyl groups distributed more or
less randomly along the chain. Beta oxidation was
blocked at many points and the result was a foamy
pollution crisis in sewage plants in the United States
and in some other countries. Since 1966, only bio-
degradable detergents having straight hydrocarbon
chains have been sold.
In fact, cells are able to deal with small amounts of
these hard-to-oxidize substrates. The O
2
-dependent
reactions called oxidation and oxidation are used.
These are related also to the oxidation of hydrocarbons
which we will consider next.
Oxidation of saturated hydrocarbons. Although
the initial oxidation step is chemically difficult, the
tissues of our bodies are able to metabolize saturated
hydrocarbons such as n-heptane slowly, and some
microorganisms oxidize straight-chain hydrocarbons
rapidly.
30,31
Strains of Pseudomonas and of the yeast
Candida have been used to convert petroleum into
CH
2
CH
2
CH
2
C S CoA
O
4 3 2
1
A CH
3
branch here
blocks oxidation
A CH
3
branch here
leads to formation
of propionyl-CoA
edible proteins.
9
The first step in oxidation of alkanes is usually an
O
2
-requiring hydroxylation (Chapter 18) to a primary
alcohol. Further oxidation of the alcohol to an acyl-CoA
derivative, presumably via the aldehyde (Eq. 17-2), is a
frequently encountered biochemical oxidation sequence.
Alpha oxidation and omega oxidation. Animal
tissues degrade such straight-chain fatty acids as
palmitic acid, stearic acid, and oleic acid almost entirely
by oxidation, but plant cells often oxidize fatty acids
one carbon at a time. The initial attack may involve
hydroxylation on the -carbon atom (Eq. 17-3) to
formeither the D- or the L-2-hydroxy acid.
17,18,32,32a
The
L-hydroxy acids are oxidized rapidly, perhaps by dehy-
drogenation to the oxo acids (Eq. 17-3, step b) and
oxidative decarboxylation, possibly utilizing H
2
O
2
(see
Eq. 15-36). The D-hydroxy acids tend to accumulate
CH
2
OH
C
O
S CoA
n-Octane
Hydroxylation
Octanol
O
2
C
7
H
15
C
7
H
15
CHO
C
7
H
15
Acyl-CoA
(17-2)
(17-3)
R CH
2
COO

R CH
2
C S CoA
O
R C
C S CoA
H
OH
O
H
C S CoA
O
R C
COO

O
H
OH
R C
O
H
R C
H
O
R C
O
O

R C
S
O
CoA
a
CoASH
b
c
d
O
2
Fe
2+
2-Oxoglutarate
Ascorbate
Thiamin
diphosphate
(2S), l-
HCOOH
CO
2
Oxidation
(2R), d-
e
Ch 17IND page - E - 2/14/03, 11:15 AM 942
943
and are normally present in green leaves. However,
they too are oxidized further, with retention of the
hydrogen as indicated by the shaded squares in Eq. 17-3,
step e. This suggests a new type of dehydrogenation
with concurrent decarboxylation. Alpha oxidation also
occurs to some extent in animal tissues. For example,
when oxidation is blocked by the presence of a
methyl side chain, the body may use oxidation to get
past the block (see Refsum disease, Box 17-A). As in
plants, this occurs principally in the peroxisomes
3335
and is important for degradation not only of poly-
prenyl chains but also bile acids. In the brain some of
the fatty acyl groups of sphingolipids are hydroxylated
to -hydroxyacyl groups.
36
Alpha oxidation in animal
cells occurs after conversion of free fatty acids to their
acyl-CoA derivatives (Eq. 7-3, step a). This is followed
by a 2-oxoglutarate-dependent hydroxylation (step b,
see also Eq. 18-51) to form the 2-hydroxyacyl-CoA,
which is cleaved in a standard thiamin diphosphate-
requiring cleavage (step c). The products are formyl-
CoA, which is hydrolyzed and oxidized to CO
2
, and
a fatty aldehyde which is metabolized further by
oxidation.
34a
A. The Oxidation of Fatty Acids
BOX 17-A REFSUM DISEASE
b
c
Oxidation is blocked here
Oxidation (Eq. 17-3)
Degradation via oxidation
( )
3
a
Phytol
Phytanic acid
H CH
2
CH CH
2
CH
2
CH
3
CH
2
CH CH
2
C
O
CH
3
OH
R CH
2
C
CH
3
H
C
S
O
CoA
CH
3
CH
2
C
S
O
CoA
CH
3
C
S
O
CoA
CH
3
CH
CH
3
C
O
3
+ 3
S CoA
+
In this autosomally inherited disorder of lipid
metabolism the 20-carbon branched-chain fatty acid
phytanic acid accumulates in tissues. Phytanic acid
is normally formed in the body (step a in the accom-
panying scheme) from the polyprenyl plant alcohol
phytol, which is found as an ester in the chloro-
phyll present in the diet (Fig. 23-20). Although only
a small fraction of the ingested phytol is oxidized
to phytanic acid, this acid accumulates to a certain
extent in animal fats and is present in dairy products.
Because oxidation is blocked, the first step (step b)
in degradation of phytanic acid is oxidation in
peroxisomes.
a
The remainder of the molecule
undergoes oxidation (step c) to three molecules
of propionyl-CoA, three of acetyl-CoA, and one of
isobutyryl-CoA. The disease, which was described
by Refsum in 1946, causes severe damage to nerves
and brain as well as lipid accumulation and early
death.
b d
This rare disorder apparently results
from a defect in the initial hydroxylation. The
causes of the neurological symptoms of Refsum
disease are not clear, but it is possible that the iso-
prenoid phytanic acid interferes with prenylation
of membrane proteins.
b
a
Singh, I., Pahan, K., Dhaunsi, G. S., Lazo, O., and Ozand, P.
(1993) J. Biol. Chem. 268, 99729979
b
Steinberg, D. (1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet, A. L.,
Sly, W. S., and Valle, D., eds), pp. 23512369, McGraw-Hill,
New York
c
Steinberg, D., Herndon, J. H., Jr., Uhlendorf, B. W., Mize, C. E.,
Avigan, J., and Milne, G. W. A. (1967) Science 156, 17401742
d
Muralidharan, V. B., and Kishimoto, Y. (1984) J. Biol. Chem. 259,
1302113026
In plants -dioxygenases (Chapter 18) convert
free fatty acids into 2(R)-hydroperoxy derivatives (Eq.
7-3, step d).
32a
These may be decarboxylated to fatty
aldehydes (step e, see also Eq. 15-36) but may also give
rise to a variety of other products. Compounds arising
from linoleic and linolenic acids are numerous and
include epoxides, epoxy alcohols, dihydroxy acids,
short-chain aldehydes, divinyl ethers, and jasmonic
acid (Eq. 21-18).
32a
On other occasions, omega () oxidation occurs
at the opposite end of the chain to yield a dicarboxylic
acid. Within the human body 3,6-dimethyloctanoic
acid and other branched-chain acids are degraded largely
via oxidation. The initial oxidative attack is by a
hydroxylase of the cytochrome P450 group (Chapter
18). These enzymes act not only on fatty acids but also
on prostaglandins, sterols, and many other lipids. In
the animal body fatty acids are sometimes hydroxylated
both at the terminal () position and at the next (-2 or
2) carbon. In plants hydroxylation may occur at the
2, 3, and 4 positions as well.
17,37
Dicarboxylates
resulting from oxidation of straight-chain fatty acids
Ch 17IND page - E - 2/14/03, 11:15 AM 943
944 Chapter 17. The Organization of Metabolism
can undergo oxidation from both ends. The resulting
short-chain dicarboxylates, which appear to be formed
primarily in the peroxisomes,
38
may be converted by
further oxidation into succinyl-CoA and free succi-
nate.
39
Incomplete oxidation in mitochondria (Fig.
17-1) releases small amounts of 3()-hydroxy fatty
acids, which also undergo oxidation and give rise
to free 3-hydroxydicarboxylic acids which may be
excreted in the urine.
40
2. Carnitine and Mitochondrial Permeability
A major factor controlling the oxidation of fatty
acids is the rate of entry into the mitochondria. While
some long-chain fatty acids (perhaps 30% of the total)
enter mitochondria as such and are converted to CoA
derivatives in the matrix, the majority are activated
to acyl-CoA derivatives on the inner surface of the
outer membranes of the mitochondria. Penetration of
these acyl-CoA derivatives through the mitochondrial
inner membrane is facilitated by L-carnitine.
4144
Carnitine is present in nearly all organisms and in
all animal tissues. The highest concentration is found
in muscle where it accounts for almost 0.1% of the dry
matter. Carnitine was first isolated from meat extracts
in 1905 but the first clue to its biological action was
obtained in 1948 when Fraenkel and associates described
a new dietary factor required by the mealworm, Tenebrio
molitor. At first designated vitamin B
t
, it was identified
in 1952 as carnitine. Most organisms synthesize their
own carnitine from lysine side chains (Eq. 24-30).
The inner membrane of mitochondria contains a long-
chain acyltransferase (carnitine palmitoyltransferase I)
that catalyzes transfer of the fatty acyl group from
CoA to the hydroxyl group of carnitine (Eq. 17-4).
4547a
Perhaps acyl carnitine derivatives pass through the
membrane more easily than do acyl-CoA derivatives
because the positive and negative charges can swing
together and neutralize each other as shown in Eq. 17-4.
Inside the mitochondrion the acyl group is transferred
back from carnitine onto CoA (Eq. 17-4, reverse) by
carnitine palmitoyltransferase II prior to initiation of
oxidation.
Tissues contain not only long-chain acylcarnitines
but also acetylcarnitine and other short-chain acylcar-
nitines, some with branched chains.
41
By accepting
acetyl groups from acetyl-CoA, carnitine causes the
release of free coenzyme A which can then be reused.
l-Carnitine
N COO

OH H
CH
3
H
3
C
H
3
C
+
R C
O
S CoA
R C
O
O
H
N
C
O
O
CH
3
CH
3
CH
3
+
CoA SH
Acyl-CoA
Carnitine
Acyl carnitine

Acyltransferase
(17-4)
Thus, carnitine may have a regulatory function. In
flight muscles of insects acetylcarnitine serves as a
reservoir for acetyl groups. Carnitine acyltransferases
that act on short-chain acyl-CoA molecules are also
present in peroxisomes and microsomes, suggesting
that carnitine may assist in transferring acetyl groups
and other short acyl groups between cell compartments.
For example, acetyl groups from peroxisomal oxida-
tion can be transferred into mitochondria where they
can be oxidized in the citric acid cycle.
41
3. Human Disorders of Fatty Acid Oxidation
Mitochondrial oxidation of fatty acids is the
principal source of energy for the heart. Consequently,
inherited defects of fatty acid oxidation or of carnitine-
assisted transport often appear as serious heart disease
(inherited cardiomyopathy). These may involve heart
failure, pulmonary edema, or sudden infant death.
As many as 1 in 10,000 persons may inherit such prob-
lems.
4850a
The proteins that may be defective include
a plasma membrane carnitine transporter; carnitine
palmitoyltransferases; carnitine/acylcarnitine trans-
locase; long-chain, medium-chain, and short-chain
acyl-CoA dehydrogenases; 2,4-dienoyl-CoA reductase
(Eq. 17-1); and long-chain 3-hydroxyacyl-CoA dehydro-
genase. Some of these are indicated in Fig. 17-2.
Several cases of genetically transmitted carnitine
deficiency in children have been recorded. These
children have weak muscles and their mitochondria
oxidize long-chain fatty acids slowly. If the inner
mitochondrial membrane carnitine palmitoyltrans-
ferase II is lacking, long-chain acylcarnitines accumu-
late in the mitochondria and appear to have damaging
effects on membranes. In the unrelated condition of
acute myocardial ischemia (lack of oxygen, e.g.,
during a heart attack) there is also a large accumulation
of long-chain acylcarnitines.
51,52
These compounds may
induce cardiac arrhythmia and may also account for
Ch 17IND page - E - 2/14/03, 11:15 AM 944
945
Medium- and
short-chain
fatty acids
Long-chain
fatty acids
Carnitine
Acyl-CoA
CoA
Acylcarnitine
Mitochondrial
membranes
Inner Outer
Matrix
Cytosol
Plasma membrane
*
*
Acetyl-CoA
3-Oxoacyl-CoA
Enoyl-CoA
H
2
O
3-Hydroxyacyl-CoA
Acyl-CoA
Carnitine
CoA
Acyl-CoA
dehydrogenase *
Long-chain
3-hydroxyacyl-CoA
dehydrogenase
*
*
Transporter
CPT II
Translocase
CPT I
Beta
oxidation
sudden death from deficiency of carnitine palmitoyl-
transferase II. Treatment of disorders of carnitine
metabolism with daily oral ingestion of several grams
of carnitine is helpful, especially for deficiency of the
plasma membrane transporter.
50a,53
Metabolic abnor-
malities may be corrected completely.
50a
One of the most frequent defects of fatty acid
oxidation is deficiency of a mitochondrial acyl-CoA
dehydrogenase.
50
If the long-chain-specific enzyme
is lacking, the rate of oxidation of such substrates
as octanoate is much less than normal and afflicted
individuals excrete in their urine hexanedioic (adipic),
octanedioic, and decanedioic acids, all products of
oxidation.
54
Much more common is the lack of the
mitochondrial medium-chain acyl-CoA dehydrogenase.
Again, dicarboxylic acids, which are presumably
generated by oxidation in the peroxisomes, are
present in blood and urine. Patients must avoid fasting
and may benefit from extra carnitine.
A deficiency of very long-chain fatty acid oxidation
in peroxisomes is apparently caused by a defective
transporter of the ABC type (Chapter 8).
55
The disease,
X-linked adrenoleukodystrophy (ALD), has received
considerable publicity because of attempts to treat it
with Lorenzos oil, a mixture of triglycerides of oleic
and the C
22
monoenoic erucic acid. The hope has
Figure 17-2 Some specific defects in proteins of oxidation and acyl-carnitine transport causing cardiomyopathy are
indicated by the green asterisks. CPT I and CPT II are carnitine palmitoyltransferases I and II. After Kelly and Strauss.
48
been that these acids would flush out the very long-
chain fatty acids that accumulate in the myelin sheath
of neurons in the central nervous system and may be
responsible for the worst consequences of the disease.
However, there has been only limited success.
56,57
Several genetic diseases involve the development
of peroxisomes.
14,35,58,59
Most serious is the Zellweger
syndrome in which there are no functional peroxi-
somes. Only ghosts of peroxisomes are present and
they fail to take up proteins containing the C-terminal
peroxisome-targeting sequence SKL.
60,60a
There are
many symptoms and death occurs within the first
year. Less serious disorders include the presence of
catalaseless peroxisomes.
60a
4. Ketone Bodies
When a fatty acid with an even number of carbon
atoms is broken down through oxidation the last
intermediate before complete conversion to acetyl-CoA
is the four-carbon acetoacetyl-CoA:
Acetoacetyl-CoA
H
3
C
C
CH
2
C
S
CoA
O O
A. The Oxidation of Fatty Acids
Ch 17IND page - E - 2/14/03, 11:15 AM 945
946 Chapter 17. The Organization of Metabolism
Acetoacetyl-CoA appears to be in equilibrium with
acetyl-CoA within the body and is an important meta-
bolic intermediate. It can be cleaved to two molecules
of acetyl-CoA which can enter the citric acid cycle.
It is also a precursor for synthesis of polyprenyl
(isoprenoid) compounds, and it can give rise to free
acetoacetate, an important constituent of blood.
Acetoacetate is a -oxoacid that can undergo decar-
boxylation to acetone or can be reduced by an NADH-
dependent dehydrogenase to D-3-hydroxybutyrate.
Notice that the configuration of this compound is
opposite to that of L-3-hydroxybutyryl-CoA which is
formed during oxidation of fatty acids (Fig. 17-1).
D-3-Hydroxybutyrate is sometimes stored as a polymer
in bacteria (Box 21-D).
The three compounds, acetoacetate, acetone, and
3-hydroxybutyrate, are known as ketone bodies.
60b
The inability of the animal body to form the glucose
precursors, pyruvate or oxaloacetate, from acetyl units
sometimes causes severe metabolic problems. The
condition known as ketosis, in which excessive
amounts of ketone bodies are present in the blood,
develops when too much acetyl-CoA is produced
and its combustion in the critic acid cycle is slow.
Ketosis often develops in patients with Type I diabetes
mellitus (Box 17-G), in anyone with high fevers, and
during starvation. Ketosis is dangerous, if severe,
because formation of ketone bodies produces hydrogen
ions (Eq. 17-5) and acidifies the blood. Thousands of
young persons with insulin-dependent diabetes die
annually from ketoacidosis.
Rat blood normally contains about 0.07 mM aceto-
acetate, 0.18 mM hydroxybutyrate, and a variable
amount of acetone. These amounts increase to 0.5 mM
acetoacetate and 1.6 mM hydroxybutyrate after 48 h
of starvation. On the other hand, the blood glucose
concentration falls from 6 to 4 mM after 48 h starva-
tion.
61
Under these conditions acetoacetate and hydroxy-
butyrate are an important alternative energy source
for muscle and other tissues.
62,63
Acetoacetate can be
thought of as a transport form of acetyl units, which
can be reconverted to acetyl-CoA and oxidized in the
citric acid cycle.
Some free acetoacetate is formed by direct hydrol-
ysis of acetoacetyl-CoA. In rats, ~ 11% of the hydroxy-
butyrate that is excreted in the urine comes from
acetoacetate generated in this way.
64
However, most
acetoacetate arises in the liver indirectly in a two-step
process (Eq. 17-5) that is closely related to the synthesis
CH
3
C
CH
2
COO

H
OH
d-3-Hydroxybutyrate
of cholesterol and other polyprenyl compounds. Step
a of this sequence is a Claisen condensation, catalyzed
by 3-hydroxy-3-methyl-glutaryl CoA synthase (HMG-
CoA synthase)
64ac
and followed by hydrolysis of one
thioester linkage. It is therefore similar to the citrate
synthase reaction (Eq. 13-38). Step c is a simple aldol
cleavage. The overall reaction has the stoichiometry
of a direct hydrolysis of acetoacetyl-CoA. Liver mito-
chondria contain most of the bodys HMG-CoA syn-
thase and are the major site of ketone body formation
(ketogenesis). Cholesterol is synthesized from HMG-
CoA that is formed in the cytoplasm (Chapter 22).
Utilization of 3-hydroxybutyrate or acetoacetate
for energy requires their reconversion to acetyl-CoA
as indicated in Eq. 17-6. All of the reactions of this
sequence may be nearly at equilibrium in tissues that
use ketone bodies for energy.
61
Acetone, in the small amounts normally present
in the body, is metabolized by hydroxylation to acetol
(Eq. 17-7, step a), hydroxylation and dehydration to
methylglyoxal (step b), and conversion to D-lactate and
pyruvate. A second pathway via 1,2-propanediol and
L-lactate is also shown in Eq. 17-7. During fasting the
acetone content of human blood may rise to as much
as 1.6 mM. As much as two-thirds of this may be
converted to glucose.
6569
Accumulation of acetone
CH
3
C
S CoA
O
2
H
3
C C CH
2
O
C
O
S CoA
H
3
C C CH
2
COO

O
+ H
+
Acetyl-CoA
SH CoA
Acetoacetyl-CoA
SH CoA
Acetoacetate
Acetyl-CoA
a
b
c
S-3-Hydroxy-3-methylglutaryl-CoA (HMG-CoA)
HMG-CoA
synthase
HMG-CoA lyase
C
CH
2
HO
CH
2
C
O
S CoA
OOC
H
3
C

CoA S C CH
3
O
H
2
O
(17-5)
Ch 17IND page - E - 2/14/03, 11:15 AM 946
947
appears to induce the synthesis of the hydroxylases
needed for methylglyoxal formation,
68
and the pyru-
vate formed by Eq. 17-7 may give rise to glucose by
the gluconeogenic pathway. However, at high acetone
concentrations most metabolism may take place through
a poorly understood conversion of 1,2-propanediol to
acetate and formate or CO
2
.
69
No net conversion of
acetate into glucose can occur in animals, but isotopic
labels from acetate can enter glucose via acetyl-CoA
and the citric acid cycle.
B. Catabolism of Propionyl Coenzyme A and
Propionate
Beta oxidation of fatty acids with an odd number
of carbon atoms leads to the formation of propionyl-
CoA as well as acetyl-CoA. The three-carbon propionyl
unit is also produced by degradation of cholesterol
and other isoprenoid compounds and of isoleucine,
valine, threonine, and methionine. Human beings
ingest small amounts of free propionic acid, e.g., from
Swiss cheese (which is cultivated with propionic acid-
producing bacteria) and from propionate added to
bread as a fungicide. In ruminant animals, such as
cattle and sheep, the ingested food undergoes extensive
fermentation in the rumen, a large digestive organ
containing cellulose-digesting bacteria and protozoa.
Major products of the rumen fermentations include
acetate, propionate, and butyrate. Propionate is an
important source of energy for these animals.
1. The Malonic Semialdehyde Pathways
The most obvious route of metabolism of
propionyl-CoA is further oxidation which leads to
3-hydroxypropionyl-CoA (Fig. 17-3, step a). This
appears to be the major pathway in green plants.
17
Continuation of the oxidation via steps ac of Fig.
17-3 produces the CoA derivative of malonic semialde-
hyde. The latter can, in turn, be oxidized to malonyl-
CoA, a -oxoacid which can be decarboxylated to
acetyl-CoA. The necessary enzymes have been found
in Clostridium kluyveri,
70
but the pathway appears to be
little used.
Nevertheless, malonyl-CoA is a major metabolite.
It is an intermediate in fatty acid synthesis (see Fig.
17-12) and is formed in the peroxisomal oxidation
of odd chain-length dicarboxylic acids.
70a
Excess
malonyl-CoA is decarboxylated in peroxisomes, and
lack of the decarboxylase enzyme in mammals causes
the lethal malonic aciduria.
70a
Some propionyl-CoA
may also be metabolized by this pathway. The modi-
fied oxidation sequence indicated on the left side of
Fig. 17-3 is used in green plants and in many micro-
organisms. 3-Hydroxypropionyl-CoA is hydrolyzed
to free -hydroxypropionate, which is then oxidized to
malonic semialdehyde and converted to acetyl-CoA
by reactions that have not been completely described.
Another possible pathway of propionate metabolism
is the direct conversion to pyruvate via oxidation
into lactate, a mechanism that may be employed by
some bacteria. Another route to lactate is through
addition of water to acrylyl-CoA, the product of step a
of Fig. 17-3. The water molecule adds in the wrong
way, the OH

ion going to the carbon instead of


the (Eq. 17-8). An enzyme with an active site similar
to that of histidine ammonia-lyase (Eq. 14-48) could
NAD
+
NADH
Acetoacetate 3-Hydroxybutyrate
Succinyl-CoA
Succinate
(see Eq. 12-50)
Acetoacetyl-CoA
Thiolase CoA SH
2 Acetyl-CoA
Oxidation via citric acid cycle (17-6)
a
O
2
Hydroxylation
C
O
H
3
C CH
3
Acetone
C
O
H
3
C CH
2
OH
Acetol
b
Hydroxylation
C
O
H
3
C CHO
Methylglyoxal
Glyoxalase
(Eq. 13-22)
H
3
C COO

OH H
d-Lactate
Further
metabolism
2[H]
HCOO

CH
3
COO

O
2
ATP + 2[H]
-4[H]
l-Lactate
H
3
C CH
2
OH
H HO
l-1,2-Propanediol
Pyruvate
(17-7)
B. Catabolism of Propionyl Coenzyme A and Propionate
Ch 17IND page - E - 2/14/03, 11:15 AM 947
948 Chapter 17. The Organization of Metabolism
H
3
C
CH
2
C
S CoA
O
H
2
C
CH
C
S CoA
O
CH
2
CH
2
C
S CoA
O
HO
CH
2
CH
2
COO

HO
CH
3
C
O
S CoA

OOC
CH
2
C
S CoA
O
H
3
C
CH
2
COO

3
2
1
Propionate
CoA-SH
ATP
Propionyl-CoA
a
b
c
Acrylyl-CoA
3-Hydroxypropionyl-CoA
H
2
O
CoA-SH
3-Hydroxypropionate
Malonic semialdehyde
Acetyl-CoA
CO
2
CoA-SH
2[H]
Acyl-CoA oxidase
H
2
O
Hydrolase
NAD
+
NAD
+
2[H]
Malonyl-CoA
Pyruvate
CO
2
Oxaloacetate
Biosynthesis
H
3
C
C
C
S CoA
O
H COO

d
Succinate
GDP + P
i
GTP
C
CH
2
C
S CoA
O

OOC
H
2
Succinyl-CoA
H
3
C
C
C
S CoA
O

OOC H
(R)-Methylmalonyl-CoA
(S)-Methylmalonyl-CoA
Racemase
Oxidation
CO
2
NAD
+
CO
2
ATP
ADP + P
i
CO
2
Biotin-enzyme
Catabolism of odd and branched
fatty acids and of isoleucine
M
o
d
i
f
i
e
d


O
x
i
d
a
t
i
o
n

p
a
t
h
w
a
y
Vitamin B
12
coenzyme
M
e
t
h
y
l
m
a
l
o
n
y
l
-
C
o
A

p
a
t
h
w
a
y
C
CH
2
C
S CoA
O
O
H
O
C
CH
2
COO

H
Figure 17-3 Catabolism of propionate and propionyl-CoA. In the names for methylmalonyl-CoA the R and S refer to the
methylmalonyl part of the structure. Coenzyme A is also chiral.
presumably catalyze such a reaction. Lactyl-CoA
could be converted to pyruvate readily. Clostridium
propionicum does interconvert propionate, lactate, and
pyruvate via acrylyl-CoA and lactyl-CoA as part of a
fermentation of alanine (Fig. 24-19).
7174
The enzyme
that catalyzes hydration of acrylyl-CoA in this case is
a complex flavoprotein that may function via a free
radical mechanism.
71,72,74
H
2
C
C
C
S
O
CoA
H
3
C
C
C
S
O
CoA
H HO
H
d-Lactyl-CoA
H
2
O
(17-8)
Ch 17IND page - E - 2/14/03, 11:15 AM 948
949
BOX 17-B METHYLMALONIC ACIDURIA
In this hereditary disease up to 1 2 g of methyl-
malonic acid per day (compared to a normal of <5
mg/day) is excreted in the urine, and a high level
of the compound is present in blood. Two causes
of the rare disease are known.
a d
One is the lack of
functional vitamin B
12
-containing coenzyme. This
can be a result of a mutation in any one of several
different genes involved in the synthesis and trans-
port of the cobalamin coenzyme.
e
Cultured fibro-
blasts from patients with this form of the disease
contain a very low level of the vitamin B
12
coenzyme
(Chapter 16), and addition of excess vitamin B
12
to
the diet may restore coenzyme synthesis to normal.
Among elderly patients a smaller increase in methyl-
malonic acid excretion is a good indicator of vitamin
B
12
deficiency. A second form of the disease, which
does not respond to vitamin B
12
, arises from a defect
in the methylmalonyl mutase protein. Methyl-
malonic aciduria is often a very severe disease,
frequently resulting in death in infancy. Surprisingly,
some children with the condition are healthy and
develop normally.
a,f
A closely related disease is caused by a deficiency
of propionyl-CoA carboxylase.
a
This may be a result
of a defective structural gene for one of the two sub-
units of the enzyme, of a defect in the enzyme that
attaches biotin to carboxylases, or of biotinitase, the
enzyme that hydrolytically releases biotin from
linkage with lysine (Chapter 14). The latter two
defects lead to a multiple carboxylase deficiency and
to methylmalonyl aciduria as well as ketoacidosis
and propionic acidemia.
g
Both methylmalonic aciduria and propionyl-CoA
decarboxylase deficiency are usually accompanied by
severe ketosis, hypoglycemia, and hyperglycinemia.
The cause of these conditions is not entirely clear.
However, methylmalonyl-CoA, which accumulates
in methylmalonic aciduria, is a known inhibitor of
pyruvate carboxylase. Therefore, ketosis may develop
because of impaired conversion of pyruvate to oxalo-
acetate.
Patients with propionic or methylmalonic
acidemia also secrete 2,3-butanediols (D-,L- or meso)
and usually also 1,2-propandiol in their urine.
Secretion of 1,2-propanediol is also observed during
starvation and in diabetic ketoacidosis. Propanediol
may be formed from acetone (Eq. 17-7), and butane-
diols may originate from acetoin, which is a side
reaction product of pyruvate dehydrogenase. How-
ever, in the metabolic defects under consideration
here, acetoin may be formed by hydroxylation of
methylethyl ketone which can arise, as does acetone,
by decarboxylation of an oxoacid precursor formed
by oxidation.
h
Methylmalonic aciduria is rare and can be
diagnosed incorrectly. In 1989 a woman in St. Louis,
Missouri, was convicted and sentenced to life in
prison for murdering her 5-month-old son by
poisoning with ethylene glycol. While in prison
she gave birth to another son who soon fell ill of
methylmalonyl aciduria and was successfully treated.
Reexamination of the evidence revealed that the first
boy had died of the same disease and the mother
was released.
i
a
Fenton, W. A., and Rosenberg, L. E. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 14231449,
McGraw-Hill, New York
b
Matsui, S. M., Mahoney, M. J., and Rosenberg, L. E. (1983) N.
Engl. J. Med. 308, 857861
c
Hubbard, S. R., Wei, L., Ellis, L., and Hendrickson, W. A. (1994)
Nature (London) 372, 746754
d
Luschinsky Drennan, C., Matthews, R. G., Rosenblatt, D. S.,
Ledley, F. D., Fenton, W. A., and Ludwig, M. L. (1996) Proc. Natl.
Acad. Sci. U.S.A. 93, 55505555
e
Fenton, W. A., and Rosenberg, L. E. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 2 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 31293149,
McGraw-Hill, New York
f
Ledley, F. D., Levy, H. L., Shih, V. E., Benjamin, R., and
Mahoney, M. J. (1984) N. Engl. J. Med. 311, 10151018
g
Wolf, B. (1995) in The Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 31513177, McGraw-Hill, New York
h
Casazza, J. P., Song, B. J., and Veech, R. L. (1990) Trends Biochem.
Sci. 15, 2630
i
Zurer, P. (1991) Chem. Eng. News 69 Sep 30, 78
H
3
C C
O
CO
2
CH
2
CH
3
H
3
C C
O
CH
2
CH
2
COO

B. Catabolism of Propionyl Coenzyme A and Propionate


Ch 17IND page - E - 2/14/03, 11:15 AM 949
950 Chapter 17. The Organization of Metabolism
2. The Methylmalonyl-CoA Pathway of
Propionate Utilization
Despite the simplicity and logic of the oxidation
pathway of propionate metabolism, higher animals
use primarily the more complex methylmalonyl-CoA
pathway (Fig. 17-3, step b). This is one of the two
processes in higher animals presently known to
depend upon vitamin B
12
. This vitamin has never
been found in higher plants, nor does the methyl-
malonyl pathway occur in plants. The pathway
(Fig. 17-3) begins with the biotin- and ATP-dependent
carboxylation of propionyl-CoA. The S-methylmalonyl-
CoA so formed is isomerized to R-methylmalonyl-CoA,
after which the methylmalonyl-CoA is converted to
succinyl-CoA in a vitamin B
12
coenzyme-requiring
reaction step d (Table 16-1). The succinyl-CoA is con-
verted to free succinate (with the formation of GTP
compensating for the ATP used initially). The succi-
nate, by oxidation, is converted to oxaloacetate which
is decarboxylated to pyruvate. This, in effect, removes
the carboxyl group that was put on at the beginning of
the sequence in the ATP-dependent step. Pyruvate is
converted by oxidative decarboxylation to acetyl-CoA.
A natural question is Why has this complex
pathway evolved to do something that could have
been done much more directly? One possibility is
that the presence of too much malonyl-CoA, the prod-
uct of the oxidation pathway of propionate metabo-
lism (Fig. 17-3, pathways a and c), would interfere
with lipid metabolism. Malonyl-CoA is formed in
the cytosol during fatty acid biosynthesis and retards
mitochondrial oxidation by inhibiting carnitine
palmitoyltransferase I.
46,70a,75
However, a relationship
to mitochondrial propionate catabolism is not clear.
On the other hand, the tacking on of an extra CO
2
and the use of ATP at the beginning suggests that the
methylmalonyl-CoA pathway (Fig. 17-3) is a biosynthetic
rather than a catabolic route (see Section H,4). The methyl-
malonyl pathway provides a means for converting
propionate to oxaloacetate, a transformation that is
chemically difficult.
In this context it is of interest that cows, whose
metabolism is based much more on acetate than is
ours, often develop a severe ketosis spontaneously.
A standard treatment is the administration of a large
dose of propionate which is presumably effective
because of the ease of its conversion to oxaloacetate
via the methylmalonyl-CoA pathway. It is possible
that this pathway was developed by animals as a
means of capturing propionyl units, scanty though
they may be, for conversion to oxaloacetate and use
in biosynthesis. In ruminant animals, the pathway is
especially important. Whereas we have 5.5 mM glu-
cose in our blood, the cow has only half as much, and
a substantial fraction of this glucose is derived, in the
liver, from the propionate provided by rumen micro-
organisms.
76
The need for vitamin B
12
in the formation
of propionate by these organisms also accounts for
the high requirement for cobalt in the ruminant diet
(Chapter 16).
C. The Citric Acid Cycle
To complete the oxidation of fatty acids the acetyl
units of acetyl-CoA generated in the oxidation
sequence must be oxidized to carbon dioxide and
water.
77
The citric acid (or tricarboxylic acid) cycle
by which this oxidation is accomplished is a vital part
of the metabolism of almost all aerobic creatures. It
occupies a central position in metabolism because of
the fact that acetyl-CoA is also an intermediate in the
catabolism of carbohydrates and of many amino acids
and other compounds. The cycle is depicted in detail
in Fig. 10-6 and in an abbreviated form, but with more
context, in Fig. 17-4.
1. A Clever Way to Cleave a Reluctant Bond
Oxidation of the chemically resistant two-carbon
acetyl group to CO
2
presents a chemical problem. As
we have seen (Chapter 13), cleavage of a CC bond
occurs most frequently between atoms that are and
to a carbonyl group. Such cleavage is clearly
impossible within the acetyl group. The only other
common type of cleavage is that of a CC bond
adjacent to a carbonyl group ( cleavage), a thiamin-
dependent process (Chapter 14). However, cleavage
would require the prior oxidation (hydroxylation) of
the methyl group of acetate. Although many biological
hydroxylation reactions occur, they are rarely used in
the major pathways of rapid catabolism. Perhaps this
is because the overall yield of energy obtainable via
hydroxylation is less than that gained from dehydro-
genation and use of an electron transport chain.
78
The solution to the chemical problem of oxidizing
acetyl groups efficiently is one very commonly found
in nature; a catalytic cycle. Although direct cleavage is
impossible, the two-carbon acetyl group of acetyl-CoA
can undergo a Claisen condensation with a second
compound that contains a carbonyl group. The con-
densation product has more than two carbon atoms,
and a cleavage to yield CO
2
is now possible. Since
the cycle is designed to oxidize acetyl units we can
regard acetyl-CoA as the primary substrate for the
cycle. The carbonyl compound with which it condenses
can be described as the regenerating substrate. To
complete the catalytic cycle it is necessary that two
carbon atoms be removed as CO
2
from the compound
formed by condensation of the two substrates and that
the remaining molecule be reconvertible to the original
regenerating substrate. The reader may wish to play a
Ch 17IND page - E - 2/14/03, 11:15 AM 950
951
Figure 17-4 The Krebs citric acid cycle. Some of its controlling interactions and its relationship to glycolysis. See also Figure
10-6. Positive and negative regulatory influences, whether arising by allosteric effects or via covalent modification, are indicated
by + or . Some biosynthetic reaction pathways related to the cycle are shown in green. Steps are lettered to correspond to
the numbering in Fig. 10-6, which shows more complete structural formulas. Three molecules of H
2
O (boxed) enter the cycle
at each turn, providing hydrogen atoms for generation of NADH + H
+
and reduced ubiquinone (QH
2
). The covalently
attached FAD is designated FAD

.
ATP
ATP
Malate
Fumarate

OOC
COO

Succinate
Isocitrate
ATP
ADP
Acetyl-CoA
ATP
Malonyl-CoA
Beta
oxidation
Fatty acyl-CoAs

ATP
NAD
+

+
O
S CoA
COO

O
COO

Pyruvate
O
COO

PEP
Glucose 6-P
Fructose 6-P
Fructose 1,6-P
2

Fatty acids
Oxaloacetate
+ P
i

Bacteria,
plants
+
NADH
+ H
+
Succinyl-CoA
ATP
CoASH
NAD
+
FAD

H
2
FAD

COO

COO

HO

OOC
Citrate
d
e
a
f
b,c
NAD
+

COO

O
COO

2-Oxoglutarate
NAD
+

C
4
i
H
2
O
C
4
GTP
GDP + P
i H
2
O

ATP
g
h
Accumulating
citrate is exported
from mitochondria,
inhibits early
reaction in
glycolysis, and
activates fatty
acid synthesis
In many organisms
(eukaryotes and
some prokaryotes)
ATP in excess acts
to reduce rate of
cycle. In gram-
negative bacteria
NADH rather than
ATP inhibits
C
6
Aspartate

Porphyrins
Glutamate
H
2
O
HCO
3

HCO
3

HCO
3

CO
2
CO
2
CO
2
Q
QH
2
NAD
+
NADH
+ H
+
C. The Citric Acid Cycle
Ch 17IND page - E - 2/14/03, 11:15 AM 951
952 Chapter 17. The Organization of Metabolism
game by devising suitable sequences of reactions for
an acetyl-oxidizing cycle and finding the simplest
possible regenerating substrate. Ask yourself whether
nature could have used anything simpler than oxalo-
acetate, the molecule actually employed in the citric
acid cycle.
The first step in the citric acid cycle (step a, Fig.
17-4) is the condensation of acetyl-CoA with oxalo-
acetate to form citrate. The synthase that catalyzes
this condensation also removes the CoA by hydrolysis
after it has served its function of activating a methyl
hydrogen. This hydrolysis also helps to drive the cycle
by virtue of the high group transfer potential of the
thioester linkage that is cleaved. Before the citrate can
be degraded through cleavage, the hydroxyl group
must be moved from its tertiary position to an adjacent
carbon where, as a secondary alcohol, it can be oxidized
to a carbonyl group. This is accomplished through
steps b and c, both catalyzed by the enzyme aconitase
(Eq. 13-17). Isocitrate can then be oxidized to the
-oxoacid oxalosuccinate, which does not leave the
enzyme surface but undergoes decarboxylation while
still bound (step d; also see Eq. 13-45).
The second carbon to be removed from citrate is
released as CO
2
through catalysis by the thiamin
diphosphate dependent oxidative decarboxylation
of 2-oxoglutarate (-ketoglutarate; Chapter 15). To
complete the cycle the four-carbon succinyl unit of
succinyl-CoA must be converted back to oxaloacetate
through a pathway requiring two more oxidation
steps: Succinyl-CoA is converted to free succinate
(step f) followed by a oxidation sequence (steps gi;
Figs. 10-6 and 17-4). Steps e and f accomplish a substrate-
level phosphorylation (Fig. 15-16). Succinyl-CoA is an
unstable thioester with a high group transfer potential.
Therefore, step f could be accomplished by simple
hydrolysis. However, this would be energetically
wasteful. The cleavage of succinyl-CoA is coupled to
synthesis of ATP in E. coli and higher plants and to
GTP in mammals. Some of the succinyl-CoA formed
in mitochondria is used in other ways, e.g., as in Eq.
17-6 and for biosynthesis of porphyrins.
2. Synthesis of the Regenerating Substrate
Oxaloacetate
The primary substrate of the citric acid cycle is
acetyl-CoA. Despite many references in the biochemical
literature to substrates entering the cycle as oxalo-
acetate (or as one of the immediate precursors succinate,
fumarate, or malate), these compounds are not consumed
by the cycle but are completely regenerated; hence
the term regenerating substrate, which can be applied
to any of these four substances. A prerequisite for the
operation of a catalytic cycle is that a regenerating
substrate be readily available and that its concentration
be increased if necessary to accommodate a more
rapid rate of reaction of the cycle. Oxaloacetate
can normally be formed in any amount needed for
operation of the citric acid cycle from PEP or from
pyruvate, both of these compounds being available
from metabolism of sugars.
In bacteria and green plants PEP carboxylase
(Eq. 13-53), a highly regulated enzyme, is responsible
for synthesizing oxaloacetate. In animal tissues
pyruvate carboxylase (Eq. 14-3) plays the same role.
The latter enzyme is almost inactive in the absence of
the allosteric effector acetyl-CoA. For this reason, it
went undetected for many years. In the presence of
high concentrations of acetyl-CoA the enzyme is fully
activated and provides for synthesis of a high enough
concentration of oxaloacetate to permit the cycle to
function. Even so, the oxaloacetate concentration in
mitochondria is low, only 0.1 to 0.4 x 10
6
M (10 40
molecules per mitochondrion), and is relatively con-
stant.
65,79
3. Common Features of Catalytic Cycles
The citric acid cycle is not only one of the most
important metabolic cycles in aerobic organisms,
including bacteria, protozoa, fungi, higher plants, and
animals, but also a typical catalytic cycle. Other cycles
also have one or more primary substrates and at least
one regenerating substrate. Associated with every
catalytic cycle there must be a metabolic pathway that
provides for synthesis of the regenerating substrate.
Although it usually needs to operate only slowly to
replenish regenerating substrate lost in side reactions,
the pathway also provides a mechanism for the net
biosynthesis of any desired quantity of any intermediate in
the cycle. Cells draw off from the citric acid cycle
considerable amounts of oxaloacetate, 2-oxoglutarate,
and succinyl-CoA for synthesis of other compounds.
For example, aspartate and glutamate are formed
directly from oxaloacetate and 2-oxoglutarate, respec-
tively, by transamination (Eq. 14-25).
79a,b
Citrate itself
is exported from mitochondria and used for synthesis
of fatty acids. It is often stated that the citric acid cycle
functions in biosynthesis, but when intermediates in
the cycle are drawn off for synthesis the complete cycle
does not operate. Rather, the pathway for synthesis of the
regenerating substrate, together with some of the enzymes
of the cycle, is used to construct a biosynthetic pathway.
The word amphibolic is often applied to those
metabolic sequences that are part of a catabolic cycle
and at the same time are involved in a biosynthetic
(anabolic) pathway. Another term, anaplerotic, is
sometimes used to describe pathways for the synthesis
of regenerating substrates. This word, which was
suggested by H. L. Kornberg, comes from a Greek root
meaning filling up.
80
Ch 17IND page - E - 2/14/03, 11:16 AM 952
953
4. Control of the Cycle
What factors determine the rate of oxidation by
the citric acid cycle? As with most other important
pathways of metabolism, several control mechanisms
operate and different steps may become rate limiting
under different conditions.
81
Factors influencing the
flux through the cycle include (1) the rate of generation
of acetyl groups, (2) the availability of oxaloacetate,
and (3) the rate of reoxidation of NADH to NAD
+
in
the electron transport chain. As indicated in Fig. 17-4,
acetyl-CoA is a positive effector for conversion of
pyruvate to oxaloacetate. Thus, acetyl-CoA turns on
the formation of a substance required for its own
further metabolism. However, when no pyruvate is
available operation of the cycle may be impaired by
lack of oxaloacetate. This may be the case when liver
metabolizes high concentrations of ethanol. The
latter is oxidized to acetate but it cannot provide
oxaloacetate. Accumulating NADH reduces pyruvate
to lactate, further interfering with formation of oxalo-
acetate.
82
In some individuals the accumulating
acetyl units cannot all be oxidized in the cycle and
instead are converted to the ketone bodies (Section
A,4). A similar problem arises during metabolism of
fatty acids by diabetic individuals with inadequate
insulin. The accelerated breakdown of fatty acids in
the liver overwhelms the system and results in ketosis,
even though the oxaloacetate concentration remains
normal.
83
The rates of the oxidative steps in the citric acid
cycle are limited by the rate of reoxidation of NADH
and reduced ubiquinone in the electron transport
chain which may sometimes be restricted by the avail-
ability of O
2
. However, in aerobic organisms this rate
is usually determined by the concentration of ADP
and/or P
i
available for conversion to ATP in the oxida-
tive phosphorylation process (Chapter 18). If catabo-
lism supplies an excess of ATP over that needed to
meet the cells energy needs, the concentration of ADP
falls to a low level, cutting off phosphorylation. At
the same time, ATP is present in high concentration
and acts as a feedback inhibitor for the catabolism
of carbohydrates and fats. This inhibition is exerted
at many points, a few of which are indicated in Fig.
17-4. Important sites of inhibition are the pyruvate
dehydrogenase complex,
8485a
which converts pyru-
vate into acetyl-CoA; isocitrate dehydrogenase,
86,86a
which converts isocitrate into 2-oxoglutarate; and
2-oxoglutarate dehydrogenase.
87
The enzyme
citrate synthase, which catalyzes the first reaction
of the cycle, is also inhibited by ATP.
88,89
Mitochondrial pyruvate dehydrogenase, which
contains a 60-subunit icosohedral core of dihydrolipoyl-
transacylase (Fig. 15-14), is associated with three mole-
cules of a two-subunit kinase as well as six molecules
of a structural binding protein which contains a
lipoyl group that can be reduced and acetylated by
other subunits of the core protein. The binding protein
is apparently essential to the functioning of the dehydro-
genase complex but not through its lipoyl group.
90,91
The specific pyruvate dehydrogenase kinase is thought
to be one of the most important regulatory proteins
involved in controlling energy metabolism in most
organisms.
9292b
Phosphorylation of up to three specific
serine hydroxyl groups in the thiamin-containing
decarboxylase subunit (designated E1) converts the
enzyme into an inactive form (Eq. 17-9). A specific
phosphatase reverses the inhibition. The kinase is
most active on enzymes whose core lipoyl (E2) sub-
units are reduced and acetylated, a condition favored
by high ratios of [acetyl-CoA] to free [CoASH] and of
[NADH] to [NAD
+
]. Since the kinase inactivates the
enzyme the effect is to decrease the pyruvate dehydro-
genase action when the system becomes saturated and
NADH and acetyl-CoA accumulate. Conversely, a high
[pyruvate] inhibits the kinase and increases the action
of the dehydrogenase complex. This system also permits
various external signals to be felt. For example, insulin
has a pronounced stimulatory effect on mitochondrial
energy.
65,92,93
One way in which this may be accom-
plished is through stimulation of the pyruvate dehydro-
genase phosphatase, as indicated in Eq. 17-9. A kinase
activator protein (Eq. 17-9) may also respond to various
external stimuli and may be inhibited by insulin.
92
The activities of 2-oxoglutarate dehydrogenase,
94
and to a lesser extent of pyruvate and isocitrate dehydro-
genases, are increased by increases in the free Ca
2+
concentration.
87
Calcium ions stimulate the phospha-
tase that dephosphorylates the deactivated phosphor-
ylated pyruvate dehydrogenase and activate the other
two dehydrogenases allosterically, increasing the
affinities for the substrates.
87
Phosphorylation of the
NAD
+
-dependent isocitrate dehydrogenase also de-
creases its activity. In E. coli the isocitrate dehydro-
genase kinase and a protein phosphatase exist as a
bifunctional protein able to both deactivate the dehy-
drogenase and restore its activity.
86
For this organism,
the decrease in activity forces substrate into the glyoxy-
late pathway (Section J,4) instead of the citric acid cycle.
Active pyruvate
dehydrogenase
E1
Inactive form
P-E1
ATP
ADP
P
i
H
2
O
Phosphatase, Mg
++
Kinase
activator
protein
Acetyl-CoA
NADH
+
+
(17-9)
C. The Citric Acid Cycle
Ch 17IND page - E - 2/14/03, 11:16 AM 953
954 Chapter 17. The Organization of Metabolism
BOX 17-C USE OF ISOTOPIC TRACERS IN STUDY OF THE TRICARBOXYLIC ACID CYCLE
The first use of isotopic labeling in the study of
the citric acid cycle and one of the first in the history
of biochemistry was carried out by Harland G. Wood
and C. H. Werkman in 1941.
a,b
The aim was to study
the fermentation of glycerol by propionic acid bac-
teria, a process that was not obviously related to the
citric acid cycle. Some succinate was also formed in
Glycerol propionate

+ H
+
+ H
2
O
G = 69 kJ/mol
the fermentation, and on the basis of simple mea-
surements of the fermentation balance reported in
1938 it was suggested that CO
2
was incorporated
into oxaloacetate, which was then reduced to succi-
nate. As we now know, this is indeed an essential
step in the propionic acid fermentation (Section F,3).
At the time
14
C was not available but the mass spec-
trometer, newly developed by A. O. Nier, permitted
the use of the stable
13
C as a tracer. Wood and
Werkman constructed a thermal diffusion column
and used it to prepare bicarbonate enriched in
13
C
and also built a mass spectrometer. By 1941 it was
established unequivocally that carbon dioxide was
incorporated into succinate by the bacteria.
c
To test the idea that animal tissues could also
incorporate CO
2
into succinate Wood examined
the metabolism of a pigeon liver preparation to
which malonate had been added to block succinate
dehydrogenase (Box 10-B). Surprisingly, the accu-
mulating succinate, which arose from oxalocetate
via citrate, isocitrate, and 2-oxoglutarate (traced
by green arrows in accompanying scheme), con-
tained no
13
C. Soon, however, it was shown that
CO
2
was incorporated into the carboxyl group of 2-
oxoglutarate that is adjacent to the carbonyl group.
That carboxyl is lost in conversion to succinate (Fig.
10-6) explains the lack of
13
C in succinate. It is of
historical interest that these observations were incor-
rectly interpreted by many of the biochemists of the
time. They agreed that citrate could not be a member of
the tricarboxylic acid cycle. Since citrate is a symmet-
ric compound it was thought that any
13
C incorpo-
rated into citrate would be present in equal amounts
in both terminal carboxyl groups. This would neces-
sarily result in incorporation of
13
C into succinate.
It was not until 1948 that Ogston popularized the
concept that by binding with substrates at three
points, enzymes were capable of asymmetric attack
upon symmetric substrates.
d
In other words, an
enzyme could synthesize citrate with the carbon
atoms from acetyl-CoA occupying one of the two
CH
2
COOH groups surrounding the prochiral
center. Later, the complete stereochemistry of the
citric acid cycle was elucidated through the use of a
variety of isotopic labels (see p. 704). Some of the
results are indicated by the asterisks and daggers in
the structures in Fig. 10-6.
The operation of the citric acid cycle in living
cells, organs, and whole animals has also been
observed using NMR and mass spectroscopy with
13
C-containing compounds. For example, a heart
can be perfused with a suitable oxygenated per-
fusion fluid
e
containing various
13
C-enriched sub-
strates such as [U-
13
C]fatty acids, [2-
13
C]acetate,
[3-
13
C]L-lactate, or [2,3-
13
C]propionate.
e k
NMR
spectroscopy allows direct and repeated observation
of the
13
C nuclei from a given substrate and its entry
into a variety of metabolic pathways. Because of the
high dispersion of chemical shift values for
13
C the
NMR resonance for the isotope can be seen at each
position within a single compound.
A compound that is especially easy to observe is
glutamate. This amino acid, most of which is found
in the cytoplasm, is nevertheless in relatively rapid
equilibrium with 2-oxoglutarate of the citric acid
cycle in the mitochondria. The accompanying
scheme shows where isotopic carbon from certain
compounds will be located when it first enters the
citric acid cycle and traces some of the labels into
glutamate. For example, uniformly enriched fatty
acids will introduce label into the two atoms of the
pro-S arm of citrate and into 4- and 5-positions of
glutamate whereas [2-
13
C]acetate will introduce
label only into the C4 position as marked by in the
scheme. In the NMR spectrum a singlet resonance
at 32.4 ppm will be observed. However, as succes-
sive turns of the citric acid cycle occur the isotope
will appear in increasing amounts in the adjacent
3-position of glutamate. They will be recognized
readily by the appearance of a multiplet. The initial
singlet will be flanked by a pair of peaks that arise
from spinspin coupling with the adjacent 3-
13
C of
the [2,3-
13
C]isotopomer (see accompanying figure).
After longer periods of time the central resonance
will weaken and the outer pair strengthen as the
recycling occurs.
Metabolism with [U-
13
C]fatty acids gives a
labeling pattern similar to that with [2,3-
13
C]acetate
and it has been deduced that heart muscle normally
metabolizes principally fatty acids for energy.
What will happen to the glutamate C4 resonance
if [3-
13
C]lactate is added to the perfusion solution?
It will enter both acetyl-CoA and oxaloacetate as
indicated by in the following scheme. That will
also intro-duce
13
C at C3 of glutamate. By looking at
spectra at short times the relative amounts of lactate
being oxidized via the cycle and that being converted
Ch 17IND page - E - 2/14/03, 11:16 AM 954
955
BOX 17-C (continued)
biosynthetically (anaplerotically) to glutamate can be
estimated. There is a complication that has long been
recognized. Oxaloacetate can be converted by
exchange processes to succinate. Since succinate is
symmetric the effect is to put 50% of the label into
each of the central atoms of succinate ( in scheme).
The exchange will then transfer label back into the
C2 position of oxaloacetate ( ) and through citric
acid cycle reactions into C3 of glutamate. Now the
C4 NMR resonance will contain an additional pair of
peaks arising from spinspin coupling with C2 but
which will have a different coupling constant than
that for coupling to C3.
If uniformly labeled [U-
13
C]acetate is introduced
the additional isotopomers, [3,4 -
13
C]glutamate and
[3,4,5 -
13
C]glutamate, will be formed as will others
with
13
C in the C1 and C5 positions but which will
not affect the C4 resonance. A total of nine lines will
be seen as illustrated in curve a of the accompanying
figure. We see that the multiplet patterns arising
from mass isotopomers are complex, but they can
be predicted accurately with a computer program.
f
Isotopomers of succinate have also been analyzed.
g
It is also of interest to introduce
13
C from
propionate labeled in various positions. One of
these is illustrated in the scheme. In this case the
appearance of multiplets arising from [3,4-
13
C]
glutamate verifies the existence of end-to-end
scrambling of the isotope in succinate. However,
is the scrambling complete or are some molecules
Glucose
Pyruvate
Lactate
Fatty acids
Acetyl-CoA
Citrate
Isocitrate
2-Oxoglutarate
Irreversible step
CO
2
Succinyl-CoA
Mutase (COSCoA moves)
Propionyl-CoA
Propionate
HCO
3

CO
2

OOC C
CH
2
O
COO

3 4
2
1
H
3
C C
O
S CoA
Malate
Succinate

OOC CH
2
CH
2
COO

C
HO

OOC
CH
2
CH
2
COO

COO

C
H
3
C CH C S CoA
O
COO

CO
2
HCO
3

H
3
C CH
2
COO

H
3
C CHOH COO

Acetate
H
3
C COO

OOC CH
2
CH
2
C S CoA
O
Glutamate

OOC CH
2
CH
2
CH COO

NH
3
+
5 4 3 2 1
C
C. The Citric Acid Cycle
Ch 17IND page - E - 2/14/03, 11:16 AM 955
956 Chapter 17. The Organization of Metabolism
A.
13
C-NMR spectrum of extracts of Langendorff-perfused
rat hearts perfused for 5 min with [1,2
13
C] acetate, [3
13
C]-
lactate and glucose. Only the glutamate C4 (left) and C3
(right) resonances are shown. B. Spectrum after perfusion
for 30 min. From Malloy et al.
r
C. The glutamate C4
resonance of an intact Langendorff-perfused rat heart
supplied with 2 mM [2-
13
C]acetate showing evolution of
the multiplet as a function of time after introducing the
label. The right panel shows glutamate C4 resonances
generated by a computer simulation after turnover of
citric acid cycle pools the indicated number of time. From
Jeffrey et al.
f
BOX 17-C USE OF ISOTOPIC TRACERS IN STUDY OF THE TRICARBOXYLIC ACID CYCLE (cont.)
efficiently channeled through enzymeenzyme
complexes in such a way as to avoid scrambling?
As shown in the scheme, full scrambling would give
equal labeling of C2 and C3 of oxaloacetate and of
glutamate. Experimentally greater labeling was
seen at C3 than at C2 during the first few turns of
a
Wood, H. G. (1972) in The Molecular Basis of Biological Transport
(Woessner, J. F., and Huijing, F., eds), pp. 154, Academic Press,
New York
b
Krampitz, L. O. (1988) Trends Biochem. Sci. 13, 152155
c
Wood, H. G., Werkman, C. H., Hemingway, A., and Nier, A. O.
(1941) J. Biol. Chem. 139, 377381
d
Ogston, A. G. (1948) Nature (London) 162, 936
e
Sherry, A. D., Sumegi, B., Miller, B., Cottam, G. L., Gavva, S.,
Jones, J. G., and Malloy, C. R. (1994) Biochemistry 33, 62686275
f
Jeffrey, F. M. H., Rajagopal, A., Malloy, C. R., and Sherry, A. D.
(1991) Trends Biochem. Sci. 16, 510
g
Jones, J. G., Sherry, A. D., Jeffrey, F. M. H., Storey, C. J., and
Malloy, C. R. (1993) Biochemistry 32, 1224012244
h
Des Rosiers, C., Di Donato, L., Comte, B., Laplante, A.,
Marcoux, C., David, F., Fernandez, C. A., and Brunengraber, H.
(1995) J. Biol. Chem. 270, 1002710036
i
Sherry, A. D., and Malloy, C. R. (1996) Cell Biochem. Funct. 14,
259268
j
Yudkoff, M., Nelson, D., Daikhin, Y., and Erecinska, M. (1994)
J. Biol. Chem. 269, 2741427420
k
Beylot, M., Soloviev, M. V., David, F., Landau, B. R., and
Brunengraber, H. (1995) J. Biol. Chem. 270, 15091514
l
Di Donato, L., Des Rosiers, C., Montgomery, J. A., David, F.,
Garneau, M., and Brunengraber, H. (1993) J. Biol. Chem. 268,
41704180
m
Magnusson, I., Schumann, W. C., Bartsch, G. E., Chandramouli,
V., Kumaran, K., Wahren, J., and Landau, B. R. (1991) J. Biol.
Chem. 266, 69756984
n
Chervitz, S. A., and Falke, J. J. (1995) J. Biol. Chem. 270,
2404324053
o
Hyder, F., Chase, J. R., Behar, K. L., Mason, G. F., Siddeek, M.,
Rothman, D. L., and Shulman, R. G. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 76127617
p
Cerdan, S., Knnecke, B., and Seelig, J. (1990) J. Biol. Chem. 265,
1291612926
q
Rothman, D. L., Novotny, E. J., Shulman, G. I., Howseman, A.
M., Petroff, O. A. C., Mason, G., Nixon, T., Hanstock, C. C.,
Prichard, J. W., and Shulman, R. G. (1992) Proc. Natl. Acad. Sci.
U.S.A. 89, 96039606
r
Malloy, C. R., Thompson, J. R., Jeffery, F. M. H., and Sherry, A.
D. (1990) Biochemistry 29, 67566761
the cycle suggesting that some channeling does
occur.
e
Isotopomer analysis can also be conducted by
mass spectroscopy, which is more sensitive than
NMR, using
13
C
h,k,l
or
2
H labeling.
j
Making use
of a technique like that employed by Knoop (Box
10-A), a chemical biopsy can be performed on
animals or on human beings, who may ingest gram
quantities of sodium phenylacetate without harm.
The phenylacetate is converted to an amide with
glutamine (phenylacetylglutamine) which is
excreted in the urine, from which it can easily be
recovered for analysis.
l n
This provides a non-
invasive way of studying the operation of the citric
acid cycle in the human body. Direct measurement
on animal brains
o,p
and on human limbs or brain
has also been accomplished by NMR spectroscopy
q
and may become more routine as instrumentation
is improved.
A
B
C4
C3
34.5 34.0 28.0 27.5
Chemical shift, ppm Chemical shift, ppm
C4 C3
Glutamate C4 resonance Simulated spectra
30 min
25 min
20 min
15 min
10 min
5 min
7
6
5
4
3
2 No. of turns
C
Ch 17IND page - E - 2/14/03, 11:16 AM 956
957 C. The Citric Acid Cycle
BOX 17-D FLUOROACETATE AND LETHAL SYNTHESIS
Among the most deadly of simple compounds
is sodium fluoroacetate. The LD
50
(the dose lethal
for 50% of animals receiving it) is only 0.2 mg/kg
for rats, over tenfold less than that of the nerve poi-
son diisopropylphosphofluoridate (Chapter 12).
a,b
Popular, but controversial, as the rodent poison
1080, fluoroacetate is also found in the leaves
of several poisonous plants in Africa, Australia,
and South America. Surprisingly, difluoroacetate
HCF
2
COO

is nontoxic and biochemical studies


reveal that monofluoroacetate has no toxic effect on
cells until it is converted metabolically in a lethal
synthesis to 2R,3R-2-fluorocitrate, which is a com-
petitive inhibitor of aconitase (aconitate hydratase,
Eq. 13-17).
bg
This fact was difficult to understand
since citrate formed by the reaction of fluorooxalo-
acetate and acetyl-CoA has only weak inhibitory
activity toward the same enzyme. Yet, it is the
fluorocitrate formed from fluorooxaloacetate that
contains a fluorine atom at a site that is attacked by
aconitase in the citric acid cycle.
The small van der Waals radius of fluorine
(0.135 nm), comparable to that of hydrogen (0.12 nm),
is often cited as the basis for the ability of fluoro
compounds to deceive enzymes. However, the
high electronegativity and ability to enter into hydro-
gen bonds may make F more comparable to OH in
metabolic effects. In the case of fluorocitrate it was
proposed that the inhibitory isomer binds in the
wrong way to aconitase in such a manner that the
fluorine atom is coordinated with the ferric ion at
the catalytic center.
c
However, 2R,3R-2-fluorocitrate
is a simple competitive inhibitor of aconitase but an
irreversible poison. It is especially toxic to nerves
and also appears to affect mitochondrial membranes.
Therefore, this poison may affect some other target,
such as a citrate transporter.
d
Fluoroacetate is only
one of many known naturally occurring fluorine
compounds.
c
Another example of lethal synthesis is seen in
the use of 5-fluorouracil in cancer therapy (Box 15-E).
In this compound and in many other fluorine-
containing inhibitors the F atom replaces the H atom
that is normally removed as H
+
in the enzymatic
reaction. The corresponding F
+
cannot be formed.
h
Because of the high electronegativity of fluorine a
C F bond is polarized: C
+
F

. This may have
very large effects on reactivity at adjacent positions.
For example, the reactivity of 2-fluoroglycosyl
groups toward glycosyl transfer is decreased by
several orders of magnitude (p. 597).
a
Gibble, G. W. (1973) J. Chem. Educ. 50, 460462
b
Elliott, K., and Birch, J., eds. (1972) CarbonFluorine Compounds,
Elsevier, Amsterdam
c
Glusker, J. P. (1971) in The Enzymes, 3rd ed., Vol. 5 (Boyer, P. D.,
ed), pp. 413439, Academic Press, New York
d
Kun, E. (1976) in Biochemistry Involving CarbonFluorine Bonds
(Filler, R., ed), pp. 122, American Chemical Society, Washing-
ton, DC
e
Marletta, M. A., Srere, P. A., and Walsh, C. (1981) Biochemistry
20, 37193723
f
Rokita, S. E., and Walsh, C. T. (1983) Biochemistry 22, 28212828
g
Peters, R. A. (1957) Adv. Enzymol. 18, 113159
h
Abeles, R. H., and Alston, T. A. (1990) J. Biol. Chem. 265,
1670516708
Acting to counteract any drop in ATP level, accumu-
lating ADP acts as a positive effector for isocitrate
dehydrogenases.
Another way in which the phosphorylation state
of the adenylate system can regulate the cycle depends
upon the need for GDP in step f of the cycle (Fig. 17-4).
Within mitochondria, GTP is used largely to reconvert
AMP to ADP. Consequently, formation of GDP is pro-
moted by AMP, a compound that arises in mitochon-
dria from the utilization of ATP for activation of fatty
acids (Eq. 13-44) and activation of amino acids for
protein synthesis (Eq. 17-36).
In E. coli and some other bacteria ATP does not
inhibit citrate synthase but NADH does; the control is
via the redox potential of the NAD
+
system rather
than by the level of phosphorylation of the adenine
nucleotide system.
95
Succinic dehydrogenase may be
regulated by the redox state of ubiquinone (Chapter 15).
Another mechanism of regulation may be the forma-
tion of specific proteinprotein complexes between
enzymes catalyzing reactions of the cycle.
9697a
This
may permit one enzyme to efficiently have a product
of its action transferred to the enzyme catalyzing the
next step in the cycle.
5. Catabolism of Intermediates of the Citric
Acid Cycle
Acetyl-CoA is the only substrate that can be com-
pletely oxidized to CO
2
by the reaction of the citric
acid cycle alone. Nevertheless, cells must sometimes
C
C
CH
2
COO

OH
COO

H
F
C C
H
F
H
O
O

2
3

OOC
2R,3R 2-Fluoroacetate
Ch 17IND page - E - 2/14/03, 11:16 AM 957
958 Chapter 17. The Organization of Metabolism
oxidize large amounts of one of the compounds found
in the citric acid cycle to CO
2
.
98,99
For example, bacteria
subsisting on succinate as a carbon source must oxidize
it for energy as well as convert some of it to carbo-
hydrates, lipids, and other materials. Complete com-
bustion of any citric acid cycle intermediate can be
accomplished by conversion to malate followed by
oxidation of malate to oxaloacetate (Eq. 17-10, step a)
and decarboxylation ( cleavage) to pyruvate, or (Eq.
17-10, step b) oxidation and decarboxylation of malate
by the malic enzyme (Eq. 13-45) without free oxalo-
acetate as an intermediate. Pathway b is probably the
most important. It is catalyzed by two different malic
enzymes present in animal mitochondria. One is
specific for NADP
+
while the other reacts with NAD
+
as well.
100,101
They both have complex regulatory pro-
perties. For example, the less specific NAD
+
-utilizing
enzyme is allosterically inhibited by ATP but is activated
by fumarate, succinate, or isocitrate.
100
Thus, accumu-
lation of citric acid cycle intermediates turns on the
malic enzyme, allowing the excess to leave the cycle
and reenter as acetyl groups. Since the Michaelis
constant for malate is high, this will not happen unless
malate accumulates, signaling a need for acetyl-CoA.
The NADP
+
-dependent enzyme is activated by a high
concentration of free CoA and is inhibited by NADH.
Perhaps when glycolysis becomes slow the free CoA
level rises and turns on malate oxidation.
101
On the
other hand, rapid glycolysis increases the NADH
concentration which inhibits the malic enzyme. The
result is a buildup of the oxaloacetate concentration
and an increase in activity of the citric acid cycle. The
malic enzymes are also present in the cytoplasm,
where one of them functions as part of an NADPH-
generating cycle (Eq. 17-46).
D. Oxidative Pathways Related to the Citric
Acid Cycle
In this section we will consider some other catalytic
cycles as well as some noncyclic pathways of oxidation
of one- and two-carbon substrates that are utilized by
microorganisms.
1. The -Aminobutyrate Cycle
A modification of the citric acid cycle which
involves glutamate and gamma () aminobutyrate
(GABA) has an important function in the brain (Fig.
17-5). Both glutamate and -aminobutyrate occur
in high concentrations in brain (10 and 0.8 mM, re-
spectively). Both are important neurotransmitters,
-aminobutyrate being a principal neuronal inhibitory
substance
102,103
(Chapter 30). In the -aminobutyrate
cycle acetyl-CoA and oxaloacetate are converted into
citrate (step a) in the usual way and the citrate is then
converted into 2-oxoglutarate. The latter is transformed
to L-glutamate either by direct amination (b) or by trans-
amination (c), the amino donor being -aminobutyrate.
-Aminobutyrate is formed by decarboxylation of
glutamate (Fig. 17-5, step d)
104
and is catabolized via
transamination (step e)
105
to succinic semialdehyde,
which is oxidized to succinate
106
and oxaloacetate.
The two transamination steps in the pathways may be
linked, as indicated in Fig. 17-5, to form
a complete cycle that parallels the citric
acid cycle but in which 2-oxoglutarate is
oxidized to succinate via glutamate and
-aminobutyrate. No thiamin diphosphate
is required, but 2-oxoglutarate is reductively
aminated to glutamate. The cycle is some-
times called the -aminobutyrate shunt,
and it plays a significant role in the over-
all oxidative processes of brain tissue.
This pathway is also prominent in green
plants.
107109
For example, under anaerobic
conditions the radish Raphanus sativus ac-
cumulates large amounts of -aminobutyr-
ate.
110
Most animal tissues contain very
little -aminobutyrate, although it has been
found in the oviducts of rats at concentra-
tions that exceed those in the brain.
111
2. The Dicarboxylic Acid Cycle
Some bacteria can subsist solely on
glycolate, glycine, or oxalate, all of which

OOC C
O
CH
2
COO

OOC C
O
CH
2
P
l-Malate
a
b
c
NAD
+
NADH
Malate
dehydrogenase
NAD
+
or
NADP
+
NADH
or
NADPH
CO
2
Malic enzyme

OOC C
O
CH
3
Pyruvate
Acetyl-CoA
Oxidative
decarboxylation
CO
2
Oxaloacetate
decarboxylase
(also spontaneous)
Pyruvate
kinase
GTP
GDP
PEPCK (Eq 13-46)
ATP ADP
Oxaloacetate
Phosphoenolpyruvate

OOC
H
CH
2
OH
COO

(17-10)
Ch 17IND page - E - 2/14/03, 11:16 AM 958
959 D. Oxidative Pathways Related to the Citric Acid Cycle
are converted to glyoxylate (Eq. 17-11).
Glyoxylate is oxidized to CO
2
and
water to provide energy to the
bacteria and is also utilized for
biosynthetic purposes. The energy-
yielding process is found in the
dicarboxylic acid cycle (Fig. 17-6),
which catalyzes the complete oxida-
tion of glyoxylate. Four hydrogen
atoms are removed with generation
of two molecules of NADH which
can be oxidized by the respiratory
chain to provide energy.
112,113
In the
dicarboxylic acid cycle glyoxylate is
the principal substrate and acetyl-
CoA is the regenerating substrate
rather than the principal substrate
as it is for the citric acid cycle.
The logic of the dicarboxylic
acid cycle is simple. Acetyl-CoA
contains a potentially free carboxyl
group. After the acetyl group of
acetyl-CoA has been condensed
with glyoxylate and the resulting
hydroxyl group has been oxidized,
the free carboxyl group appears in
oxaloacetate in a position to the
carbonyl group. The carboxyl
Transamination
O C COO

CH
2
CH
2
COO

C H
2
C
O CH
2
COO

H
2-Oxoglutarate
NAD
+

OOC H
2
C
CH
2
COO

Succinate
Oxaloacetate
C
CH
2
COO

H
2
C
COO

H
H
3
+
N
l-Glutamate
CH
2
CH
2
H
2
C
COO

H
3
+
N
CO
2
Glutamate
decarboxylase
(Chapter 14,
Section E)
PLP
-Aminobutyrate
NADPH
NH
4
+
d
e
b
c
Acetyl-CoA
Succinic semialdehyde
a
NAD
+
H
2
O
NAD
+
CO
2
FAD

Figure 17-5 Reactions of the -aminobutyrate (GABA) cycle.


Figure 17-6 The dicarboxylic acid cycle for oxidation of glyoxylate to
carbon dioxide. The pathway for synthesis of the regenerating substrate
is indicated by green lines. This pathway is also needed for synthesis of
carbohydrates and all other cell constituents.
O C COO

CH
3
C
CHO
COO

H
HO
C
CH
2
OH
COO

H
HO
O
C COO

H
Glyoxylate
CH
3
C
O
S CoA
Acetyl-CoA
Malate
synthase
Glyoxylate carboligase
CO
2
d-Glycerate
HO C COO

CH
2
COO

H
Malate
O C COO

CH
2
COO

Oxaloacetate
C
CH
2
OH
COO

H
O P
2-Phosphoglycerate
C
CH
2
COO

H
HO
O P
3-Phosphoglycerate
Cleavage
ATP
+2[H]
Tartronic semialdehyde
2[H]
Oxidative
decarboxylation
Cleavage
Carbohydrate synthesis
Synthesis of regenerating substrate
(2 mol)
CO
2
CO
2
Ch 17IND page - E - 2/14/03, 11:16 AM 959
960 Chapter 17. The Organization of Metabolism
donated by the glyoxylate is still in the position.
A consecutive cleavage and an oxidative cleavage
release the two carboxyl groups as carbon dioxide to
reform the regenerating substrate. The cycle is simple
and efficient. Like the citric acid cycle, it depends upon
thiamin diphosphate, without which the cleavage
would be impossible. Comparing the citric acid cycle
(Fig. 17-2) with the simpler dicarboxylic acid cycle, we
see that in the former the initial condensation product
citrate contains a hydroxyl group attached to a tertiary
carbon atom. With no adjacent hydrogen it is impossible
to oxidize it directly to the carbonyl group which is
essential for subsequent chain cleavage; hence the
dependence on aconitase to shift the OH to an adjacent
carbon. Both cycles involve oxidation of a hydroxy
acid to a ketone followed by cleavage and oxidative
cleavage. In the citric acid cycle additional oxidation
steps are needed to convert succinate back to oxalo-
acetate, corresponding to the fact that the citric acid
cycle deals with a more reduced substrate than does
the dicarboxylic acid cycle.
The synthetic pathway for the regenerating sub-
strate of the dicarboxylic acid cycle is quite complex.
Two molecules of glyoxylate undergo condensation
with decarboxylation by glyoxylate carboligase
114
(see also Chapter 14, Section D) to form tartronic
semialdehyde. The latter is reduced to D-glycerate,
which is phosphorylated to 3-phosphoglycerate and
2-phosphoglycerate. Since the phosphoglycerates are
carbohydrate precursors, this glycerate pathway
provides the organisms with a means for synthesis
of carbohydrates and other complex materials from
glyoxylate alone. At the same time, 2-phosphoglycerate
can be converted to pyruvate and the pyruvate, by
oxidative decarboxylation, to the regenerating substrate
acetyl-CoA.
E. Catabolism of Sugars
In most sugars each carbon atom bears an oxygen
atom which facilitates chemical attack by oxidation at
any point in the carbon chain. Every sugar contains a
potentially free aldehyde or ketone group, and the
carbonyl function can be moved readily to adjacent
positions by isomerases. Consequently, aldol cleavage
is also possible at many points. For these reasons, the
metabolism of carbohydrates is complex and varied.
A sugar chain can be cut in several places giving rise
to a variety of metabolic pathways. However, in the
energy economy of most organisms, including human
beings, the EmbdenMeyerhofParnas or glycolysis
pathway by which hexoses are converted to pyruvate
(Fig. 17-7) stands out above all others. We have already
considered this pathway, which is also outlined in
Figs. 10-2 and 10-3. Some history and additional
important details follow.
1. The Glycolysis Pathway
The discovery of glycolysis followed directly the
early observations of Buchner and of Harden and
Young on fermentation of sugar by yeast juice (p. 767).
Another line of research, the study of muscle, soon
converged with the investigations of alcoholic fermen-
tation. Physiologists were interested in the process
by which an isolated muscle could obtain energy for
contraction in the absence of oxygen. It was shown
by A. V. Hill that glycogen was converted to lactate
to supply the energy, and Otto Meyerhof later demon-
strated that the chemical reactions were related to
those of alcoholic fermentation. The establishment of
the structures and functions of the pyridine nucleo-
tides in 1934 (Chapter 15) coincided with important
studies by G. Embden in Frankfurt and of J. K. Parnas
in Poland. The sequence of reactions in glycolysis
soon became clear. All of the 15 enzymes catalyzing
the individual steps in the sequence have been isolated
and crystallized and are being studied in detail.
115
Formation of pyruvate. The conversion of glu-
cose to pyruvate requires ten enzymes (Fig. 17-7), and
the sequence can be divided into four stages: prepara-
tion for chain cleavage (reactions 13), cleavage and
equilibration of triose phosphates (reactions 4 and 5),
oxidative generation of ATP (reactions 6 and 7), and
conversion of 3-phosphoglycerate to pyruvate (reac-
tions 810).
In preparation for chain cleavage, free glucose is
phosphorylated to glucose 6-phosphate by ATP under
the action of hexokinase (reaction 1). Glucose 6-
phosphate can also arise by cleavage of a glucosyl unit
from glycogen by the consecutive action of glycogen
phosphorylase (reaction 1a) and phosphoglucomutase,
which transfers a phospho group from the oxygen at
C-1 to that at C-6 (reaction 1b) (see also Eq. 12-39 and
associated discussion of the mechanism of this enzyme).
Why do cells attach phospho groups to sugars to initiate
metabolism of the sugars? Four reasons can be given:
(a) The phospho group constitutes an electrically
charged handle for binding the sugar phosphate
to enzymes.
(b) There is a kinetic advantage in initiating a reac-
tion sequence with a highly irreversible reaction
HO CH
2
COO
H
3
+
N CH
2
COO
HO CH
2
COO
Glycolate
2[H]
C COO
H
O
2[H]
Transamination
Glycine
Oxalate
Glyoxylate
Oxalyl-CoA

(17-11)
Ch 17IND page - E - 2/14/03, 11:16 AM 960
961 E. Catabolism of Sugars
Figure 17-7 Outline of the glycolysis pathway by which hexoses are broken down to pyruvate. The ten enzymes needed to
convert D-glucose to pyruvate are numbered. The pathway from glycogen using glycogen phosphorylase is also included, as is
the reduction of pyruvate to lactate (step 11). Steps 6a7, which are involved in ATP synthesis via thioester and acyl phosphate
intermediates, are emphasized. See also Figures 10-2 and 10-3, which contain some additional information.
O
O
CH
2
OP
OH
HO
OH
P
CH
2
OP
C
C
C
O
H
HO
HO
C
CH
2
OP
OH

H
H
C
C
CH
2
OP
H OH
H O
C
C
CH
2
OP
H OH
OH H
S
E
C
C
CH
2
OP
H OH
O
S
E
Fructose 6-P
ATP
ADP
3
Fructose 1,6-P
2
4
Dihydroxyacetone-P
Glyceraldehyde 3-P
5
E-SH
Thioester intermediate
NAD
+
NADH + H
+
Reoxidation to NAD
+
in mitochondria
8
HO
O
HO
CH
2
OH
OH
OH
1
-d-Glucose
C
6
H
12
O
6
Glucose 6-P
1b
2
Glucose 1-P
P
i
Glycogen
1a
Equations below are balanced for one triose phosphate or 1/2 glucose
C
C
CH
2
OP
H OH
O O P
P
i
E-SH
Acyl phosphate intermediate
l-Lactic acid C
3
H
6
O
3
ADP ATP
7
C
CH
2
OP
COO

HO
H
3-Phosphoglycerate
C
CH
2
OH
H
COO

OP
COO

C
CH
2
OP
COO

C
CH
3
O
9
P
i
ATP ADP
(Resynthesis of ATP
compensates for that
used in steps 1 and 3)
Pyruvate
C
3
H
3
O
3

10
11
CO
2
Acetyl-CoA to
citric acid cycle
6a
6b
6c
C
CH
3
COOH
H
HO
ATP ADP
1
UTP
Ch 17IND page - E - 2/14/03, 11:16 AM 961
962 Chapter 17. The Organization of Metabolism
such as the phosphorylation of glucose.
(c) Phosphate esters are unable to diffuse out of cells
easily and be lost.
(d) There is at least a possibility that the phospho
groups may function in catalysis.
Reaction 2 of Fig. 17-7 is a simple isomerization
that moves the carbonyl group to C-2 so that cleav-
age to two three-carbon fragments can occur. Before
cleavage a second phosphorylation (reaction 3) takes
place to form fructose 1,6-bisphosphate. This ensures
that when fructose bisphosphate is cleaved by aldolase
each of the two halves will have a phosphate handle.
This second priming reaction (reaction 3) is the first
step in the series that is unique to glycolysis. The
catalyst for the reaction, phosphofructokinase, is
carefully controlled, as discussed in Chapter 11 (see
Fig. 11-2).
Fructose bisphosphate is cleaved by action of an
aldolase (reaction 4) to give glyceraldehyde 3-phosphate
and dihydroxyacetone phosphate. These two triose
phosphates are then equilibrated by triose phosphate
isomerase (reaction 5; see also Chapter 13). As a result,
both halves of the hexose can be metabolized further
via glyceraldehyde 3-P to pyruvate. The oxidation of
glyceraldehyde 3-P to the corresponding carboxylic
acid, 3-phosphoglyceric acid (Fig. 17-7, reactions 6 and
7), is coupled to synthesis of a molecule of ATP from
ADP and P
i
. This means that two molecules of ATP
are formed per hexose cleaved, and that two molecules
of NAD
+
are converted to NADH in the process.
The conversion of 3-phosphoglycerate to pyru-
vate begins with transfer of a phospho group from the
C-3 to the C-2 oxygen (reaction 8) and is followed by
dehydration through an , elimination catalyzed by
enolase (reaction 9). The product, phosphoenolpyru-
vate (PEP), has a high group transfer potential. Its
phospho group can be transferred easily to ADP via
the action of the enzyme pyruvate kinase, to leave
the enol of pyruvic acid which is spontaneously con-
verted to the much more stable pyruvate ion (see Eq.
7-59). Because two molecules of PEP are formed from
each glucose molecule, the process provides for the
recovery of the two molecules of ATP that were expended
in the initial formation of fructose 1,6-bisphosphate
from glucose. Several isoenzyme forms exist in mam-
mals. Most of these are allosterically activated by
fructose 1,6-bisphosphate.
115a,b
However, the enzyme
from trypanosomes is activated by fructose 2,6-P
2
.
115c
The further metabolism of pyruvate. In the
aerobic metabolism that is characteristic of most tissues
of our bodies, pyruvate is oxidatively decarboxylated
to acetyl-CoA, which can then be completely oxidized
in the citric acid cycle (Fig. 17-4). The NADH produced
in reaction 6 of Fig. 17-7, as well as in the oxidative
decarboxylation of pyruvate and in subsequent reac-
tions of the citric acid cycle, is reoxidized in the electron
transport chain of the mitochondria as described in
detail in Chapter 18 (see Fig. 18-5). An important
alternative fate of pyruvate is to enter into fermentation
reactions. For example, the enzyme lactate dehydro-
genase (Fig. 17-7, reaction 11) catalyzes reduction by
NADH of pyruvate to L-lactate, or, for some bacteria,
to D-lactate. This reaction can be coupled to the NADH-
producing reaction 6 to give a balanced process by which
glucose is fermented to lactic acid in the absence of
oxygen (see also Eq. 10-3). In a similar process, yeast
cells decarboxylate pyruvate ( cleavage) to acetalde-
hyde which is reduced to ethanol using the NADH
produced in reaction 6. These fermentation reactions
are summarized in Fig. 10-3 and, along with many others,
are discussed further in Section F of this chapter.
2. Generation of ATP by Substrate Oxidation
The formation of ATP from ADP and P
i
is a vital
process for all cells. It is usually referred to as phos-
phorylation and includes oxidative phosphoryla-
tion associated with the passage of electrons through
an electron transport chain usually in mitochondria;
photosynthetic phosphorylation, a similar process
occurring in chloroplasts under the influence of light;
and substrate-level phosphorylation. Only the last
is fully understood chemically. The dehydrogenation
of glyceraldehyde 3-P and the accompanying ATP
formation (reactions 6 and 7, Fig. 17-7; Fig. 15-6) is the
best known example of substrate-level phosphoryla-
tion and is tremendously important for yeasts and
other microorganisms that live anaerobically. They
depend upon this one reaction for their entire supply
of energy. The conversion of glucose either to lactate
or to ethanol and CO
2
is accompanied by a net synthesis
of only two molecules of ATP and it is most logical
to view these as arising from oxidation of glyceralde-
hyde 3-P. The formation of ATP from PEP and ADP
in reaction 10 of Fig. 17-7 can be regarded as the recap-
turing of ATP spent in the priming reactions of steps
1 and 3. With a gain of only two molecules of ATP for
each molecule of hexose fermented, it is not surprising
that yeast must ferment enormous amounts of sugar to
meet its energy needs.
Each glucose unit of glycogen stored in our bodies
can be converted to pyruvate with an apparent net gain
of three molecules of ATP. However, two molecules of
ATP were needed for the initial synthesis of each hexose
unit of glycogen (Fig. 12-2). Therefore, the overall net
yield for fermentation of stored polysaccharide is still
only two ATP per hexose. The fermentation of glycogen
accounts for the very rapid generation of lactic acid
during intense muscular activity. However, in most
circumstances within aerobic tissues reoxidation of
NADH occurs via the electron transport chain of mito-
chondria with a much higher yield of ATP. Substrate-
Ch 17IND page - E - 2/14/03, 11:16 AM 962
963
level phosphorylation can also follow oxidative decar-
boxylation of an -oxoacid. For example, in the citric
acid cycle GTP is formed following oxidative decar-
boxylation of 2-oxoglutarate (Fig. 17-4, steps e and f ).
3. The Pentose Phosphate Pathways
A second way of cleaving glucose 6-phosphate
utilizes sequences involving the five-carbon pentose
sugars. They are referred to as pentose phosphate
pathways, the phosphogluconate pathway, or the
hexose monophosphate shunt. Historically, the evidence
for such routes dates from the experiments of Warburg
on the oxidation of glucose 6-P to 6-phosphogluconate
(Chapter 15). For many years the oxidation remained
an enzymatic reaction without a defined pathway.
However, it was assumed to be part of an alternative
method of degradation of glucose. Supporting evidence
was found in the observation that tissues continue to
respire in the presence of a high concentration of
fluoride ion, a known inhibitor of the enolase reaction
and capable of almost completely blocking glycolysis.
Some tissues, e.g., liver, are especially active in respira-
tion through this alternative pathway, whose details
were elucidated by Horecker and associates.
116,117
We
now know that the pentose phosphate pathways are
multiple as well as multipurpose. They function in
catabolism and also, when operating in the reverse
direction, as a reductive pentose phosphate pathway
that lies at the heart of the sugar-forming reactions of
photosynthesis.
The oxidative pentose pathway provides a means
for cutting the chain of a sugar molecule one carbon at a
time, with the carbon removed appearing as CO
2
. The
enzymes required can be grouped into three distinct
systems, all of which are found in the cytosol of animal
cells: (i) a dehydrogenationdecarboxylation system,
(ii) an isomerizing system, and (iii) a sugar rearrange-
ment system. The dehydrogenationdecarboxylation
system cleaves glucose 6-P to CO
2
and the pentose
phosphate, ribulose 5-P (Eq. 17-12). Three enzymes are
required, the first being glucose 6-P dehydrogenase
117a
(Eq. 17-12, step a; see also Eq. 15-10). The immediate
product, a lactone, undergoes spontaneous hydrolysis.
However, the action of gluconolactonase (Eq. 17-12,
step b) causes a more rapid ring opening. A second
dehydrogenation is catalyzed by 6-phosphogluconate
dehydrogenase (Eq. 17-12, step c),
117b
and this reaction
is immediately followed by a decarboxylation cata-
lyzed by the same enzyme (as in Eq. 13-45). The value
of G for oxidation of glucose 6-P to ribulose 5-P by
NADP
+
according to Eq. 17-12 is 30.8 kJ mol
1
, a
negative enough value to drive the [NADPH]/[NADP
+
]
ratio to an equilibrium value of over 2000 at a CO
2
tension of 0.05 atm.
The isomerizing system, consisting of two enzymes,
HO
O
HO
OH
OH
OP
COO

C OH
C HO H
C OH
C OH
CH
2
OP
H
H
H
CH
2
OH
C O
C OH
C OH
CH
2
OP
H
H
b
a
Glucose 6-P
NADP
+
NADPH
6-Phosphogluconolactone
6-Phosphogluconate
NADP
+
NADPH
+ H
+
CO
2
c
Ribulose 5-P
Gluconolactonase
6-Phosphogluconate
dehydrogenase
Glucose-6-phosphate
dehydrogenase
COOH
C OH
C O
C OH
C OH
CH
2
OP
H
H
H
HO
O
HO
OH O
OP
(17-12)
(17-13)
E. Catabolism of Sugars
CH
2
OH
C O
C OH
C OH
CH
2
OP
CHO
C OH
C OH
C OH
CH
2
OP
CH
2
OH
C O
CH
C OH
CH
2
OP
HO
H
H
H
H
H
H
Ribose 5-P
Ribulose 5-P
Xylulose 5-P
Epimerase
Isomerase
(Chapter 13)
Ch 17IND page - E - 2/14/03, 11:16 AM 963
964 Chapter 17. The Organization of Metabolism
atoms are chopped off one at a time (Fig. 17-8A) leaving
a three-carbon triose phosphate as the product. Since
the dehydrogenation system works only on glucose
6-P, a part of the sugar rearrangement system must
be utilized between each of the three oxidation steps.
Notice that a C
5
unit (ribose 5-P) is used in the first
reaction with transketolase but is regenerated at the
end of the sequence. This C
5
unit is the regenerating
substrate for the cycle. As indicated by the dashed
arrows, it is formed readily in any quantity needed
by oxidation of glucose 6-P. Before the C
5
unit that
is formed in each oxidation step can be processed by
the sugar rearrangement reactions, it must be isomer-
ized
117c,118,118a,b
from ribulose 5-P to xylulose 5-P; before
the C
5
unit, produced at the end of the sequence in Fig.
17-8, can be reutilized as a regenerating substrate, it
must be isomerized to ribose 5-P. Thus, the cycle is
quite complex. The same C
5
substrates appear at
several points in Fig. 17-8A and substrates from differ-
ent parts of the cycle become scrambled and the path-
way does not degrade all the hexose molecules in a
uniform manner. For this reason, Zubay described the
pentose phosphate pathways as a swamp.
119
The oxidative pentose phosphate cycle is often
presented as a means for complete oxidation of hexoses
to CO
2
. For this to happen the C
3
unit indicated as the
product in Fig. 17-8A must be converted (through the
action of aldolase, a phosphatase, and hexose phosphate
isomerase) back to one-half of a molecule of glucose-6-P
which can enter the cycle at the beginning. On the
other hand, alternative ways of degrading the C
3
pro-
duct glyceraldehyde-P are available. For example, using
glycolytic enzymes, it can be oxidized to pyruvate and
to CO
2
via the citric acid cycle.
As a general rule, NAD
+
is associated with catabolic
reactions and it is somewhat unusual to find NADP
+
acting as an oxidant. However, in mammals the en-
zymes of the pentose phosphate pathway are specific
for NADP
+
. The reason is thought to lie in the need
of NADPH for biosynthesis (Section I). On this basis,
the occurrence of the pentose phosphate pathway in
tissues having an unusually active biosynthetic func-
tion (liver and mammary gland) is understandable.
In these tissues the cycle may operate as indicated in
Fig. 17-8A with the C
3
product also being used in
biosynthesis. Furthermore, any of the products from
C
4
to C
7
may be withdrawn in any desired amounts
without disrupting the smooth operation of the cycle.
For example, the C
4
intermediate erythrose 4-P is
required in synthesis of aromatic amino acids by bac-
teria and plants (but not in animals). Ribose 5-P is
needed for formation of several amino acids and of
nucleic acids by all organisms. In some circumstances
the formation of ribose 5-P may be the only essential
function for the pentose phosphate pathway.
120
Several studies of the metabolism of isotopically
labeled glucose
121122a
have been in accord with
interconverts three pentose phosphates (Eq. 17-13).
As a consequence the three compounds exist as an
equilibrium mixture. Both xylulose 5-P and ribose 5-P
are needed for further reactions in the pathways.
The ingenious sugar rearrangement system uses
two enzymes, transketolase and transaldolase.
Both catalyze chain cleavage and transfer reactions
(Eqs. 17-14 and 17-15) that involve the same group of
substrates. These enzymes use the two basic types of
CC bond cleavage, adjacent to a carbonyl group ()
and one carbon removed from a carbonyl group ().
Both types are needed in the pentose phosphate path-
ways just as they are in the citric acid cycle. The en-
zymes of the pentose phosphate pathway are found
in the cytoplasm of both animal and plant cells.
117c
Mammalian cells appear to have an additional set that
is active in the endoplasmic reticulum and plants have
another set in the chloroplasts.
117c
An oxidative pentose phosphate cycle. Putting
the three enzyme systems together, we can form a
cycle that oxidizes hexose phosphates. Three carbon
(17-14)
(17-15)
CH
2
OH
C O
C
R
CHO
R
C
R
CH
2
OH
C O
C
R
HO H
H O
Xylulose 5-P,
fructose 6-P,
or sedoheptulose 7-P
Ribose 5-P,
erythrose 4-P, or
glyceraldehyde 3-P
H HO
Transketolase (TK),
a thiamin diphosphate
enzyme (Chapter 14)
CH
2
OH
C O
C
C
CHO
R
CHO
R
CH
2
OH
C O
C
C
R
R
HO H
H OH
HO
H
H
OH
Transaldolase (TA)
(Chapter 13)
Ch 17IND page - E - 2/14/03, 11:16 AM 964
965
Figure 17-8 The pentose phosphate pathways. (A) Oxi-
dation of a hexose (C
6
) to three molecules of CO
2
and a
three-carbon fragment with the option of removing C
3
, C
4
,
C
5
, and C
7
units for biosynthesis (dashed arrows). (B) Non-
oxidative pentose pathways: 2
1
/2 C
6
3 C
5
or 2 C
6
3 C
4
or
3
1
/2 C
6
3 C
7
.
C
6
C
5
C
5
C
7
C
3
C
6
CO
2
CO
2
C
5
C
6
C
4
C
3
CO
2
C
5
Degradation
via pyruvate
or conversion to
C
6
and recycling
TA
TK
TK
2 NAD
+
2 NAD
+
2 NAD
+
C
5
TK
C
5
C
6
C
4
C
3
C
7
TA
C
3
C
6
C
5
TK
2
1
/2 C
6
2
1
/2 C
6
3 C
5
3 C
5
A B
operation of the pentose phosphate pathway as is
depicted in Fig. 17-8. However, Williams and associates
proposed a modification in the sugar rearrangement
sequence in liver
123126
to include the formation of
arabinose 5-P (from ribose 5-P), an octulose bisphos-
phate, and an octulose 8-monophosphate. Many
investigators argue that these additional reactions are
of minor significance.
121,122,127
The measured concen-
trations of pentose phosphate pathway intermediates
in rat livers are close to those predicated for a near-
equilibrium state from equilibrium constants measured
for the individual steps of Fig. 17-8.
128
Most of the
concentrations are in the 4- to 10-M range but the
level of erythrose 4-P, which is predicted to be ~ 0.2 M,
is too low to measure.
In contrast to animals, the resurrection plant
Craterostigma plantaginenm accumulates large amounts
of a 2-oxo-octulose. This plant is one of a small group
of angiosperms that can withstand severe dehydration
and are able to rehydrate and resume normal metabo-
lism within a few hours. During desiccation much of
the octulose is converted into sucrose. The plant has
extra transketolase genes which may be essential for
this rapid interconversion of sugars.
129
Nonoxidative pentose phosphate pathways.
The sugar rearrangement system together with the
glycolytic enzymes that convert glucose 6-P to glycer-
aldehyde 3-P can function to transform hexose phos-
phates into pentose phosphates (Fig. 17-8B; Eq. 17-16)
which may be utilized for nucleic acid synthesis in
erythrocytes and other cells.
130,131
2
1
/2 C
6
3 C
5
The reader can easily show that the same enzymes
will catalyze the net conversion of hexose phosphate
to erythrose 4-phosphate or to sedoheptulose 7-phos-
phate (Eq. 17-17):
2 C
6
3 C
4
; 3
1
/2 C
6
3 C
7
An investigation of metabolism of the red lipid-
forming yeast Rhodotorula gracilis (which lacks phos-
phofructokinase and is thus unable to break down
sugars through the glycolytic pathway) indicated that
20% of the glucose is oxidized through the pentose
phosphate pathways while 80% is altered by the nonoxi-
dative pentose phosphate pathway.
100
However, it is
not clear how the C
3
unit used in the nonoxidative
pathway (Fig. 17-8B) is formed if glycolysis is blocked.
A number of fermentations are also based on the
pentose phosphate pathways (Section F,5).
4. The EntnerDoudoroff Pathway
An additional way of cleaving a six-carbon sugar
chain provides the basis for the EntnerDoudoroff
pathway which is used by Zymomonas lindneri and
many other species of bacteria. Glucose 6-P is oxidized
first to 6-phosphogluconate, which is converted by
dehydration to a 2-oxo-3-deoxy derivative (Eq. 17-18,
E. Catabolism of Sugars
COO

C O
CH
2
HC
HC
CH
2
OP
OH
2-Oxo-3-deoxy-6-phosphogluconate
OH
COO

C O
CH
3
Pyruvate
H
2
O
6-Phosphogluconate
a
b
+
An aldolase
HC
HC
CH
2
OP
OH
O
Glyceraldehyde 3-P (17-18)
(17-17)
(17-16)
Ch 17IND page - E - 2/14/03, 11:16 AM 965
966 Chapter 17. The Organization of Metabolism
step a). The resulting 2-oxo-3-deoxy sugar is cleaved
by an aldolase (Eq. 17-18, step b) to pyruvate and
glyceraldehyde 3-P, which are then metabolized in
standard ways.
F. Fermentation: Life without Oxygen
Pasteur recognized in 1860 that fermentation was
not a spontaneous process but a result of life in the
near absence of air.
132
He realized that yeasts decompose
much more sugar under anaerobic conditions than
they do aerobically, and that the anaerobic fermentation
was essential to the life of these organisms. In addition
to the alcoholic fermentation of yeast, there are many
other fermentations which have been attractive subjects
for biochemical study. If life evolved at a time when
no oxygen was available, the most primitive organisms
must have used fermentations. They may be the oldest
as well as the simplest ways in which cells obtain
energy. The enzymes of the glycolysis pathway are
found in the small genomes of Mycoplasma, Haemophila,
and Methanococcus.
133,134
Fermentation is also a vital process in the human
body. Our muscles usually receive enough oxygen to
oxidize pyruvate and to obtain ATP through aerobic
metabolism, but there are circumstances in which the
oxygen supply is inadequate. During extreme exertion,
after most oxygen is consumed, muscle cells produce
lactate by fermentation. White muscle of fish and fowl
has little aerobic metabolism and normally yields L-lactic
acid as a principal end product. Likewise, a variety
of tissues within the human body, including the
transparent lens and cornea of our eyes, are poorly
supplied with blood and depend upon fermentation
of glucose to lactic acid. Red blood cells and skin and
sometimes adipose tissue are also major producers of
lactic acid.
135
Of the ~ 115 g of lactic acid present in a
70-kg human body, about 29% comes from erythro-
cytes, 29% from skin, 17% from the brain, and 16%
from skeletal muscle.
136
Because lactic acid lowers the
pH of cells it must be removed efficiently.
Some of the lactic acid formed in muscle and
most of the lactate formed in less aerobic tissues (e.g.,
adipose tissue)
136a
enters the bloodstream, which
normally contains 12 mM lactate,
136
and is carried to
the liver where it is reoxidized to pyruvate. Part of the
pyruvate is then oxidized via the citric acid cycle while
a larger part is reconverted to glucose (Section J,5).
This glucose may be released into the bloodstream and
returned to the muscles. The overall process is known
as the Cori cycle. Lactic acid accumulates in muscle
after vigorous exercise. It is exported to the liver slowly,
but if mild exercise continues the lactate may be
largely oxidized within muscle via the tricarboxylic
acid cycle. Recent NMR studies have shown that lactic
acid is formed rapidly during muscular contraction,
even when exercise is mild.
136b
During the initial 15
ms of contraction the ATP utilized is regenerated from
creatine phosphate (Eq. 6-67). During the remainder of
the contraction (up to ~100 ms) glycogen is converted
to lactic acid to provide ATP and to replenish the
creatine phosphate. In the resting period following
contraction most of the lactate is either dehydrogenated
to pyruvate and oxidized in mitochondria or exported
to other tissues. The glycogen stores in muscle are
renewed by synthesis from blood glucose. Lactic acid
is a convenient energy carrier and a precursor for
gluconeogenesis which can be transferred between
tissues easily.
136c
Cancer cells often take advantage of
this opportunity to grow rapidly using fermentation
of glucose to lactic acid as a source of energy.
136d
Alcoholic fermentation allows roots of some plants
to survive short periods of flooding. Ethanol does not
acidify the tissues as does lactic acid, avoiding possible
damage from low pH.
137,138
Goldfish can also use the
ethanolic fermentation for short times, excreting the
ethanol.
139
1. Fermentations Based on the Embden
Meyerhof Pathway
Homolactic and alcoholic fermentations. The
reactions by which glucose can be converted to lactate
and, by yeast cells, to ethanol and CO
2
(Figs. 10-3 and
17-7) illustrate several features common to all fermenta-
tions. The NADH produced in the oxidation step is
reoxidized in a reaction by which substrate is reduced
to the final end product. The NAD alternates between
oxidized and reduced forms. This coupling of oxida-
tion steps with reduction steps in exact equivalence is
characteristic of all true (anaerobic) fermentations. The
formation of ATP from ADP and P
i
by substrate-level
phosphorylation is also common to all fermentations.
The stoichiometry is often nearly exact and simple. For
example, according to the reaction of Eq. 17-19, which is
outlined step-by-step in Fig. 17-7, a net total of two moles
of ATP is formed per mole of glucose fermented.
Energy relationships. If we disregard the syn-
thesis of ATP, the equations for the lactic acid and
ethanol fermentations are given by Eqs. 17-19 and 17-20.
Glucose 2 lactate

+ 2 H
+
G (pH 7) = 196 kJ mol
1
( 46.8 kcal mol
1
)
Glucose 2 CO
2
+ 2 ethanol
G = 235 kJ mol
1
The Gibbs energy changes are negative and of sufficient
magnitude that the reactions will unquestionably go to
completion. However, the synthesis of two molecules
of ATP from inorganic phosphate and ADP, a reaction
(17-19)
(17-20)
Ch 17IND page - E - 2/14/03, 11:16 AM 966
967
(Eq. 17-21) for which G is substantially positive, is
coupled to the fermentation.
ADP
3
+ HPO
4
2
+ H
+
ATP
4
+ H
2
O
G (pH 7) = + 34.5 kJ mol
1
(Table 6-5)
To obtain the net Gibbs energy change for the complete
reaction we must add 2 x 34.5 = + 69.0 kJ to the values
of G for Eqs. 17-19 and 17-20. When this is done we
see that the net Gibbs energy changes are still highly
negative, that the reactions will proceed to completion,
and that these fermentations can serve as an usable
energy source for organisms.
Biochemists sometimes divide G for the ATP
synthesis in a coupled reaction sequence (in this case
+69 kJ) by the overall Gibbs energy decrease for the
coupled process (196 or 235 kJ mol
1
) to obtain an
efficiency. In the present case the efficiency would
be 35% and 29% for coupling of Eq. 17-21 (for 2 mol of
ATP) to Eqs. 17-19 and 17-20, respectively. According
to this calculation, nature is approximately one-third
efficient in the utilization of available metabolic Gibbs
energy for ATP synthesis. However, it is important to
realize that this calculation of efficiency has no exact
thermodynamic meaning. Furthermore, the utilization
of ATP formed by a cell for various purposes is far
from 100% efficient.
Why are the Gibbs energy decreases for Eqs. 17-19
and 17-20 so large? No overall oxidation takes place;
there is only a rearrangement of the existing bonds
between atoms of the substrate. Why does this rear-
rangement of bonds lead to a substantial negative G?
An answer is suggested by an examination of the
numbers of each type of bond in the substrate and in
the products. During the conversion from glucose
to two molecules of lactate one CC bond, one CO
bond, and one OH bond are lost and one CH bond
and one C=O are gained. If we add up the bond
energies for these bonds (Table 6-6) we find that the
difference (H) between substrate and products
amounts to only about 20 kJ/mol. However, lactic
acid contains a carboxyl group, and carboxyl groups
have a special stability as a result of resonance. The
extra resonance energy of a carboxyl group (Table 6-6)
is ~ 117 kJ (28 kcal) per mole or 234 kJ/mol for two
carboxyl groups. This is approximately the same as
the Gibbs energy change (Eq. 17-19) for fermentation
of glucose to lactate. Thus, the energy available results
largely from the rearrangement of bonds by which the
carboxyl groups of lactate are formed. Likewise, the
resonance stabilization of CO
2
is given by Pauling as
151 kJ/mol, again of just the right magnitude to explain
G in alcoholic fermentations (Eq. 17-20).
On this basis we can state as a general rule that
fermentations can occur when substrates consisting of
largely singly bonded atoms and groups, such as the
carbonyl groups that are not highly stabilized by
resonance, are converted to products containing car-
boxyl groups or to CO
2
. If we assume an efficiency of
~ 30%, the energy available will be about sufficient for
synthesis of one ATP molecule for each carboxyl group
or CO
2
created. Bear in mind that generation of ATP
also depends upon availability of a mechanism. It is of
interest that most synthesis of ATP is linked directly to
the chemical processes by which carboxyl groups or
CO
2
molecules are created in a fermentation process.
The most important single reaction is the oxidation
of the aldehyde group of glyceraldehyde 3-P to the
carboxyl group of 3-phosphoglycerate (steps 6a 6c
and 7 in Fig. 17-7; see also Fig. 15-6).
Compare the fermentation of glucose with the
complete oxidation of the sugar to carbon dioxide and
water (Eq. 17-22), a process which yeast cells (as well
as our own cells) carry out in the presence of air. The
overall Gibbs energy change is over 10 times greater
than that for fermentation, a fact that permits the cell
Glucose + 6 O
2
6 CO
2
+ 6 H
2
O
G = 2872 kJ ( 686.5 kcal) mol
1
to form an enormously greater quantity of ATP. The
net gain in ATP synthesis, accompanying Eq. 17-22, is
usually about 38 mol of ATP 19 times more than is
available from fermentation of glucose. Thus, the
explanation of Pasteurs observation that yeast decom-
poses much less sugar in the presence of air than in its
absence is clear. Also, we can understand why a cell,
living anaerobically, must metabolize a very large amount
of substrate to grow. (Recall from Chapter 6 that ~ 1 mol
of ATP energy is needed to produce 10 g of cells.)
Variations of the alcoholic and homolactic
fermentations. The course of a fermentation is often
affected drastically by changes in conditions. Many
variations can be visualized by reference to Fig. 17-9,
which shows a number of available metabolic sequences.
We have already discussed the conversion of glucose to
triose phosphate and via reaction pathway a to pyruvate,
via reaction c to lactate, and via reaction d to ethanol.
If bisulfite is added to a fermenting culture of
yeast, the acetaldehyde formed through reaction d is
trapped as the bisulfite adduct blocking the reduction
of acetaldehyde to ethanol, an essential part of the
fermentation. Yeast cells accommodate this change by
using the accumulating NADH to reduce half of the
triose-P to glycerol through pathway b. Two enzymes
are needed, a dehydrogenase and a phosphatase, to
hydrolytically cleave off the phosphate. The balanced
reaction is given by Eq. 17-23:
Glucose glycerol + acetaldehyde (trapped) + CO
2
G (pH 7) = 105 kJ mol
1
In this reaction only one molecule of CO
2
is produced
(17-22)
F. Fermentation: Life without Oxygen
(17-23)
(17-21)
Ch 17IND page - E - 2/14/03, 11:16 AM 967
968 Chapter 17. The Organization of Metabolism
but the overall Gibbs energy change is still adequate
to make the reaction highly spontaneous. However
(referring to Fig. 17-9), we see that the net synthesis
of ATP is now apparently zero. The fermentation
apparently does not permit cell growth. Nevertheless,
it has been used industrially for production of glycerol.
Reduction of dihydroxyacetone phosphate to
glycerol phosphate also occurs in insect flight muscle
and apparently operates as an alternative to lactic acid
formation in that tissue. There is no net gain of ATP in
the conversion of free glucose to glycerol phosphate
and pyruvate, but using stored glycogen in muscle as
the starting material, the dismutation of triose-P to
glycerol-P and pyruvate provides one ATP per glucose
unit rapidly during the vigorous contraction of the
powerful insect flight muscle. During the slower recov-
ery phase, glycerol-P is thought to be reoxidized after
entering the mitochondria of these highly aerobic cells.
Thus, the transport of glycerol-P into mitochondria
serves as a means for transporting reducing equivalents
derived from reoxidation of NADH into the mitochon-
dria. Indeed, the significance of glycerol-P to muscle
metabolism may be more related to this function than
to the rapid formation of ATP (see Chapter 18).
Why does the glycolysis sequence begin with
phosphorylation of glucose by ATP? The phospho
groups probably provide convenient handles and
doubtless assist in substrate recognition. There may
be a kinetic advantage but also a danger. Unless there
is adequate regulation the turbo design, in which
ATP is used at the outset to drive glycolysis, may lead
to accumulation of phosphorylated intermediates and
to inadequate concentrations of ATP and inorganic
phosphate.
139a,b
Yeast cells guard against this problem
by synthesizing trehalose 6-phosphate, which acts as
a feedback inhibitor of hexokinase.
139a
Trypanosomes
utilize a different type of control. The enzymes that
convert glucose into 3-phosphoglycerate are present
in membrane-bounded organelles called glycosomes.
Phosphoglycerate is exported from them into the
cytosol where glycolysis is completed.
139b
Since inor-
ganic phosphate is essential for ATP formation, if the
P
i
concentration falls too low the rate of fermentation
by yeast juice is greatly decreased, an observation
made by Harden and Young
139c
in 1906.
2. The Mixed Acid Fermentation
Enterobacteria, including E. coli, convert glucose to
ethanol and acetic acid and either formic acid or CO
2
and H
2
derived from it. The stoichiometry is variable
but the fermentation can be described in an idealized
form as follows:
Glucose + H
2
O ethanol + acetate

+ H
+
+ 2 H
2
+ 2 CO
2
G (pH 7) = 225 kJ mol
1
The details of the process and the oxidationreduc-
tion balance can be pictured as in Eq. 17-25. Pyruvate
is cleaved by the pyruvate formatelyase reaction
(Eq. 15-37) to acetyl-CoA and formic acid. Half of
the acetyl-CoA is cleaved to acetate via acetyl-P with
generation of ATP, while the other half is reduced in
two steps to ethanol using the two molecules of NADH
produced in the initial oxidation of triose phosphate
(Eq. 17-25). The overall energy yield is three molecules
of ATP per glucose. The efficiency is thus (3 x 34.5)
225 = 46%. Some of the glucose is converted to
D-lactic and to succinic acids (pathway f, Fig. 17-9);
hence the name mixed acid fermentation. Table 17-1
gives typical yields of the mixed acid fermentation of
E. coli. Among the four major products are acetate,
ethanol, H
2
, and CO
2
, as shown in Eq. 17-25. How-
ever, at high pH formate accumulated instead of CO
2
.
TABLE 17-1
Products of the Mixed Acid Fermentation by
E. coli at Low and High Values of pH
a
Acetate 36 39
Ethanol 50 51
H
2
70 0.3
CO
2
88 1.7
Formate 2.4 86
Lactate 79 70
Succinate 11 15
Glycerol 1.4 0.3
Acetoin 0.1 0.2
Butanediol 0.3 0.2
a
From Tempest, D. W. and Neijssel, O. M.
140
Based on data of
Blackwood.
141
Product pH 6.2 pH 7.8
(Millimole formed from
100 mmol of glucose)
Glucose
2 NADH
2 ATP
2 Pyruvate
2 HCOOH
2 Acetyl-CoA
ATP
NADH
Acetate Acetaldehyde
NADH
Ethanol
2 H
2
Formic
hydrogen-lyase
2 CO
2
Pyruvate
formate-lyase
(17-25)
(17-24)
Ch 17IND page - E - 2/14/03, 11:17 AM 968
969
Figure 17-9 Reaction sequences in fermentation based on the EmbdenMeyerhofParnas pathway. Oxidation steps
(producing NADH + H
+
) are marked O; reduction steps (using NADH + H
+
) are marked R.
CO
2
Ethanol Acetaldehyde
Acetyl-P
Acetate
Adduct
Oxaloacetate
Malate
Succinate
CoA
transfer
Succinyl-CoA
Methylmalonyl-CoA
Propionyl-CoA
Propionate
1-Propanol
E
Biotin
CO
2
ATP
Formate
H
2
CO
2
R
ATP
O
R
HSO
3
R
R
R
R
HCO
3
O
(2 mol)
HCO
3
d
f
Acetolactate
Acetoin
CO
2
2,3-Butanediol
R
CoA
Acetoacetyl-CoA
Butyryl-CoA
Acetoacetate
Acetone
2-Propanol
Butyrate
1-Butanol
ATP
ATP
CO
2
R
R
R
R
R
1,3-Bisphosphoglycerate
ATP
PEP
Lactate
Pyruvate
CO
2
Acetyl-CoA
ATP
R
CO
2
O
(2 mol)
c
e
g
Glucose
1
2
Glycogen
Glycerol-P Triose-P
Glycerol
ATP
1
2
ATP
1
2
P
i
O
P
i
R
b a

Over one-third of the glucose was fermented to lactate


in both cases.
In some mixed acid fermentations (e.g., that of
Shigella) formic acid accumulates, but in other cases
(e.g., with E. coli at pH 6) it is converted to CO
2
and
H
2
(Eq. 17-25). The equilibration of formic acid
with CO
2
and hydrogen is catalyzed by the formic
hydrogenlyase system which consists of two iron
sulfur enzymes. The selenium-containing formate
dehydrogenase (Eq. 16-63) catalyzes oxidation of
formate to CO
2
by NAD
+
, while a membrane-bound
hydrogenase (Eq. 16-48) equilibrates NADH + H
+
with NAD
+
+ H
2
. Hydrogenase also serves to release
H
2
from excess NADH. Krebs pointed out that an
excess of NADH may arise because growth of cells
requires biosynthesis of many components such as
amino acids. When glucose is the sole source of
carbon, biosynthetic reactions involve an excess of
oxidation steps over reduction steps.
142
The excess
of reducing equivalents may be released as H
2
or
F. Fermentation: Life without Oxygen
Ch 17IND page - E - 2/14/03, 11:17 AM 969
970 Chapter 17. The Organization of Metabolism
may be used to form highly reduced products such as
succinate.
Among such genera as Aerobacter and Serratia part
of the pyruvate formed is condensed with decarboxyl-
ation to form S acetolactate,
143
which is decarboxyl-
ated to acetoin (Eq. 17-26; pathway g of Fig. 17-9).
The acetoin is reduced with NADH to 2,3-butanediol,
while a third molecule of pyruvate is converted to
ethanol, hydrogen, and CO
2
(Eq. 17-26). The reaction
provides the basis for industrial production of butane-
diol, which can be dehydrated nonenzymatically to
butadiene.
Mixed acid fermentations are not limited to bacteria.
For example, trichomonads, parasitic flagellated pro-
tozoa, have no mitochondria. They export pyruvate
into the bloodstreams of their hosts and also contain
particles called hydrogenosomes which can convert
pyruvate to acetate, succinate, CO
2
, and H
2
.
144
Hydro-
genosomes are bounded by double membranes and
have a common evolutionary relationship with both
mitochondria and bacteria. The enzyme that catalyzes
pyruvate cleavage in hydrogenosomes apparently
does not contain lipoate and may be related to the
pyruvateferredoxin oxidoreductase of clostridia
(Eq. 15-35). The hydrogenosomes also contain an
active hydrogenase.
Many invertebrate animals are true facultative
anaerobes, able to survive for long periods, sometimes
indefinitely, without oxygen.
145 147
Among these are
Ascaris (Fig. 1-14), oysters, and other molluscs. Succi-
nate and alanine are among the main end products
of anaerobic metabolism. The former may arise by a
mixed acid fermentation that also produces pyruvate.
The pyruvate is converted to acetate to balance the
fermentation in Ascaris lumbricoides, which is in effect
an obligate anaerobe. However, in molluscs the pyru-
vate may undergo transamination with glutamate to
form alanine and 2-oxoglutarate; the oxoglutarate may
be oxidatively decarboxylated to succinate. The reactions
depend upon the availability of a store of glutamate or
of other amino acids, such as arginine, that can give
rise to glutamate.
3. The Propionic Acid Fermentation
Propionic (propanoic) acid-producing bacteria are
numerous in the digestive tract of ruminants. Within
the rumen some bacteria digest cellulose to form
glucose, which is then converted to lactate and other
products. The propionic acid bacteria can convert
either glucose or lactate into propionic and acetic acids
which are absorbed into the bloodstream of the host.
Usually some succinic acid is also formed.
The basis of the propionic acid fermentation is
conversion of pyruvate to oxaloacetate by carboxyla-
tion and the further conversion through succinate and
succinyl-CoA to methylmalonyl-CoA and propionyl-
CoA, reactions which are almost the exact reverse of
those for the oxidation of propionate in the animal
body (Fig. 17-3, pathway d). However, whereas the
carboxylation of pyruvate to oxaloacetate in the
animal body requires ATP, the propionic acid bacteria
save one equivalent of ATP by using a carboxyltrans-
ferase (p. 725). This enzyme donates a carboxyl group
from a preformed carboxybiotin compound generated
in the decarboxylation of methylmalonyl-CoA in the
next to final step of the reaction sequence (Fig. 17-10).
A second molecule of ATP is saved by linking directly
the conversion of succinate to succinyl-CoA to the
cleavage of propionyl-CoA to propionate through the
use of a CoA transferase (Eq. 12-50). To provide for
oxidationreduction balance, two-thirds of the glucose
goes to propionate and one-third to acetate (Eq. 17-27):
1
1
/2 Glucose
2 propionate

+ acetate

+ 3 H
+
+ CO
2
+ H
2
O
G (pH 7) = 465 kJ per 1
1
/2 mol of glucose
More carboxyl groups and CO
2
molecules are formed
in this fermentation (2
2
/3 per glucose molecule) than
in the regular lactic acid fermentation. The yield of
ATP (also 2
2
/3 mol/mol of glucose fermented) is corre-
spondingly greater and G is more negative.
Using the same mechanism (Fig. 17-10), propionic
acid bacteria are also able to ferment lactate, the pro-
duct of fermentation by other bacteria, to propionate
and acetate (Eq. 17-28). The net gain is one molecule
of ATP. This reaction probably accounts for the niche
3 Pyruvate
H
3
C
C
C
COO

O
CH
3
HO
3 ATP
3 NADH
Acetyl-CoA
1
2 1 / Glucose
H
3
C
C
C
H
O
CH
3
HO
CO
2
two
molecules
Ethanol
H
2
+ CO
2
Acetolactate
CO
2
2 NADH
Acetoin
NADH
2,3-Butanediol
(17-26)
(17-27)
Ch 17IND page - E - 2/14/03, 11:17 AM 970
971
(17-28)
Figure 17-10 Propionic acid fermentation of Propionobacte-
ria and Veillonella. Oxidation steps are designated by the
symbol O and reduction steps by R. The two coupled
reactions marked by asterisks are catalyzed by carboxyl-
transferase.
F. Fermentation: Life without Oxygen
Oxaloacetate
R
Malate
R
Succinate
Succinyl-CoA
Methylmalonyl-CoA
Propionyl-CoA
Propionate
Glucose
1
2
O ATP
Pyruvate
CO
2
O
E-biotin
Mutase requiring
vitamin B
12
coenzyme
Racemase
Methylmalonyl-CoA
These two
reactions are
linked. The
energy of the
propionyl-CoA
linkage is
conserved as
the CoA is
transferred
to succinate.
EnzymebiotinCO
2
Acetyl-CoA
ATP
Acetate
Reverse
of citric
acid cycle
reactions
Lactate
O
*
*
3 Lactate

2 propionate

+ acetate

+ H
2
O + CO
2
G (pH 7) = 171 kJ per 3 mol of lactate
in the ecology of the animal rumen that is occupied by
propionic acid bacteria.
4. Butyric Acid and Butanol-Forming
Fermentations
A variety of fermentations are carried out by bac-
teria of the genus Clostridium and by the rumen organ-
isms Eubacterium (Butyribacterium) and Butyrivibrio.
For example, glucose may be converted to butyric and
acetic acids together with CO
2
and H
2
(Eqs. 17-29 and
17-30).
2 Glucose + 2 H
2
O butyrate

+ 2 acetate

+ 4 CO
2
+ 6 H
2
+ 3 H
+
G (pH 7) = 479 kJ per 2 mol of glucose
The yield of ATP (3
1
/2 mol/mol of glucose) is the
highest we have discussed giving an efficiency of 50%.
Another fermentation yields butanol, isopropanol,
ethanol, and acetone.
The fermentation of Eq. 17-31 is catalyzed by
Clostridium kluyveri. The value of G is one of the
lowest that we have considered but is still enough to
provide easily for the synthesis of one molecule of ATP.
2 CH
3
CH CHCOO

+ 2 H
2
O
Crotonate
butyrate

+ 2 acetate

+ H
+
G (pH 7) = 105 kJ mol
1
The energy of the butyryl-CoA linkage and of one of
the acetyl-CoA linkages is conserved and utilized in
the initial formation of crotonyl-CoA (Eq. 17-32). That
leaves one acetyl-CoA which can be converted via
acetyl-P to acetate with formation of ATP.
2 Glucose
4 NADH
2 ATP
2 H
2
Two used below
4 Pyruvate
4 CO
2
+ 4 H
2
4 Acetyl-CoA
1 Acetoacetyl-CoA
-Hydroxybutyryl-CoA
Crotonyl-CoA
Butyryl-CoA Butyrate

ATP
2 Acetate

NADH
NADH
(17-30)
(17-29)
(17-31)
Ch 17IND page - E - 2/14/03, 11:17 AM 971
972 Chapter 17. The Organization of Metabolism
(17-34)
Glucose
Glucose 6-P
6-Phosphogluconate
CO
2
Pentose-P
NADH
NADH
Acetaldehyde Ethanol Acetyl-P
Glyceraldehyde 3-P
2 ATP
NADH Pyruvate
Lactate
ATP
P
i
Phospho-
ketolase
(17-33)
5. Fermentations Based on the Phospho-
gluconate and Pentose Phosphate Pathways
Some lactic acid bacteria of the genus Lactobacillus,
as well as Leuconostoc mesenteroides and Zymomonas
mobilis, carry out the heterolactic fermentation (Eq.
17-33) which is based on the reactions of the pentose
phosphate pathway. These organisms lack aldolase,
the key enzyme necessary for cleavage of fructose
1,6-bisphosphate to the triose phosphates. Glucose is
converted to ribulose 5-P using the oxidative reactions
of the pentose phosphate pathway. The ribulose-
phosphate is cleaved by phosphoketolase (Eq. 14-23)
to acetyl-phosphate and glyceraldehyde 3-phosphate,
which are converted to ethanol and lactate, respectively.
The overall yield is only one ATP per glucose fermented.
This is generated in the substrate level oxidative phos-
phorylation catalyzed by phosphoketolase. Metabolic
engineering of Zymomonas was accomplished by trans-
ferring from other bacteria two operons that provide
for assimilation of xylose and a complete set of enzymes
for the pentose phosphate pathway. The engineered
bacteria are able to convert pentose phosphates nonoxi-
datively (see Fig. 17-8) into glyceraldehyde 3-phosphate,
which is converted to ethanol in high yield and with
much greater synthesis of ATP than according to Eq.
17-33.
148
A variation of the heterolactic fermentation is used
by Bifidobacterium (Eq. 17-34).
149
Phosphoketolase and
a phosphohexoketolase, which cleaves fructose 6-P
to erythrose 4-P and acetyl-P, are required, as are the
enzymes of the sugar rearrangement system (Section
E,3). The net yield of ATP is 2
1
2 molecules per mole-
cule of glucose.
G. Biosynthesis
In this section and sections H K the general
principles and strategy of synthesis of the many carbon
compounds found in living things will be considered.
Since green plants and autotrophic bacteria are able to
assemble all of their needed carbon compounds from
CO
2
, let us first examine the mechanisms by which
this is accomplished. We will also need to ask how
some organisms are able to subsist on such simple
compounds as methane, formate, or acetate.
2 Crotonate

2 Crotonyl-CoA
Butyryl-CoA
Butyrate

ATP
Acetate

Acetyl-P
NAD
+
NADH
Acetoacetyl-CoA
2 Acetyl-CoA
Acetate

CoA
transferase
Reactions
linked by
energy-
conserving
CoA transferase
(17-32)
Phospho-
hexoketolase
Acetyl-P
3 ATP
3 Acetate

P
i
Erythrose 4-P
Sugar rearrangement
2 Pentose-P
2 Triose-P
2 P
i
2 Acetyl-P
2 Lactate

4 ATP
2 NADH
2 Pyruvate

2 Glucose
2 Fructose 6-P
2 ATP
Ch 17IND page - E - 2/14/03, 11:17 AM 972
973
1. Metabolic Loops and Biosynthetic Families
As was pointed out in Chapter 10, routes of bio-
synthesis (anabolism) often closely parallel pathways
of biodegradation (catabolism). Thus, catabolism
begins with hydrolytic breakdown of polymeric mole-
cules; the resulting monomers are then cleaved into
small two- and three-carbon fragments. Biosynthesis
begins with formation of monomeric units from small
pieces followed by assembly of the monomers into
polymers. The mechanisms of the individual reactions
of biosynthesis and biodegradation are also often
closely parallel. However, in most instances, there are
clear-cut differences. A first principle of biosynthesis
is that biosynthetic pathways, although related to catabolic
pathways, differ from them in distinct ways and are often
catalyzed by completely different sets of enzymes.
The sum of the pathways of biosynthesis and
biodegradation form a continuous loop a series of
reactions that take place concurrently and often within
the same part of a cell. Metabolic loops often begin in
the central pathways of carbohydrate metabolism with
three- or four-carbon compounds such as phospho-
glycerate, pyruvate, and oxaloacetate. After loss of
some atoms as CO
2
the remainder of the compound
rejoins the mainstream of metabolism by entering a
catabolic pathway leading to acetyl-CoA and oxidation
in the citric acid cycle. Not all of the loops are closed
within a given species. For example, human beings are
unable to synthesize the vitamins and the essential amino
acids. We depend upon other organisms to make
these compounds, but we do degrade them. Some
metabolites, such as uric acid, are excreted by humans
and are further catabolized by bacteria. From a chemi-
cal viewpoint the whole of nature can be regarded as
an enormously complex set of branching and inter-
connecting metabolic cycles. Thus, the synthetic path-
ways used by autotrophs are all parts of metabolic
loops terminating in oxidation back to CO
2
.
It is often not possible to state at what point in a
metabolic loop biosynthesis has been completed and
biodegradation begins. An end product X that serves
one need of a cell may be a precursor to another cell
component Y which is then degraded to complete the
loop. The reactions that convert X to Y can be regarded
as either biosynthetic (for Y) or catabolic (for X).
2. Key Intermediates and Biosynthetic
Families
In examining routes of biosynthesis it is helpful
to identify some key intermediates. One of these is
3-phosphoglycerate. This compound is a primary
product of photosynthesis and may reasonably be
regarded as the starting material from which all other
carbon compounds in nature are formed. Phospho-
glycerate, in most organisms, is readily interconvertible
with both glucose and phosphoenolpyruvate (PEP).
Any of these three compounds can serve as the pre-
cursor for synthesis of other organic materials. A first
stage in biosynthesis consists of those reactions by
which 3-phosphoglycerate or PEP arise, whether it be
from CO
2
, formate, acetate, lipids, or polysaccharides.
The further biosynthetic pathways from 3-phos-
phoglycerate to the myriad amino acids, nucleotides,
lipids, and miscellaneous compounds found in cells
are complex and numerous. However, the basic features
are relatively simple. Figure 17-11 indicates the origins
of many substances including the 20 amino acids
present in proteins, nucleotides, and lipids. Among
the additional key biosynthetic precursors that can be
identified from this chart are glucose 6-phosphate,
pyruvate, oxaloacetate, acetyl-CoA, 2-oxoglutarate,
and succinyl-CoA.
The amino acid serine originates almost directly
from 3-phosphoglycerate. Aspartate arises from oxalo-
acetate and glutamate from 2-oxoglutarate. These
three amino acids each are converted to families of
other compounds.
150
A little attention paid to establish-
ing correct family relationships will make the study of
biochemistry easier. Besides the serine, aspartate, and
glutamateoxoglutarate families, a fourth large family
originates directly from pyruvate and a fifth (mostly
lipids) from acetyl-CoA. The aromatic amino acids are
formed from erythrose 4-P and PEP via the key inter-
mediate chorismic acid (Box 9-E; Fig. 25-1). Other
families of compounds arise from glucose 6-P and from
the pentose phosphates. These groups have been set
off roughly by the boxes outlined in green in Fig. 17-11.
H. Harnessing the Energy of ATP for
Biosynthesis
In the past it seemed reasonable to think that some
biosynthetic pathways involved exact reversal of
catabolic pathways. For example, it was observed that
glycogen phosphorylase catalyzed elongation of gly-
cogen branches by transfer of glycosyl groups from
glucose 1-phosphate. Likewise, the enzymes needed
for the oxidation of fatty acid derivatives, when
isolated from mitochondria, catalyze formation of fatty
acyl-CoA derivatives from acetyl-CoA and a reducing
agent such as NADH. However, reactant concentrations
within cells are rarely appropriate for reversal of a
catabolic sequence. For catabolic sequences the Gibbs
energy change is usually distinctly negative and reversal
requires high concentrations of end products. However,
the latter are often removed promptly from the cells.
For example, NADH produced in degradation of
fatty acids is oxidized to NAD
+
and is therefore never
available in sufficient concentrations to reverse the
oxidation sequence.
H. Harnessing the Energy of ATP for Biosynthesis
Ch 17IND page - E - 2/14/03, 11:17 AM 973
974 Chapter 17. The Organization of Metabolism
Figure 17-11 Some major biosynthetic pathways. Some key intermediates are enclosed in boxes and the 20 common
amino acids of proteins are encircled. Key intermediates for each family are in shaded boxes or elipses. Green lines trace
the reactions of the glyoxylate pathway and of glucogenesis.
Trp
Erythrose 4-P
Polysaccharides
Muramic,
neuraminic acids
Ribulose 1,5-P
2
Glyoxylate
one C
units
2-Oxoisovalerate
Prephenate
Malate
Succinate
Glyoxylate
Isocitrate
Polyprenyls
in part
Hydroxyproline
Corrins, porphins
RNA, DNA
Glucose 6-P
Glyceraldehyde 3-P
Chorismate
Citrate
Tyr
Phe
Val
Fatty acids
Polyketides
-Hydroxy--
methylglutaryl-CoA
Leu
ATP
Malonyl-CoA
Oxaloacetate
Acetyl-CoA
Lys -Oxoglutarate
Pro
Arg
Gln
-Aminobutyrate
His
Purine
nucleotides
Lactate
Ala
ATP
Pyruvate
Cys
Succinyl-CoA
Phosphoenolpyruvate
Arogenate
Glu
Pyrimidine
nucleotides
-Oxobutyrate
Thr
Asn
Met
Homoserine
Ile
Lys
in part
Asp
Ser
Gly
Pentose
phosphates
3 P-glycerate
CO
2
CO
2
Natures answer to the problem of reversing a catabolic
pathway lies in the coupling of cleavage of ATP to the
biosynthetic reaction. The concept was introduced in
Chapter 10, in which one sequence for linking hydrol-
ysis of ATP to biosynthesis was discussed. However,
living cells employ several different methods of har-
nessing the Gibbs energy of hydrolysis of ATP to drive
biosynthetic processes. Many otherwise strange aspects
of metabolism become clear if it is recognized that they
provide a means for coupling ATP cleavage to biosyn-
thesis. A few of the most important of these coupling
mechanisms are summarized in this section.
1. Group Activation
Consider the formation of an ester (or of an amide)
from a free carboxylic acid and an alcohol (or amine)
by elimination of a molecule of water (Eq. 17-35).
The reaction is thermodynamically unfavorable with
values of G (pH 7) ranging from ~ + 10 to 30 kJ mol
1
depending on conditions and structures of the specific
compounds. Long ago, organic chemists learned that
such reactions can be made to proceed by careful
removal of the water that is generated (Eq. 17-35).
Ch 17IND page - E - 2/14/03, 11:17 AM 974
975
However, it is often better to acti-
vate the carboxylic acid by con-
version to an acyl chloride or an
anhydride:
Nucleophilic attack on the carbonyl
group of such a compound results
in displacement of a good leaving
group, Cl

or RCOO

. Nature
has followed the same approach in
forming from carboxylic acids acyl
phosphates or acyl-CoA deriva-
tives.
The virtue of these activated
acyl compounds in biosynthetic
reactions was considered in
Chapter 12 and Table 15-1. Just
as a carboxylic acid can be converted
to an active acyl derivative, so other
groups can be activated. ATP and
other compounds with phospho
groups of high group transfer poten-
tial are active phospho compounds.
Sulfate is converted to a phospho-
sulfate anhydride, an active sulfo
derivative. Sugars are converted
to compounds such as glucose 1-P
or sucrose, which contain active
glycosyl groups. The group trans-
fer potentials of the latter, though
not as great as that of the phospho
groups of ATP, are still high enough
to make glucose 1-phosphate and
sucrose effective glycosylating
reagents. Table 17-2 lists several
of the more important activated
groups.
Group activation usually takes
place at the expense of ATP cleavage.
R C
O
Cl
R C
O
O C
O
R Acyl chloride
Acid anhydride
H
2
O
O R
N R
H
R C
O
OH
N R
H
R C
O
O R
H
H
or
or
(17-35)
H. Harnessing the Energy of ATP for Biosynthesis
R C
CH
2
Enoyl
C
O
R
Acyl
O P
O

O
P O R
O
O

Pyrophosphate
O P
O

O
C R
O
P
O

O
Phospho
S
O
O

O
Sulfo
Group Typical activated forms
O P
O

O
C R
CH
2
Enol phosphate
Sucrose
O
Glycosyl
N P
O

O
C N R
NH
2
+
H H
Guanidine phosphate
H
2
N C
O
Carbamoyl
R
Alkyl
S-Adenosylmethionine
H
3
C S R'
R
+
R C
CH
2
O P O

O
Enol phosphate
H
2
N C
O
O P
O
O

Carbamoyl phosphate
O S
O
O

O
P O
O
R
O

R C
O
O P
O
O R'
O

Acyl phosphate
R C
O
S R'
Thioester
Acyl phosphate
in PAPS (3-Phosphoadenosine-5-phosphosulfate)
HO
O
HO
CH2
OH
O
HO
O
CH2 HO
HO
OH
CH2OH
Glucosyl
Fructosyl
O
O PO3
2
Glycosyl phosphate
TABLE 17-2
Activated Groups Used in Biosynthesis
Ch 17IND page - E - 2/14/03, 11:17 AM 975
976 Chapter 17. The Organization of Metabolism
R C O
O
P
O
O

Acyl
phosphate
Activated
acyl group
Activated
phospho group
As pointed out in Chapter 12, acyl phosphates play
a central role in metabolism by virtue of the fact that
they contain both an activated acyl group and an
activated phospho group. The high group transfer
potential can be conserved in subsequent reactions in
either one group or the other (but not in both). Thus, dis-
placement on P by an oxygen of ADP will regenerate
ATP and attack on C by an SH will give a thioester.
Several of the other compounds in Table 17-2 can
also be split in two ways to yield different activated
groups, e.g., the phosphosulfate anhydride, enoyl
phosphate, and carbamoyl phosphate. It is probably
only through intermediates of this type that cleavage
of ATP can be coupled to synthesis of activated groups.
Such common intermediates are essential to the
synthesis of ATP by substrate-level phosphorylation
(Fig. 15-6).
2. Hydrolysis of Pyrophosphate
The splitting of inorganic pyrophosphate (PP
i
)
into two inorganic phosphate ions is catalyzed by
pyrophosphatases (p. 636)
150a,b
that apparently occur
universally. Their function appears to be simply to
remove the product PP
i
from reactions that produce it,
shifting the equilibrium toward formation of a desired
compound. An example is the formation of aminoacyl-
tRNA molecules needed for protein synthesis. As
shown in Eq. 17-36, the process requires the use of
two ATP molecules to activate one amino acid. While
the spending of two ATPs for the addition of one
monomer unit to a polymer does not appear necessary
from a thermodynamic viewpoint, it is frequently
observed, and there is no doubt that hydrolysis of PP
i
ensures that the reaction will go virtually to completion.
Transfer RNAs tend to become saturated with amino
acids according to Eq. 17-36 even if the concentration
of free amino acid in the cytoplasm is low. On the
other hand, kinetic considerations may be involved.
Perhaps the biosynthetic sequence would move too
slowly if it were not for the extra boost given by the
removal of PP
i
. Part of the explanation for the com-
plexity may depend on control mechanisms which
are only incompletely understood.
In some metabolic reactions pyrophosphate esters
are formed by consecutive transfer of the terminal
phospho groups of two ATP molecules onto a hydroxyl
group. Such esters often react with elimination of PP
i
,
e.g., in polymerization of prenyl units (reaction type 6B,
Table 10-1; Fig. 22-1). Again, hydrolysis to P
i
follows.
Thus, cleavage of pyrophosphate is a second very general
method for coupling ATP cleavage to synthetic reactions.
Although pyrophosphatases are ubiquitous, there
are organisms in which PP
i
is conserved by the cell
and replaces ATP in several glycolytic reactions. These
include Propionibacterium,
151,152
sulfate-reducing bac-
teria,
153
the photosynthetic Rhodospirillum, and the
parasitic Entamoeba histolytica.
152,154
In the latter the
internal concentration of PP
i
is about 0.2 mM. Green
plants also accumulate PP
i
at concentrations of up to
0.2 mM.
155
Apparently, pyrophosphate is not always
hydrolyzed immediately. Another mystery of metabo-
lism is the accumulation of inorganic polyphosphate
in chains of tens to many hundreds of phospho groups
linked, as in pyrophosphate, by phosphoanhydride
bonds. These polyphosphates are present in many
bacteria, including E. coli, and also in fungi, plants,
and animals.
156156b
They constitute a store of energy
as well as of phosphate. Various other functions have
also been proposed. A polyphosphate kinase transfers
a terminal phospho group from polyphosphate chains
onto ADP to form ATP. This source of metabolic energy
is evidently essential to the ability of Pseudomonas
aeruginosa to form biofilms.
156a
Both endophosphatases
and exophosphatases, of uncertain function, can
degrade the chains hydrolytically. An exophosphatase
from E. coli can completely hydrolyze polyphosphate
chains of 1000 units processively without release of
intermediates.
156b
C C
O

O R
H
NH
3
+
C C
R
H
NH
3
+
O
P
O
O O Adenosine
C C
R
H
NH
3
+
O
O
tRNA
Amino acid
H
2
O
2 P
i
PP
i
Pyrophosphatase
ATP
Aminoacyl adenylate
(activated amino acid)
tRNA (with free OH)
Activated amino acid
in final form ready for
protein synthesis
AMP
ATP
Adenylate kinase
2 ADP
Net: 2 ATP + amino acid + tRNA
aminoacyl tRNA + 2 ADP + 2 P
i
(17-36)
Ch 17IND page - E - 2/14/03, 11:17 AM 976
977
In a few instances group activation is coupled to
cleavage of ATP at C-5' presumably with formation of
bound tripolyphosphate (PPP
i
). The latter is hydro-
lyzed to P
i
and PP
i
and ultimately to three molecules
of P
i
. An example is the formation of S-adenosyl-
methionine
157
shown in Eq. 17-37. The reaction is a
displacement on the 5'-methylene group of ATP by the
sulfur atom of methionine. While the initial product
may be enzyme-bound PPP
i
, it is P
i
and PP
i
that are
released from the enzyme, the P
i
arising from the
terminal phosphorus (P

) of ATP.
157
The S-adenosyl-
methionine formed has the S configuration around the
sulfur.
158
3. Coupling by Phosphorylation and
Subsequent Cleavage by a Phosphatase
A third general method for coupling the hydrolysis
of ATP to drive a synthetic sequence is to transfer the
terminal phospho group from ATP to a hydroxyl group
somewhere on a substrate. Then, after the substrate
has undergone a synthetic reaction, the phosphate is
removed by action of a phosphatase. For example,
in the activation of sulfate (Eq. 17-38),
159
the overall
standard Gibbs energy change for steps a (catalyzed
by ATP sulfurylase
160,161
) and b is distinctly positive
(+12 kJ mol
1
). The equilibrium concentration of
adenylyl sulfate formed in this group activation
process is extremely low. Natures solution to this
problem is to spend another molecule of ATP to
phosphorylate the 3' OH of adenosine phospho-
sulfate. As the latter is formed, it is converted to
3'-phosphoadenosine-5'-phosphosulfate (Eq. 17-38,
step c) by a kinase, which is often part of a bifunctional
enzyme that also contains the active site of ATP sulfuryl-
ase.
162163a
Since the equilibrium in this step lies far
toward the right, the product accumulates in a sub-
stantial concentration (up to 1 mM in cell-free systems)
H. Harnessing the Energy of ATP for Biosynthesis
and serves as the active sulfo group donor in formation
of sulfate esters. The reaction cycle is completed by
two more reactions. In Eq. 17-38, step d, the sulfo
group is transferred to an acceptor, and in step e the
extra phosphate group is removed from adenosine
3',5'-bisphosphate by a specific phosphatase. Since
the reconversion of AMP to ADP requires expenditure
of still a third high-energy linkage of ATP, the overall
process makes use of three high-energy phosphate
linkages for formation of one sulfate ester.
An analogous use of ATP is found in photosyn-
thetic reduction of carbon dioxide in which ATP phos-
phorylates ribulose 5-P to ribulose bisphosphate and
the phosphate groups are removed later by phosphatase
action on fructose bisphosphate and sedoheptulose
bisphosphate (Section J,2). Phosphatases involved
in synthetic pathways usually have a high substrate
specificity and are to be distinguished from nonspecific
phosphatases which are essentially digestive enzymes
(Chapter 12).
4. Carboxylation and Decarboxylation:
Synthesis of Fatty Acids
A fourth way in which cleavage of ATP can be
coupled to biosynthesis was recognized in about 1958
when Wakil and coworkers discovered that synthesis
of fatty acids in animal cytoplasm is stimulated by
carbon dioxide. However, when
14
CO
2
was used in
O
Adenine
OH OH
+
CH
2
S
CH
3
H
3
N
C
CH
2
CH
2
OOC H

+
ATP
P
i
+PP
i
S-Adenosylmethionine
(AdoMet or SAM - older)
See also p. 581
Methionine
H
3
N
C
CH
2
CH
2
S
CH
3
OOC H

+
S configuration
around sulfur
(17-37)
a
d
b
H
2
O
PP
i
3
G (pH 7) =
33 kJ mol
-1
2P
i
+ H
+
G
0
= +45 kJ mol
-1
ATP
3
SO
4
2
ATP sulfurylase
ATP
ADP
Acceptor
Adenosine 3,5-
bisphosphate
Acceptor-sulfate Phosphatase
H
2
O
AMP + P
i
3-Phosphoadenosine-
5-Phosphosulfate (PAPS)

O S O
O
O
P
O
O Adenosine
O

Adenylyl sulfate

O S O
O
O
P
O
O CH
2
O

O
Adenosine
O OH
2
O
3
P
e
c
(17-38)
Ch 17IND page - E - 2/14/03, 11:17 AM 977
978 Chapter 17. The Organization of Metabolism
(17-39)
R C
O
S
E
C C
S
O

OOC
H
R
H
ACP
C C
O

S
H
H ACP
C
O
S
E
R
R
C
C
C
S
O O
H H
ACP
E S

*
-Oxoacyl-ACP
CO
2
*
(17-40)
the experiment no radioactivity appeared in the fatty
acids formed. Rather, it was found that acetyl-CoA
was carboxylated to malonyl-CoA in an ATP- and
biotin-requiring process (Eq. 17-39; see also Chapter
13). The carboxyl group formed in this reaction is later
converted back to CO
2
in a decarboxylation (Fig. 17-12).
We know now that in most bacteria and green
plants both an acetyl group of acetyl-CoA and a malo-
nyl group of malonyl-CoA are transferred (steps a
and d of Fig. 17-12) to the sulfur atoms of the phospho-
pantetheine groups of a low-molecular-weight acyl
carrier protein (ACP; Chapter 14). The malonyl
group of the malonyl-ACP is then condensed (step f
of Fig. 17-12) with an acetyl group, which has been
transferred from acetyl-ACP onto a thiol group of the
enzyme (E in Eq. 17-40). The enolate anion indicated
in this equation is generated by decarboxylation of the
malonyl-ACP. It is this decarboxylation that drives the
reaction to completion and, in effect, links CC bond
formation to the cleavage of the ATP required for the
carboxylation step. A related sequence involving multi-
functional proteins is used by animals and fungi
164
(Section J,6).
Carboxylation followed by a later decarboxylation
is an important pattern in other biosynthetic pathways,
too. Sometimes the decarboxylation follows the carboxyl-
ation by many steps. For example, pyruvate (or PEP)
is converted to uridylic acid (Eq. 17-41; details are
shown in Fig. 25-14):
I. Reducing Agents for Biosynthesis
Still another difference between biosynthesis of
fatty acids and oxidation (in mammals) is that the
former has an absolute requirement for NADPH
(Fig. 17-12) while the latter requires NAD
+
and flavo-
proteins (Fig. 17-1). This fact, together with many
other observations, has led to the generalization
that biosynthetic reduction reactions usually require
NADPH rather than NADH. Many measurements
have shown that in the cytosol of eukaryotic cells the
ratio [NADPH]/[NADP
+
] is high, whereas the ratio
[NADH]/[NAD
+
] is low. Thus, the NAD
+
/NADH
system is kept highly oxidized, in line with the role of
NAD
+
as a principal biochemical oxidant, while the
NADP
+
/NADPH system is kept reduced.
The use of NADPH in step g of Fig. 17-12 ensures
that significant amounts of the -oxoacyl-ACP deriva-
tive are reduced to the alcohol. Another difference
between oxidation and biosynthesis is that the alcohol
formed in this reduction step in the biosynthetic process
N
N O
O
COO

O
2
O
3
PO
OH OH
H
Transamination
Orotidine 5'-P
5'-Uridylic acid (UMP)
CO
2
Aspartate
Oxaloacetate
Pyruvate
ATP
ADP + P
i
HCO
3

(17-41)
CH
3
C
O
S CoA
CH
2
C
O
S CoA
HCO
3

ATP

OOC
ADP + P
i
Acetyl-CoA
Activated hydrogens
Group to be removed later
by decarboxylation
Malonyl-CoA
Ch 17IND page - E - 2/14/03, 11:17 AM 978
979
Figure 17-12 The reactions of cytoplasmic biosynthesis of saturated fatty acids. Compare with pathway of oxidation
(Fig. 17-1).
ATP
HCO
3

ADP + P
i
Biotin
R
C
S
O
E
(CH
3
)
ACP-SH
CoA-SH
R
C
C
C
S
O
H H
H H
ACP
R
C
C
C
S
O
ACP
H
H
R
C
C
C
S
O
H H
ACP
O
(CH
3
)
Acyl-ACP;
recycled
each time.
A new 2C
fragment
is added
to the
chain
Acyl-ACP
Enoyl-ACP
H
2
O
NADP
+
NADPH + H
+
NADP
+
NADPH + H
+

O
C
C
C
S
O
H H
ACP
O
Malonyl-ACP
H
2
C
C
S
O

ACP
Stabilized enolate anion
Chain growth is initiated
by transfer of acetyl group
to peripheral SH
group of enzyme
CO
2
CO
2
Photosynthesis
Pyruvate
CO
2
Synthesis of
autotrophic
bacteria
3-Phospho-
glycerate
H
3
C
C
S
O
CoA
Acetyl-CoA

O
C
C
C
S
O
H H
CoA
O
Malonyl-CoA
R
C
C
C
S
O
H H
H H
CoA
Lipids
E-SH
CH
3
C
S
O
ACP
Acetyl-ACP
ACP-SH
of fatty acid synthesis
This pathway is
utilized for initiation
d--Hydroxyacyl-ACP
R
C
C
C
S
O
H H
ACP
H OH
Catabolism of
carbohydrates
Catabolism of
Leu, Ile, Val,
Met, polyphenyl
components
CO
2
f
g
h
i
k
j
d--Oxo-acyl-ACP
c
b
a
e
d
I. Reducing Agents for Biosynthesis
Ch 17IND page - E - 2/14/03, 11:17 AM 979
980 Chapter 17. The Organization of Metabolism
has the D configuration while the corresponding alcohol
in oxidation has the L configuration.
1. Reversing an Oxidative Step with a Strong
Reducing Agent
The second reduction step in biosynthesis of fatty
acids in the rat liver (step i) also required NADPH.
The corresponding step in oxidation utilizes FAD,
but NADPH is a stronger reducing agent than FADH
2
.
Therefore, use of a reduced pyridine nucleotide again
provides a thermodynamic advantage in pushing the
reaction in the biosynthetic direction. Interesting
variations have been observed among different species.
For example, fatty acid synthesis in the rat requires
only NADPH, but the multienzyme complexes from
Mycobacterium phlei, Euglena gracilis, and the yeast
Saccharomyces cerevisiae all give much better synthesis
with a mixture of NADPH and NADH than with
NADPH alone.
165
Apparently, NADPH is required in
step g and NADH in step i. This seems reasonable
because the equilibrium in step i lies far toward the
product formation, and NADH at a very low concen-
tration could carry out the reduction.
2. Regulation of the State of Reduction of the
NAD and NADP Systems
The ratio [NAD
+
]/[NADH] appears to be main-
tained at a relatively constant value and in equilibrium
with a series of different reduced and oxidized substrate
pairs. Thus, it was observed that in the cytoplasm
of rat liver cells, the dehydrogenations catalyzed by
lactate dehydrogenase, sn-glycerol 3-phosphate dehy-
drogenase, and malate dehydrogenase are all at equi-
librium with the same ratio of [NAD
+
]/[NADH].
166
In one experiment rat livers were removed and frozen
in less than 8 s by freeze-clamping (Section L,2)
and the concentrations of different components of
the cytoplasm determined
167
; the ratio [NAD
+
]/
[NADH] was found to be 634, while the ratio of
[lactate]/[pyruvate] was 14.2. From these values an
apparent equilibrium constant for reaction c of Eq.
17-42 was calculated as K
c
= 9.0 x 10
3
. The known
equilibrium constant for the reaction (from in vitro
experiments) is 8.8 x 10
3
(Eq. 17-43). In a similar way
it was shown that several other dehydrogenation
reactions are nearly at equilibrium. This conclusion
has been confirmed more recently by NMR observa-
tions.
168
Now consider Eq. 17-42, step a, the ADP- and P
i
-
requiring oxidation of glyceraldehyde 3-phosphate
(Fig. 15-6). Experimental measurements indicated
that this reaction is also at equilibrium in the cyto-
plasm. In one series of experiments the measured
phosphorylation state ratio [ATP]/[ADP][P
i
] was 709,
while the ratio [3-phosphoglycerate]/[glyceraldehyde
3-phosphate] was 55.5. The overall equilibrium
constant for Eq. 17-42a is given by Eq. 17-44. That
calculated from known equilibrium constants is 60.
From these data Krebs and Veech concluded that the
oxidation state of the NAD system is determined largely
by the phosphorylation state ratio of the adenylate
system.
169
If the ATP level is high the equilibrium in
Eq. 17-42a will be reached at a higher [NAD
+
]/[NADH]
ratio and lactate may be oxidized to pyruvate to adjust
the [lactate]/[pyruvate] ratio.
It is important not to confuse the reactions of Eq.
17-42 as they occur in an aerobic cell with the tightly
coupled pair of redox reactions in the homolactate
fermentation (Fig. 10-3; Eq. 17-19). The reactions of
steps a and c of Eq. 17-42 are essentially at equilibrium,
but the reaction of step b may be relatively slow. Fur-
thermore, pyruvate is utilized in many other metabolic
pathways and ATP is hydrolyzed and converted to ADP
through innumerable processes taking place within
the cell. Reduced NAD does not cycle between the two
enzymes in a stoichiometric way and the reducing
equivalents of NADH formed are, in large measure,
transferred to the mitochondria. The proper view of
the reactions of Eq. 17-42 is that the redox pairs repre-
sent a kind of redox buffer system that poises the
NAD
+
/NADH couple at a ratio appropriate for its
metabolic function.
Glyceraldehyde 3-P
3-Phosphoglycerate Pyruvate
Further
metabolism
Lactate
NAD
+
NADH + H
+
P
i
ADP
ATP
b
c
a
Slower
(17-42)
(17-43)
(17-44)
K
c
'
[
.
(pH 7, 38 C) =
[lactate]
[pyruvate]
NAD ]
[NADH]
=
+

8 8 10
3
K
a
'
[ [
]
/
(pH 7, 38 C) =
[ATP]
[ADP] [P ]
3-phosphoglycerate]
[glyceraldehyde phosphate]
NADH]
[NAD
= 709 55.5
i
+

1 634 62 =
Ch 17IND page - E - 2/14/03, 11:17 AM 980
981 I. Reducing Agents for Biosynthesis
Somewhat surprisingly, within the mitochondria
the ratio [NAD
+
]/[NADH] is 100 times lower than in
the cytoplasm. Even though mitochondria are the site
of oxidation of NADH to NAD
+
, the intense catabolic
activity occurring in the oxidation pathway and the
citric acid cycle ensure extremely rapid production
of NADH. Furthermore, the reduction state of NAD
is apparently buffered by the low potential of the
-hydroxybutyrateacetoacetate couple (Chapter 18,
Section C,2). Mitochondrial pyridine nucleotides also
appear to be at equilibrium with glutamate dehydro-
genase.
169
How is the cytoplasmic [NADPH]/[NADP
+
] ratio
maintained at a value higher than that of [NADH]/
[NAD
+
]? Part of the answer is from operation of the
pentose phosphate pathway (Section E,3). The reac-
tions of Eq. 17-12, if they attained equilibrium, would
give a ratio of cytosolic [NADPH]/[NADP
+
] > 2000 at
0.05 atm CO
2
. Compare this with the ratio 1/634 for
[NADH/[NAD
+
] deduced from the observation on the
reactions of Eq. 17-42.
Consider also the following transhydrogenation
reaction (Eq. 17-45):
NADH + NADP
+
NAD
+
+ NADPH
There are soluble enzymes that catalyze this reaction
for which K equals ~ 1. Within mitochondria an energy-
linked system (Chapter 18) involving the membrane
shifts the equilibrium to favor NADPH. However,
within the cytoplasm, the reaction of Eq. 17-45 is driven
by coupling ATP cleavage to the transhydrogenation
via carboxylation followed by eventual decarboxylation.
One cycle that accomplishes this is given in Eq. 17-46.
The first step (step a) is ATP-dependent carboxylation
of pyruvate to oxaloacetate, a reaction that occurs only
within mitochondria (Eq. 14-3). Oxaloacetate can be
reduced by malate dehydrogenase using NADH (Eq.
17-46, step b), and the resulting malate can be exported
from the mitochondria. In the cytoplasm the malate is
oxidized to pyruvate, with decarboxylation, by the
(17-45)
malic enzyme (Eq. 13-45). The malic enzyme (Eq. 17-46,
step c) is specific for NADP
+
, is very active, and also
appears to operate at or near equilibrium within the
cytoplasm. On this basis, using known equilibrium
constants, it is easy to show that the ratio [NADPH]/
[NADP
+
] will be ~ 10
5
times higher at equilibrium than
the ratio [NADH]/[NAD
+
].
169,170
Since NADPH is continuously used in biosynthetic
reactions, and is thereby reconverted to NADP
+
, the
cycle of Eq. 17-46 must operate continuously. As in Eq.
17-42, a true equilibrium does not exist but steps b and
c are both essentially at equilibrium. These equilibria,
together with those of Eq. 17-42 for the NAD system,
ensure the correct redox potential of both pyridine
nucleotide coenzymes in the cytoplasm.
Malate is not the only form in which C
4
compounds
are exported from mitochondria. Much oxaloacetate
is combined with acetyl-CoA to form citrate; the latter
leaves the mitochondria and is cleaved by the ATP-
dependent citrate-cleaving enzymes (Eq. 13-39). This,
in effect, exports both acetyl-CoA (needed for lipid
synthesis) and oxaloacetate which is reduced to malate
within the cytoplasm. Alternatively, oxaloacetate may
be transaminated to aspartate. The aspartate, after
leaving the mitochondria, may be converted in another
transamination reaction back to oxaloacetate. All of
these are part of the nonequilibrium process by which
C
4
compounds diffuse out of the mitochondria before
completing the reaction sequence of Eq. 17-46 and
entering into other metabolic processes. Note that the
reaction of Eq. 17-46 leads to the export of reducing
equivalents from mitochondria, the opposite of the
process catalyzed by the malateaspartate shuttle
which is discussed in Chapter 18 (Fig. 18-18). The
two processes are presumably active under different
conditions.
While the difference in the redox potential of the
two pyridine nucleotide systems is clear-cut in mam-
malian tissues, in E. coli the apparent potentials of the
two systems are more nearly the same.
171
3. Reduced Ferredoxin in Reductive
Biosynthesis
Both the NAD
+
and NADP
+
systems have standard
electrode potentials E (pH 7) of 0.32 V. However,
because of the differences in concentration ratios, the
NAD
+
system operates at a less negative potential
( 0.24 V) and the NADP
+
system at a more negative
potential ( 0.38 V) within the cytoplasm of eukaryotes.
In green plants and in many bacteria a still more power-
ful reducing agent is available in the form of reduced
ferredoxin. The value of E (pH 7) for clostridial ferre-
doxin is 0.41 V, corresponding to a Gibbs energy
change for the two-electron reduction of a substrate
~ 18 kJ mol
1
more negative than the corresponding
ADP
+
P
i
ATP HCO
3

Pyruvate
Mitochondria
Cytoplasm
Malate
NAD
+
NADH
NADP
+
NADPH CO
2
Malic
enzyme
Pyruvate
Oxaloacetate
c
b
a
(17-46)
Ch 17IND page - E - 2/14/03, 11:18 AM 981
982 Chapter 17. The Organization of Metabolism
value of G for reduction by NADPH. Using reduced
ferredoxin (Fd) some photosynthetic bacteria and
anaerobic bacteria are able to carry out reductions that
are virtually impossible with the pyridine nucleotide
system. For example, pyruvate and 2-oxoglutarate
can be formed from acetyl-CoA (Eq. 15-35) and succinyl-
CoA, respectively (Eq. 17-47).
172173a
In our bodies
the reaction of Eq. 17-47, with NAD
+
as the oxidant,
goes only in the opposite direction and is essentially
irreversible.
J. Constructing the Monomer Units
Now let us consider the synthesis of the monomeric
units from which biopolymers are made. How can
simple one-carbon compounds such as CO
2
and formic
acid be incorporated into complex carbon compounds?
How can carbon chains grow in length or be shortened?
How are branched chains and rings formed?
1. Carbonyl Groups in Chain Formation and
Cleavage
Except for some vitamin B
12
-dependent reactions,
the cleavage or formation of carboncarbon bonds
usually depends upon the participation of carbonyl
groups. For this reason, carbonyl groups have a central
mechanistic role in biosynthesis. The activation of
hydrogen atoms to carbonyl groups permits con-
densations to occur during biosynthesis. Aldol or
Claisen condensations require the participation of two
carbonyl compounds. Carbonyl compounds are also
essential to thiamin diphosphate-dependent condensa-
tions and the aldehyde pyridoxal phosphate is needed
for most C C bond cleavage or formation within
amino acids.
Because of the importance of carbonyl groups to
the mechanism of condensation reactions, much of the
assembly of either straight-chain or branched-carbon
skeletons takes place between compounds in which
the average oxidation state of the carbon atoms is
similar to that in carbohydrates (or in formaldehyde,
H
2
CO). The diversity of chemical reactions possible
with compounds at this state of oxidation is a maximum,
a fact that may explain why carbohydrates and closely
related substances are major biosynthetic precursors
and why the average state of oxidation of the carbon in
most living things is similar to that in carbohydrates.
174
This fact may also be related to the presumed occurrence
of formaldehyde as a principal component of the
earths atmosphere in the past and to the ability of
formaldehyde to condense to form carbohydrates.
In Fig. 17-13 several biochemicals have been
arranged according to the oxidation state of carbon.
Most of the important biosynthetic intermediates lie
within 2 electrons per carbon atom of the oxidation
state of carbohydrates. As the chain length grows,
they tend to fall even closer. It is extremely difficult
to move through enzymatic processes between 2C, 3C,
and 4C compounds (i.e., vertically in Fig. 17-13) except
at the oxidation level of carbohydrates or somewhat
to its right, at a slightly higher oxidation level. On the
other hand, it is often possible to move horizontally
with ease using oxidationreduction reactions. Thus,
fatty acids are assembled from acetate units, which lie
at the same oxidation state as carbohydrates and, after
assembly, are reduced.
Among compounds of the same overall oxidation
state, e.g., acetic acid and sugars, the oxidation states
of individual carbon atoms can be quite different.
Thus, in a sugar every carbon atom can be regarded as
immediately derived from formaldehyde, but in acetic
acid one end has been oxidized to a carboxyl group
and the other has been reduced to a methyl group.
Such internal oxidationreduction reactions play an
important role in the chemical manipulations necessary
to assemble the carbon skeletons needed by a cell.
Decarboxylation is a feature of many biosynthetic
routes. Referring again to Fig. 17-13, notice that many
of the biosynthetic intermediates such as pyruvate,
oxoglutarate, and oxaloacetate are more oxidized than
the carbohydrate level. However, their decarboxylation
products, which become incorporated into the com-
pounds being synthesized, are closer to the oxidation
level of carbohydrates.
2. Starting with CO
2
There are three known pathways by which auto-
trophic organisms can use CO
2
to synthesize triose
phosphates or 3-phosphoglycerate, three-carbon com-
pounds from which all other biochemical substances
can be formed.
175 177
The first of these is the reductive
tricarboxylic cycle. This is a reversal of the oxidative
citric acid cycle in which reduced ferredoxin is used as
a reductant in the reaction of Eq. 17-47 to incorporate
CO
2
into pyruvate. Succinyl-CoA can react with CO
2
in the same type of reaction to form 2-oxoglutarate,
accomplishing the reversal of the only irreversible step
in the citric acid cycle. Using these reactions photosyn-
thetic bacteria and some anaerobes that can generate a
high ratio of reduced to oxidized ferredoxin carry out
the reductive tricarboxylic acid cycle. Together with
Acetyl-CoA
CO
2
Pyruvate

+ CoA
Fd
red
Fd
ox
G' (pH 7) = +17 kJ mol
1
(17-47)
Ch 17IND page - E - 2/14/03, 11:18 AM 982
983
Figure 17-13 Some biochemical compounds arranged in order of average oxidation state of the carbon atoms and by
carbon-chain lengths. Black horizontal arrows mark some biological interconversions among compounds with the same
chain length, while green lines show changes in chain length and are often accompanied by decarboxylation.
HC
COH)
n
O
H
(H
CH
2
OH
(CH
2
)
n
H
COOH
C
H
O)
n
(
Carbohydrates
0 -2 +2 +4 -4 -6
CHO
COH
CH
2
OH
H
0 +4e

CH
4
+2e

2e

4e

CH
3
OH H
2
CO HCOOH CO
2
Multicarbon
compounds
with same
oxidation state
in all internal
carbons
CH
2
OH
COH
CH
2
OH
H
Acetaldehyde Acetic acid Glycine
+6
2C
Dihydroxy-
acetone P
Serine
Glyceraldehyde
4C
Alanine Pyruvic acid Acetone
COOH
CH
2
CH
3
COOH
CH
2
CH
2
OH
COOH
CH
2
CHO
COOH
CH
2
COOH
Malonic
semialdehyde
n-Butanol
Glycerol
Oxaloacetic
acid
COH
COH
CH
3
CH
3
H
H
C
COH
CH
3
CH
3
O
H
C
C
CH
3
CH
3
O
O
CH
2
CH
2
CHO
COOH
Acetoin
Methylmalonic
acid
CO
2
CO
2
COOH
C
CH
2
CH
2
COOH
O
2-Oxoglutaric
acid
Oxalosuccinic
acid
Citric acid
CO
2
5 & 6C
CO
2
Total number of electrons from carbohydrate level
3C
Glyoxylic
acid
COOH
C
CH
2
OH
O
COOH
HC
CH
2
OH
OH
COOH
HC
CH
3
COOH
CH
2
CH
2
CH
2
OH
CH
2
OH
CHO
CH
2
OH
COOH
CH
2
OH
COOH
CHO
COOH
COOH
CH
2
OH
CH
3
CHO
CH
3
COOH
CH
3
COOH
CH
2
NH
2
CH
3
HCOH
CH
3
CH
3
C
CH
3
O
COOH
H
2
NCH
CH
3
COOH
C
CH
3
O
COOH
HCOH
HC O
COOH
C
CH
2
CH
3
COOH
CH
2
HOCH
CH
3
COOH
HC
CH
2
OH
CH
3
COOH
C
CH
2
O
COOH
Malic
acid
Fumaric acid
COOH
CH
COOH
CH
Succinic acid
COOH
CH
2
COOH
CH
2
Acetoacetic acid
COOH
CH
2
CH
3
C O
Valine
COOH
C
CH
3
CH
3
CO
2
CO
2
J. Constructing the Monomer Units
Ch 17IND page - E - 2/14/03, 11:18 AM 983
984 Chapter 17. The Organization of Metabolism
(17-49)
Eq. 17-47, the cycle provides for the complete synthesis
of pyruvate from CO
2
.
178,179
A quantitatively much more important pathway
of CO
2
fixation is the reductive pentose phosphate
pathway (ribulose bisphosphate cycle or Calvin
Benson cycle; Fig. 17-14). This sequence of reactions,
which takes place in the chloroplasts of green plants
and also in many chemiautotrophic bacteria, is essen-
tially a way of reversing the oxidative pentose phos-
phate pathway (Fig. 17-8). The latter accomplishes the
complete oxidation of glucose or of glucose 1-phosphate
by NADP
+
(Eq. 17-48):
Glucose 1-P
2
+ ATP
4
+ 8 H
2
O + 12 NADP
+

6 CO
2
+ 12 NADPH + ADP
3
+ 2HPO
4
2
+ 13 H
+
G (pH 7) = 299 kJ mol
1
It would be almost impossible for a green plant
to fix CO
2
using photochemically generated NADPH
by an exact reversal of Eq. 17-48 because of the high
positive Gibbs energy change. To solve this thermo-
dynamic problem the reductive pentose phosphate
pathway has been modified in a way that couples ATP
cleavage to the synthesis.
The reductive carboxylation system is shown
within the green shaded box of Fig. 17-14. Ribulose
5-phosphate is the starting compound and in the first
step one molecule of ATP is expended to form ribulose
1,5-bisphosphate. The latter is carboxylated and
cleaved to two molecules of 3-phosphoglycerate. This
reaction was discussed in Chapter 13. The reductive
step (step c) of the system employs NADPH together
with ATP. Except for the use of the NADP system
instead of the NAD system, it is exactly the reverse of
the glyceraldehyde phosphate dehydrogenase reaction
of glycolysis. Looking at the first three steps of Fig.
17-14 it is clear that in the reductive pentose phosphate
pathway three molecules of ATP are utilized for each
CO
2
incorporated. On the other hand, in the oxidative
direction no ATP is generated by the operation of the
pentose phosphate pathway.
The reactions enclosed within the shaded box of
Fig. 17-14 do not give the whole story about the cou-
pling mechanism. A phospho group was transferred
from ATP in step a and to complete the hydrolysis it
must be removed in some future step. This is indicated
in a general way in Fig. 17-14 by the reaction steps
d, e, and f. Step f represents the action of specific phos-
phatases that remove phospho groups from the seven-
carbon sedoheptulose bisphosphate and from fructose
bisphosphate. In either case the resulting ketose
monophosphate reacts with an aldose (via transketolase,
step g) to regenerate ribulose 5-phosphate, the CO
2
acceptor. The overall reductive pentose phosphate
cycle (Fig. 17-14B) is easy to understand as a reversal
of the oxidative pentose phosphate pathway in which
the oxidative decarboxylation system of Eq. 17-12 is
replaced by the reductive carboxylation system of Fig.
17-14A. The scheme as written in Fig. 17-14B shows
the incorporation of three molecules of CO
2
. The
reductive carboxylation system operates three times
with a net production of one molecule of triose phos-
phate. As with other biosynthetic cycles, any amount
of any of the intermediate metabolites may be with-
drawn into various biosynthetic pathways without
disruption of the flow through the cycle.
The overall reaction of carbon dioxide reduction
in the CalvinBenson cycle (Fig. 17-14) becomes
6 CO
2
+ 12 NADPH + 18 ATP
4
+ 11 H
2
O
glucose-1-P
2-
+ 12 NADP
+
+ 18 ADP
3
17 HPO
4
2
+ 6 H
+
The Gibbs energy change G (pH 7) is now 357 kJ
mol
1
instead of the + 299 kJ mol
1
required to reverse
the reaction of Eq. 17-48.
The third pathway for reduction of CO
2
to acetyl-
CoA is utilized by acetogenic bacteria, by methanogens,
and probably by sulfate-reducing bacteria.
179 181
This acetyl-CoA pathway (or WoodLjungdahl
pathway) involves reduction by H
2
of one of the
two molecules of CO
2
to the methyl group of methyl-
tetrahydromethanopterin in methanogens and of
methyltetra-hydrofolate in acetogens. The pathway
utilized by methanogens is illustrated in Fig. 15-22.
182
184
A similar process utilizing H
2
as the reductant is
employed by acetogens.
179,185 188a
In both cases a
methyl corrinoid is formed and its methyl group is
condensed with a molecule of carbon monoxide bound
to a copper ion in a NiCu cluster.
189a,b
The resulting
acetyl group is transferred to a molecule of coenzyme
A as illustrated in Eq. 16-52.
189
The bound CO is
formed by reduction of CO
2
, again using H
2
as the
reductant.
190
The overall reaction for acetyl-CoA
synthesis is given by Eq. 17-50. Conversion of acetyl-
CoA to pyruvate via Eq. 17-47 leads into the glucogen-
ic pathway.
(17-48)
CH
3
Pterin
CH
3
Co (Corrin)
CH
3
C
O
S CoA + 3 H
2
O
CO
2
6-Electron reduction
(Fig. 15-22)
CoA-SH CO
CH
3
C
O
S CoA
Carbon monoxide
dehydrogenase
CO CO
2
2e
2 CO
2
+ 4 H
2
+ CoASH
(Eq. 16-52)
Overall:

Ni
Cu
(17-50)
Ch 17IND page - E - 2/14/03, 11:18 AM 984
985
An alternative pathway by which some acetogenic
bacteria form acetate is via reversal of the glycine decar-
boxylase reaction of Fig. 15-20. Methylene-THF is
formed by reduction of CO
2
, and together with NH
3
and CO
2
a lipoamide group of the enzyme and PLP
forms glycine. The latter reacts with a second methylene-
THF to form serine, which can be deaminated to pyru-
vate and assimilated. Methanogens may use similar
pathways but ones that involve methanopterin (Fig.
15-17).
191
3. Biosynthesis from Other Single-Carbon
Compounds
Various bacteria and fungi are able to subsist on
such one-carbon compounds as methane, methanol,
methylamine, formaldehyde, and formate.
192 197
Energy
is obtained by oxidation to CO
2
. Methylotrophic
bacteria initiate oxidation of methane by hydroxylation
(Chapter 18) and dehydrogenate the resulting methanol
or exogenous methanol using the PPQ cofactor (Eq.
15-51).
198
Further dehydrogenation to formate and of
formate to CO
2
via formate dehydrogenase (Eq. 16-63)
completes the process. Some methylotrophic bacteria
incorporate CO
2
for biosynthetic purposes via the
ribulose bisphosphate (CalvinBenson) cycle but
many use pathways that begin with formaldehyde
(or methylene-THF). Others employ variations of
the reductive pentose phosphate pathway to convert
formaldehyde to triose phosphate. In one of these, the
ribulose monophosphate cycle or Quayle cycle,
192,193
ribulose 5-P undergoes an aldol condensation with
formaldehyde to give a 3-oxo-hexulose 6-phosphate
(Eq. 17-51, step a). The latter is isomerized to fructose
6-P (Eq. 17-51, step b). If this equation is applied to the
BOX 17-E
14
C AND THE CALVINBENSON CYCLE
The chemical nature of photosynthesis had
intrigued chemists for decades but little was learned
about the details until radioactive
14
C became avail-
able. Discovered in 1940 by Ruben and Kamen, the
isotope was available in quantity by 1946 as a prod-
uct of nuclear reactors. Initial studies of photosyn-
thesis had been conducted by Ruben and Kamen
using
11
C but
14
C made rapid progress possible. In
1946 Melvin Calvin and Andrew A. Benson began
their studies that elucidated the mechanism of incor-
poration of CO
2
into organic materials.
A key development was two-dimensional paper
chromatography with radioautography (Box 3-C).
A suspension of the alga Chlorella (Fig. 1-11) was
allowed to photosynthesize in air. At a certain time,
a portion of H
14
CO
3
was injected into the system,
and after a few seconds of photosynthesis with
14
C
present the suspension of algae was run into hot
methanol to denature proteins and to stop the reac-
tion. The soluble materials extracted from the algal
cells were concentrated and chromatographed;
radioautographs were then prepared. It was found
that after 10 s of photosynthesis in the presence of
14
CO
2
, the algae contained a dozen or more
14
C
labeled compounds. These included malic acid,
aspartic acid, phosphoenolpyruvate, alanine, triose
phosphates, and other sugar phosphates and diphos-
phates. However, during the first five seconds a single
compound, 3-phosphoglycerate, contained most of the
radioactivity.
a,b
This finding suggested that a two-
carbon regenerating substrate might be carboxylated
by
14
CO
2
to phosphoglycerate. Search for this two-
carbon compound was unsuccessful, but Benson,
in Calvins laboratory, soon identified ribulose
a
Benson, A. A., Bassham, J. A., Calvin, M., Goodale, T. C., Haas,
V. A., and Stepka, W. (1950) J. Am. Chem. Soc. 72, 17101718
b
Benson, A. A. (1951) J. Am. Chem. Soc. 73, 29712972
c
Benson, A. A., Kawaguchi, S., Hayes, P., and Calvin, M. (1952)
J. Am. Chem. Soc. 74, 44774482
d
Bassham, J. A., Benson, A. A., Kay, L. D., Harris, A. Z., Wilson,
A. T., and Calvin, M. (1954) J. Am. Chem. Soc. 76, 17601770
e
Calvin, M., and Bassham, J. A. (1962) The Photosynthesis of
Carbon Compounds, Benjamin, New York
f
Fuller, R. C. (1999) Photosynth Res. 62, 129
bisphosphate,
c
which kinetic studies proved to be
the true regenerating substrate.
c,d
Its carboxylation
and cleavage
e
represent the first step in what has
come to be known as the CalvinBenson cycle (Fig.
17-14).
f
Chromatogram of extract of the alga Scenedesmus after photosyn-
thesis in the presence of
14
CO
2
for 10 s. Courtesy of J. A. Bassham.
The origin of the chromatogram is at the lower right corner.
Dihydroxyacetone phosphate
Ribulose phosphate
Ribose phosphate
Fructose phosphate & mannose phosphate
Glucose phosphate & sedoheptulose phosphate
Ribulose diphosphate & hexose diphosphate
Phosphoenolpyruvate
Phosphoglycerate
J. Constructing the Monomer Units
Ch 17IND page - E - 2/14/03, 11:18 AM 985
986 Chapter 17. The Organization of Metabolism
Ribulose 5-phosphate
ATP
ADP
b
a
CO
2
, H
2
O
Ribulose
bisphosphate
carboxylase
3-Phosphoglycerate
c
2 ATP
2 ADP + 2P
i
2 NADPH
2 NADP
+
R'CHO
Transketolase g
Specific
phosphatase
H
2
O
P
i
R(CHOH)CHO
Recycle
CH
2
OH
C
CH
2
OP
O
Aldolase
R CHO (An aldose phosphate)
CH
2
OP
C O
C HO
C OH
R
H
H
CH
2
OH
C O
C HO
C OH
R
H
H
f
e
d
Products
Carbo-
hydrates,
etc.
CH
2
OH
C
C
O
C
CH
2
OP
OH
OH
H
H
CH
2
OP
C
C
O
C
CH
2
OP
OH
OH
H
H
COO

C
CH
2
OP
OH H 2
CHO
C
CH
2
OP
OH H 2
Figure 17-14 (A) The reductive carboxylation system used in reductive pentose phosphate pathway (CalvinBenson cycle).
The essential reactions of this system are enclosed within the dashed box. Typical subsequent reactions follow. The phosphatase
action completes the phosphorylationdephosphorylation cycle. (B) The reductive pentose phosphate cycle arranged to
show the combining of three CO
2
molecules to form one molecule of triose phosphate. Abbreviations are RCS, reductive
carboxylation system (from above); A, aldolase, Pase, specific phosphatase; and TK, transketolase.
three C
5
sugars three molecules of fructose 6-phosphate
will be formed. One of these can be phosphorylated by
ATP to fructose 1,6-bisphosphate, which can be cleaved
by aldolase. One of the resulting triose phosphates
can then be removed for biosynthesis and the second,
together with the other two molecules of fructose 6-P,
can be recycled through the sugar rearrangement
sequence of Fig. 17-8B to regenerate the three ribulose
5-P molecules that serve as the regenerating substrate.
In bacteria, which lack formate dehydrogenase,
formaldehyde can be oxidized to CO
2
toprovide energy
beginning with the reactions of Eq. 17-51. The result-
ing fructose 6-P is isomerized to glucose 6-P, which is
then dehydrogenated via Eq. 17-12 to form CO
2
and
the regenerating substrate ribulose 5-phosphate.
A number of pseudomonads and other bacteria
convert C
1
compounds to acetate via tetrahydrofolic
acid-bound intermediates and CO
2
using the serine
pathway
179,192,193
shown in Fig. 17-15. This is a cyclic
process for converting one molecule of formaldehyde
(bound to tetrahydrofolate) plus one of CO
2
into acetate.
The regenerating substrate is glyoxylate. Before
condensation with the active formaldehyde of meth-
A B
Growth
C
3
(Glyceraldehyde 3-P)
C
5
C
7
A
C
4
C
5
C
3
C
5
C
3
C
6
TK
P'ase
TK
P'ase
CO
2
CO
2
CO
2
C
3
3 ATP
2 NADPH
Aldolase
3 ATP
2 NADPH
3 ATP
2 NADPH
ylene THF, the glyoxylate undergoes transamination to
glycine (Fig. 17-15, step a). The glycine plus formalde-
hyde forms serine (step b), which is then transaminated
to hydroxypyruvate, again using step a. Glyoxylate
plus formaldehyde could have been joined in a thiamin-
dependent condensation. However, as in the -amino-
butyrate shunt (Fig. 17-5), the coupled transamination
step of Fig 17-15 permits use of PLP-dependent CC
bond formation.
Conversion of hydroxypyruvate to PEP (Fig. 17-15)
involves reduction by NADH and phosphorylation by
ATP to form 3-phosphoglycerate, which is converted
to PEP as in glycolysis. The conversion of malate to
acetate and glyoxylate via malyl-CoA and isocitrate
lyase (Eq. 13-40) forms the product acetate and regen-
erates glyoxylate. As with other metabolic cycles,
various intermediates, such as PEP, can be withdrawn
for biosynthesis. However, there must be an indepen-
dent route of synthesis of the regenerating substrate
glyoxylate. One way in which this can be accomplished
is to form glycine via the reversal of the glycine decar-
boxylase pathway as is indicated by the shaded green
lines in Fig. 17-15.
Ch 17IND page - E - 2/14/03, 11:18 AM 986
987
4. The Glyoxylate Pathways
The reductive carboxylation of acetyl-CoA to
pyruvate (Eq. 17-47) occurs only in a few types of
bacteria. For most species, from microorganisms to
animals, the oxidative decarboxylation of pyruvate
to acetyl-CoA is irreversible. This fact has many
important consequences. For example, carbohydrate
is readily converted to fat; because of the irreversibility
of this process, excess calories lead to the deposition
of fats. However, in animals fat cannot be used to
generate most of the biosynthetic intermediates needed
for formation of carbohydrates and proteins because
those intermediates originate largely from C
3
units.
This limitation on the conversion of C
2
acetyl units
to C
3
metabolites is overcome in many organisms by
J. Constructing the Monomer Units
CH
2
OH
C
C
C
Ribulose 5-P
H OH
CH
2
OH H
O
O P
C
C
C
C
Hexulose 6-P
H OH
CH
2
OH H
O
O P
CH
2
OH
H OH
CH
2
OH
b
Isomerase
C
C
C
C
Fructose 6-P
H OH
CH
2
OH H
HO
O P
CH
2
OH
O
H
CH
2
OH
a
H
C O
H
(17-51)
Figure 17-15 One of the serine pathways for assimilation of one-carbon compounds.
CH
2
O (Formaldehyde)
(Fig. 15-43)
2 Tetrahydrofolate
5,10-Methylene-THF
(Fig. 15-20)
THF
(Fig. 15-18)
Serine
Aminotransferase
Hydroxypyruvate
Glycerate
Biosynthesis
Lyase
Glyoxylate
Acetyl-CoA
Malate
Oxaloacetate
PEP
Glycine
NADH
ATP
3-Phosphoglycerate
Malyl-CoA
ATP
NADH
NH
4
+
CO
2
Overall: CH
2
O + CO
2
+ 2NADH acetate

+ H
+
+ H
2
O
CO
2
P
i
(Eq. 13-53)
Acetate

(Eq. 17-52)
ATP
O
C
CH
2
HO
COO

C
CH
2
O
3
PO
COO

H OH
2
OOC
C
CH
2
C
S
CoA
O
H OH

b
a
Ch 17IND page - E - 2/14/03, 11:18 AM 987
988 Chapter 17. The Organization of Metabolism
Fatty acids
oxidation
Aspartate
Oxaloacetate
Citrate synthase
Citrate
Aconitase
Succinate
Pyruvate
L-malate
Isocitrate
2-Oxoglutarate
Glutamate
Oxaloacetate
NAD
+
Malate Oxidation
part of citric
acid cycle
H
2
O
Fumarate
FAD*
c
cleavage
Glyoxylate
h
Succinate
Regulated
in E. coli
CO
2
2-Oxo-
glutarate

Acetyl-CoA
Malate synthase d
CO
2
2[H]
Oxaloacetate Gluconeogenesis
fungi
Citrate
Acetyl-CoA
a
CYTOSOL
MITOCHONDRION
Aspartate
e
f
g
h
b
a
i
GLYOXYSOME
j
Isocitrate lyase
Acetyl-CoA
CH
2
C
C
O
COO

H
C H H
OH
O
COO

C
O
COO

H
Figure 17-16 The glyoxylate pathway. The green line traces the pathway of labeled carbon from fatty acids or acetyl-CoA
into malate and other products.
the glyoxylate cycle (Fig. 17-16), which converts two
acetyl units into one C
4
unit. The cycle provides a way
for organisms, such as E. coli,
111,199
Saccharomyces,
200
Tetrahymena, and the nematode Caenorhabditis,
201
to
subsist on acetate as a sole or major carbon source.
It is especially prominent in plants that store large
amounts of fat in their seeds (oil seeds). In the
germinating oil seed the glyoxylate cycle allows fat
to be converted rapidly to sucrose, cellulose, and other
carbohydrates needed for growth.
A key enzyme in the glyoxylate cycle is isocitrate
lyase, which cleaves isocitrate (Eq. 13-40) to succinate
and glyoxylate. The latter is condensed with a sec-
ond acetyl group by the action of malate synthase
(Eq. 13-38). The L-malate formed in this reaction is
dehydrogenated to the regenerating substrate oxalo-
acetate. Some of the reaction steps are those of the
citric acid cycle and it appears that in bacteria there is
no spatial separation of the citric acid cycle and glyox-
ylate pathway. However, in plants the enzymes of the
glyoxylate cycle are present in specialized peroxisomes
known as glyoxysomes.
60
The glyoxysomes also con-
tain the enzymes for oxidation of fatty acids, allow-
ing for efficient conversion of fatty acids to succinate.
This compound is exported from the glyoxysomes and
enters the mitochondria where it undergoes oxidation
to oxaloacetate. The latter can be converted by PEP
carboxylase (Eq. 13-53) or by PEP carboxykinase
(Eq. 13-46) to PEP.
An acetyl-CoA-glyoxylate cycle, which catalyzes
oxidation of acetyl groups to glyoxylate, can also be
constructed from isocitrate lyase and citric acid cycle
Ch 17IND page - E - 2/14/03, 11:18 AM 988
989
(17-53)
enzymes. Glyoxylate is taken out of the cycle as the
product and succinate is recycled (Eq. 17-52). The
independent pathway for synthesis of the regenerating
substrate oxaloacetate is condensation of glyoxylate
with acetyl-CoA (malate synthetase) to form malate
and oxidation of the latter to oxaloacetate as in the
main cycle of Fig. 17-16.
5. Biosynthesis of Glucose from Three-Carbon
Compounds
Now let us consider the further conversion of PEP
and of the triose phosphates to glucose 1-phosphate,
the key intermediate in biosynthesis of other sugars
and polysaccharides. The conversion of PEP to glucose
1-P represents a reversal of part of the glycolysis
sequence. It is convenient to discuss this along with
gluconeogenesis, the reversal of the complete glyco-
lysis sequence from lactic acid. This is an essential
part of the Cori cycle (Section F) in our own bodies, and
the same process may be used to convert pyruvate
derived from deamination of alanine or serine (Chapter
24) into carbohydrates.
Just as with the pentose phosphate cycle, an exact
reversal of the glycolysis sequence (Eq. 17-53) is pre-
cluded on thermodynamic grounds. Even at very high
values of the phosphorylation state ratio R
p
, the reaction:
2 Lactate

+ 3 ATP
4
+ 2 H
2
O C
6
H
10
O
5
(glycogen)
+ 3 ADP
3
+ 3 HPO
4
2
+ H
+
G (pH 7) = + 107 kJ per glycosyl unit
would be unlikely to go to completion. The actual
pathways used for gluconeogenesis (Fig. 17-17, green
lines) differ from those of glycolysis (black lines) in
three significant ways. First, while glycogen breakdown
is initiated by the reaction with inorganic phosphate
catalyzed by phosphorylase (Fig. 17-17, step a), the
biosynthetic sequence from glucose 1-P, via uridine
diphosphate glucose (Fig. 17-17, step b; see also Eq.
17-56), is coupled to cleavage of ATP. Second, in the
catabolic process (glycolysis) fructose 6-P is converted
to fructose 1,6-P
2
through the action of a kinase (Fig.
17-17, step c), which is then cleaved by aldolase. The
resulting triose phosphate is degraded to PEP. In
glucogenesis a phosphatase is used to form fructose P
from fructose P
2
(Fig. 17-17, step d). Third, during
gly-colysis PEP is converted to pyruvate by a kinase
with generation of ATP (Fig. 17-17, step e). During
glucogenesis pyruvate is converted to PEP indirectly
via oxaloacetate (Fig. 17-17, steps f and g) using pyru-
vate carboxylase (Eq. 14-3) and PEP carboxykinase
(Eq. 13-46). This is another example of the coupling
of ATP cleavage through a carboxylationdecarboxyl-
ation sequence. The net effect is to use two molecules
of ATP (actually one ATP and one GTP) rather than one
to convert pyruvate to PEP.
The overall reaction for reversal of glycolysis to
form glycogen (Eq. 17-54) now has a comfortably
negative standard Gibbs energy change as a result
of coupling the cleavage of 7 ATP to the reaction.
2 Lactate

+ 7 ATP
4
+ 6 H
2
O
glycogen + 7 ADP
3
+ 7 HPO
4
2
+ 5 H
+
G (pH 7) = 31 kJ mol
1
per glycosyl unit
J. Constructing the Monomer Units
Acetyl-CoA
Citrate
Isocitrate
Glyoxylate
Succinate
oxidation
Oxaloacetate
(17-52)
(17-54)
Figure 17-17 Comparison of glycolytic pathway (left) with
pathway of gluconeogenesis (right, green arrows).
Glycogen
Glucose 1-P
Glucose 6-P
Fructose 6-P
Fructose 1,6-bisphosphate
UDP-glucose
UDP
UTP
ATP
ADP
P
i
Phosphorylase
Triose phosphates
ADP
ATP + P
i
ADP
ATP + P
i
Phosphoenolpyruvate (PEP)
ADP
ATP
Pyruvate
Further
metabolism
HCO
3

ATP ADP + P
i
Oxaloacetate
NADH (or NADPH)
b a
PP
i
2 P
i
ADP
ATP P
i
Phosphatase
H
2
O
d c Kinase
NAD
+
Lactate,
Alanine,
etc.
Precursors
f
g
e
Ch 17IND page - E - 2/14/03, 11:18 AM 989
990 Chapter 17. The Organization of Metabolism
reactions for fatty acid biosynthesis. Why does
oxidation require CoA derivatives while biosynthesis
requires the more complex acyl carrier protein (ACP)?
The reason may involve control. ACP is a complex
handle able to hold the growing fatty acid chain and
to guide it from one enzyme to the next. In E. coli the
various enzymes catalyzing the reactions of Fig. 17-12
are found in the cytosol and behave as independent
proteins. The same is true for fatty acid synthases of
higher plants which resemble those of bacteria.
205,205a
It is thought that the ACP molecule lies at the
center of the complex and that the growing fatty acid
chain on the end of the phosphopantetheine prosthetic
group moves from one subunit to the other.
164,206
The
process is started by a primer which is usually acetyl-
CoA in E. coli. Its acyl group is transferred first to the
central molecule of ACP (step a, Fig. 17-12) and then to
a peripheral thiol group, probably that of a cysteine
side chain on a separate protein subunit (step b, Fig.
17-12). Next, a malonyl group is transferred (step d)
from malonyl-CoA to the free thiol group on the ACP.
The condensation (steps e and f ) occurs with the freeing
of the peripheral thiol group. The latter does not come
into use again until the -oxoacyl group formed has
undergone the complete sequence of reduction reactions
(steps gi). Then the growing chain is again transferred
to the peripheral SH (step j) and a new malonyl unit
is introduced on the central ACP.
After the chain reaches a length of 12 carbon atoms,
the acyl group tends to be transferred off to a CoA
molecule (step k) rather than to pass around the cycle
again. Thus, chain growth is terminated. This tendency
systematically increases as the chain grows longer.
In higher animals as well as in Mycobacterium,
207
yeast,
208
and Euglena, the fatty acid synthase consists
of only one or two multifunctional proteins. The
synthase from animal tissues has seven catalytic
activities in a single 263-kDa 2500-residue protein.
209
The protein consists of a series of domains that contain
the various catalytic activities needed for the entire
synthetic sequence. One domain contains an ACP-like
site with a bound 4'-phosphopantetheine as well as a
cysteine side chain in the second acylation site. This
synthase produces free fatty acids, principally the C
16
palmitate. The final step is cleavage of the acyl-CoA
by a thioesterase, one of the seven enzymatic activities
of the synthase. See Chapter 21 for further discussion.
7. The Oxoacid Chain Elongation Process
As mentioned in Section 4, glyoxylate can be
converted to oxaloacetate by condensation with
acetyl-CoA (Fig. 17-16) and the oxaloacetate can be
decarboxylated to pyruvate. This sequence of reactions
resembles that of the conversion of oxaloacetate to
2-oxoglutarate in the citric acid cycle (Fig. 17-4). Both
Two enzymes that are able to convert pyruvate
directly to PEP are found in some bacteria and plants.
In each case, as in the animal enzyme system discussed
in the preceding paragraph, the conversion involves
expenditure of two high-energy linkages of ATP. The
PEP synthase of E. coli first transfers a pyrophospho
group from ATP onto an imidazole group of histidine
in the enzyme (Eq. 17-55). A phospho group is hydro-
lyzed from this intermediate (dashed green line in Eq.
17-55, step b), ensuring that sufficient intermediate
EHisP is present. The latter reacts with pyruvate
to form PEP.
202,203
Pyruvatephosphate dikinase
is a similar enzyme first identified in tropical grasses
and known to play an important role in the CO
2
concentrating system of the so-called C
4
plants
(Chapter 23).
204
The same enzyme participates in
gluconeogenesis in Acetobacter. The reaction cycle
for this enzyme is also portrayed in Eq. 17-55. In this
case P
i
, rather than water, is the attacking nucleophile
in Eq. 17-55 and PP
i
is a product. The latter is probably
hydrolyzed by pyrophosphatase action, the end result
being an overall reaction that is the same as with PEP
synthase. Kinetic and positional isotope exchange
studies suggest that the P
i
must be bound to pyruvate
phosphate dikinase before the bound ATP can react
with the imidazole group.
202
Likewise, AMP doesnt
dissociate until P
i
has reacted to form PP
i
.
6. Building Hydrocarbon Chains with
Two-Carbon Units
Fatty acid chains are taken apart two carbon atoms
at a time by oxidation. Biosynthesis of fatty acids
reverses this process by using the two-carbon acetyl
unit of acetyl-CoA as a starting material. The coupling
of ATP cleavage to this process by a carboxylation
decarboxylation sequence, the role of acyl carrier protein
(Section H,4), and the use of NADPH as a reductant
(Section I) have been discussed and are summarized
in Fig. 17-12, which gives the complete sequence of
ATP
AMP

E-His-P

-P

E-His-P

Pyruvate
PEP
P P
i
P
i
2 P
i
H
2
O
H
2
O
PEP Synthase
P
i
E-His
Pyruvate-
phosphate
dikinase
reaction
cycle
a
b
c
(17-55)
Ch 17IND page - E - 2/14/03, 11:18 AM 990
991
are examples of a frequently used general chain elongation
process for -oxo acids. This sequence, which is illustrated
in Fig. 17-18, has four steps: (1) condensation of the
-oxo acid with an acetyl group, (2) isomerization by
dehydration and rehydration (catalyzed by aconitase
in the case of the citric acid cycle), (3) dehydrogenation,
and (4) decarboxylation. In many cases steps 3 and
4 are combined as a single enzymatic reaction. The
isomerization of the intermediate hydroxy acid in step
2 is required because the hydroxyl group, which is
attached to a tertiary carbon bearing no hydrogen,
must be moved to the adjacent carbon atom before
oxidation to a ketone can take place. However, in the
case of glyoxylate, isomerization is not necessary
because R = H.
It may be protested that the reaction of the citric
acid cycle by which oxaloacetate is converted to oxo-
glutarate does not follow exactly the pattern of Fig.
17-18. The carbon dioxide removed in the decarboxy-
lation step does not come from the part of the molecule
donated by the acetyl group but from that formed from
oxaloacetate. However, the end result is the same.
Furthermore, there are two known citrate-forming
enzymes with different stereospecificities (Chapter 13),
one of which leads to a biosynthetic pathway strictly
according to the sequence of Fig. 17-18.
At the bottom of Fig. 17-18 several stages of the
-oxo acid elongation process are arranged in tan-
dem. We see that glyoxylate (a product of the acetyl-
CoAglyoxylate cycle) can be built up systematically to
J. Constructing the Monomer Units
R C
O
COO

H
3
C C
O
S CoA
R C COO

C HO COO

H
H
R C COO

C O COO

H
R CH
2
C
O
COO

CoASH
R C
OH
COO

CH
2
COO

Isomerization by
dehydration and
rehydration
-Decarboxylation
NAD
+
NADH
Examples:
H C
O
COO

C C
O
COO

H
2
C C
O
COO

CH
2
C
O
COO

OOC CH
2

OOC
CH
2
CH
2
C
O
COO

CH
2
CH
2
C
O
COO

OOC

OOC
CH C
O
COO

H
3
C
H
3
C
CH C
O
COO

Glyoxylate
Pyruvate
Alanine
H
3
C
H
3
C
Oxaloacetate 2-Oxoglutarate
CH
2
Glutamic acid
2-Oxoglutarate
H
Lysine
Leucine
H
CH
2
H
Figure 17-18 The oxoacid chain elongation process.
Ch 17IND page - E - 2/14/03, 11:18 AM 991
992 Chapter 17. The Organization of Metabolism
pyruvate, oxaloacetate, 2-oxoglutarate, and 2-oxoadipate
(a precursor of lysine) using this one reaction sequence.
Methanogens elongate 2-oxoadipate by one and two
carbon atoms using the same sequence to give 7- and
8-carbon dicarboxylates.
210
8. Decarboxylation as a Driving Force in
Biosynthesis
Consider the relationship of the following promi-
nent biosynthetic intermediates one to another:
Utilization of acetyl-CoA for the synthesis of long-chain
fatty acids occurs via carboxylation to malonyl-CoA.
We can think of the malonyl group as a -carboxylated acetyl
group. During synthesis of a fatty acid the carboxyl
group is lost, and only the acetyl group is ultimately
incorporated into the fatty acid. In a similar way
pyruvate can be thought of as an -carboxylated acetaldehyde
and oxaloacetate as an - and -dicarboxylated acetaldehyde.
During biosynthetic reactions these three- and four-
carbon compounds also often undergo decarboxylation.
Thus, they both can be regarded as activated acetalde-
hyde units. Phosphoenolpyruvate is an -carboxylated
phosphoenol form of acetaldehyde and undergoes both
decarboxylation and dephosphorylation before con-
tributing a two-carbon unit to the final product.
It is of interest to compare two chain elongation
processes by which two-carbon units are combined.
In the synthesis of fatty acids the acetyl units are con-
densed and then are reduced to form straight hydro-
carbon chains. In the oxo-acid chain elongation
mechanism, the acetyl unit is introduced but is later
decarboxylated. Thus, the chain is increased in length
by one carbon atom at a time. These two mechanisms
account for a great deal of the biosynthesis by chain
extension. However, there are other variations. For
example, glycine (a carboxylated methylamine), under
the influence of pyridoxal phosphate and with accom-
panying decarboxylation, condenses with succinyl-CoA
(Eq. 14-32) to extend the carbon chain and at the same
time to introduce an amino group. Likewise, serine
(a carboxylated ethanolamine) condenses with
palmitoyl-CoA in biosynthesis of sphingosine (as in
Eq. 14-32). Phosphatidylserine is decarboxylated to
phosphatidylethanolamine in the final synthetic step
for that phospholipid (Fig. 21-5).
9. Stabilization and Termination of Chain
Growth by Ring Formation
Biochemical substances frequently undergo cycliza-
tion to form stable five- and six-atom ring structures.
The three-carbon glyceraldehyde phosphate exists in
solution primarily as the free aldehyde (and its cova-
lent hydrate) but glucose 6-phosphate exists largely
as the cyclic hemiacetal. In this ring form no carbonyl
group is present and further chain elongation is inhib-
ited. When the hemiacetal of glucose 6-P is enzymati-
cally isomerized to glucose 1-P the ring is firmly locked.
Glucose 1-P, in turn, serves as the biosynthetic precursor
of polysaccharides and related compounds, in all of
which the sugar rings are stable. Ring formation can
occur in lipid biosynthesis, too. Among the polyketides
(Chapter 21), polyprenyl compounds (Chapter 22),
and aromatic amino acids (Chapter 25) are many
substances in which ring formation has occurred by
ester or aldol condensations followed by reduction
and elimination processes. This is a typical sequence
for biosynthesis of highly stable aromatic rings.
10. Branched Carbon Chains
Branched carbon skeletons are formed by standard
reaction types but sometimes with addition of rear-
rangement steps. Compare the biosynthetic routes to
three different branched five-carbon units (Fig. 17-19)
The first is the use of a propionyl group to initiate
formation of a branched-chain fatty acid. Propionyl-
CoA is carboxylated to methylmalonyl-CoA, whose
acyl group is transferred to the acyl carrier protein
before condensation. Decarboxylation and reduction
yields an acyl-CoA derivative with a methyl group in
the 3-position.
The second five-carbon branched unit, in which
the branch is one carbon further down the chain, is
an intermediate in the biosynthesis of polyprenyl
(isoprenoid) compounds and steroids. Three two-
carbon units are used as the starting material with
decarboxylation of one unit. Two acetyl units are first
condensed to form acetoacetyl-CoA. Then a third
acetyl unit, which has been transferred from acetyl-
CoA onto an SH group of the enzyme, is combined
with the acetoacetyl-CoA through an ester condensa-
tion. The thioester linkage to the enzyme is hydrolyzed
to free the product 3-hydroxy-3-methylglutaryl-CoA
(HMG-CoA). This sequence is illustrated in Eq. 17-5.
The thioester group of HMG-CoA is reduced to the
C
CH
3
S-CoA O
C
H
2
C
S-CoA O
COOH
C
CH
3
COO

O
C
CH
2
COO

O P
C
H
2
C
COO

O
Acetyl-CoA
Pyruvate
Malonyl-CoA
PEP
COOH
Oxaloacetate (OAA)
Ch 17IND page - E - 2/14/03, 11:18 AM 992
993
alcohol mevalonic acid, a direct
precursor to isopentenyl pyro-
phosphate, from which the poly-
prenyl compounds are formed
(Fig. 22-1).
The third type of carbon-
branched unit is 2-oxoisovalerate,
from which valine is formed by
transamination. The starting units
are two molecules of pyruvate
which combine in a thiamin
diphosphate-dependent conden-
sation with decarboxylation. The
resulting -acetolactate contains a
branched chain but is quite unsuit-
able for formation of an amino
acid. A rearrangement moves the
methyl group to the position
(Fig. 24-17), and elimination of
water from the diol forms the enol
of the desired -oxo acid (Fig. 17-19).
The precursor of isoleucine is
formed in an analogous way by
condensation, with decarboxylation
of one molecule of pyruvate with
one of 2-oxobutyrate.
K. Biosynthesis and
Modification of Polymers
There are three chemical prob-
lems associated with the assembly
of a protein, nucleic acid, or other
biopolymer. The first is to overcome
thermodynamic barriers. The second
is to control the rate of synthesis, and
the third is to establish the pattern or
sequence in which the monomer units
are linked together. Let us look briefly
at how these three problems are
dealt with by living cells.
1. Peptides and Proteins
Activation of amino acids for
incorporation into oligopeptides
and proteins can occur via two
routes of acyl activation. In the first
of these an acyl phosphate (or acyl
adenylate) is formed and reacts with
an amino group to form a peptide
linkage (Eq. 13-4). The tripeptide
glutathione is formed in two steps
of this type (Box 11-B). In the second
method of activation aminoacyl
K. Biosynthesis and Modification of Polymers
O
C S CoA
CH
2
H
3
C Propionyl-CoA
HCO
3

Biotin
ATP
O
C S ACP
C
H
3
C

OOC
H
C
O
H
3
C
S E
Acetyl-enzyme
H
3
C
C
C
C
S ACP
O O
H
3
C H
Methylmalonyl-ACP
CO
2
O
C
H
3
C CH
2
C S CoA
O
H
3
C
C
O
S CoA
2 Acetyl-CoA
CH
2
C
C

OOC S
O
CoA
OH H
3
C
HMG-CoA
CH
2
C
CH
2

OOC CH
2
OH
OH H
3
C
Mevalonate
C CH
2
CH
2
O P P
Isopentenyl pyrophosphate
H
2
C
H
3
C
H
3
C C
COO

O
C
COO

CH
3
O
CO
2 H
3
C
C
C
COO

O
HO CH
3
H
3
C C
COO

O
H H
3
C
Valine
(Isoleucine)
-Acetolactate
1. Starter piece for branched-chain fatty acids
2. Polyisoprenoid compounds
3. Branched-chain amino acids
Figure 17-19 Biosynthetic origins of three five-carbon branched structural
units. Notice that decarboxylation is involved in driving each sequence.
Ch 17IND page - E - 2/14/03, 11:18 AM 993
994 Chapter 17. The Organization of Metabolism
adenylates are formed. They transfer their activated
aminoacyl groups onto specific tRNA molecules dur-
ing synthesis of proteins (Eq. 17-36). In other cases
activated aminoacyl groups are transferred onto SH
groups to form intermediate thioesters. An example
is the synthesis of the antibiotic gramicidin S formed
by Bacillus brevis. The antibiotic is a cyclic decapeptide
with the following five-amino-acid sequence repeated
twice in the ringlike molecule
211
:
(d-Phel-Pro l-Val l-Ornl-Leu )
2
The soluble enzyme system responsible for its
synthesis contains a large 280-kDa protein that not only
activates the amino acids as aminoacyl adenylates and
transfers them to thiol groups of 4'-phosphopantetheine
groups covalently attached to the enzyme but also
serves as a template for joining the amino acids in
proper sequence.
211 214
Four amino acidsproline,
valine, ornithine (Orn), and leucineare all bound.
A second enzyme (of mass 100 kDa) is needed for acti-
vation of phenylalanine. It is apparently the activated
phenylalanine (which at some point in the process is
isomerized from L- to D-phenylalanine) that initiates
polymer formation in a manner analogous to that of
fatty acid elongation (Fig. 17-12). Initiation occurs
when the amino group of the activated phenylalanine
(on the second enzyme) attacks the acyl group of the
aminoacyl thioester by which the activated proline is
held. Next, the freed imino group of proline attacks
the activated valine, etc., to form the pentapeptide.
Then two pentapeptides are joined and cyclized to
give the antibiotic. The sequence is absolutely specific,
and it is remarkable that this relatively small enzyme
system is able to carry out each step in the proper
sequence. Many other peptide antibiotics, such as the
bacitracins, tyrocidines,
215
and enniatins, are synthesized
in a similar way,
213,216,217
as are depsipeptides and the
immunosuppresant cyclosporin. A virtually identical
pattern is observed for formation of polyketides,
218,219
whose chemistry is considered in Chapter 21.
While peptide antibiotics are synthesized according
to enzyme-controlled polymerization patterns, both
proteins and nucleic acids are made by template
mechanisms. The sequence of their monomer units
is determined by genetically encoded information. A
key reaction in the formation of proteins is the transfer
of activated aminoacyl groups to molecules of tRNA
(Eq. 17-36). The tRNAs act as carriers or adapters as
explained in detail in Chapter 29. Each aminoacyl-
tRNA synthetase must recognize the correct tRNA
and attach the correct amino acid to it. The tRNA then
carries the activated amino acid to a ribosome, where
it is placed, at the correct moment, in the active site.
Peptidyltransferase, using a transacylation reaction,
in an insertion mechanism transfers the C terminus of
the growing peptide chain onto the amino group of
the new amino acid to give a tRNA-bound peptide one
unit longer than before.
2. Polysaccharides
Incorporation of a sugar monomer into a polysac-
charide also involves cleavage of two high-energy
phosphate linkages of ATP. However, the activation
process has its own distinctive pattern (Eq. 17-56).
Usually a sugar is first phosphorylated by a kinase
or a kinase plus a phosphomutase (Eq. 17-56, step a).
Then a nucleoside triphosphate (NuTP) reacts under
the influence of a second enzyme with elimination of
pyrophosphate and formation of a glycopyranosyl
ester of the nucleoside diphosphate, more often
known as a sugar nucleotide (Eq. 17-56, step b).
The inorganic pyrophosphate is hydrolyzed by pyro-
phosphatase while the sugar nucleotide donates the
activated glycosyl group for polymerization (Eq. 17-56,
step c). In this step the glycosyl group is transferred
with displacement of the nucleoside diphosphate.
Thus, the overall process involves first the cleavage of
ATP to ADP and P
i
, and then the cleavage of a nucleo-
side triphosphate to a nucleoside diphosphate plus P
i
.
The nucleoside triphosphate in Eq. 17-56, step b is
sometimes ATP, in which case the overall result is the
splitting of two molecules of ATP to ADP. However,
as detailed in Chapter 20, the whole series of nucleotide
handles serve to carry various activated glycosyl units.
What determines the pattern of incorporation of
sugar units into polysaccharides? Homopolysaccharides,
like cellulose and the linear amylose form of starch,
contain only one monosaccharide component in only
one type of linkage. A single synthetase enzyme can
add unit after unit of an activated sugar (UDP glucose
or other sugar nucleotide) to the growing end. However,
at least two enzymes are needed to assemble a branched
molecule such as that of the glycogen molecule. One
is the synthetase; the second is a branching enzyme,
a transglycosylase. After the chain ends attain a length
of about ten monosaccharide units the branching
enzyme attacks a glycosidic linkage somewhere in the
chain. Acting much like a hydrolase, it forms a glyco-
syl enzyme (or a stabilized carbocation) intermediate.
The enzyme does not release the severed chain frag-
ment but transfers it to another nearby site on the
branched polymer. In the synthesis of glycogen, the
chain fragment is joined to a free 6-hydroxyl group
of the glycogen, creating a new branch attached by an
-1,6-linkage.
Other carbohydrate polymers consist of repeating
oligosaccharide units. Thus, in hyaluronan units of
glucuronic acid and N-acetyl-D-glucosamine alternate
(Fig. 4-11). The O antigens of bacterial cell coats
(p. 180) contain repeating subunits made up of a
block of four or five different sugars. In these and
Ch 17IND page - E - 2/14/03, 11:18 AM 994
995
many other cases the pattern of polymerization is
established by the specificities of individual enzymes.
An enzyme capable of joining an activated glucosyl
unit to a growing polysaccharide will do so only if
the proper structure has been built up to that point.
In cases where a block of sugar units is transferred
it is usually inserted at the nonreducing end of the
polymer, which may be covalently attached to a
protein. Notice that the insertion mode of chain
growth exists for lipids, polysaccharides, and proteins.
3. Nucleic Acids
The activated nucleotides are the nucleoside
5'-triphosphates. The ribonucleotides ATP, GTP,
UTP, and CTP are needed for RNA synthesis and the
2'-deoxyribonucleotide triphosphates, dATP, dTTP,
dGTP, and dCTP for DNA synthesis. In every case,
the addition of activated monomer units to a growing
polynucleotide chain is catalyzed by an enzyme that
binds to the template nucleic acid. The choice of the
proper nucleotide unit to place next in the growing
strand is determined by the nucleotide already in place
in the complementary strand, a matter that is dealt
with in Chapters 27 and 28. The chemistry is a simple
displacement of pyrophosphate (Eq. 17-57). The
3'-hydroxyl of the polynucleotide attacks the phos-
phorus atom of the activated nucleoside triphosphate.
Thus, nucleotide chains always grow from the 5' end, with
new units being added at the 3' end.
4. Phospholipids and PhosphateSugar
Alcohol Polymers
Choline and ethanolamine are activated in much
the same way as are sugars. For example, choline can
be phosphorylated using ATP (Eq. 17-58, step a) and
the phosphocholine formed can be further converted
(Eq. 17-58, step b) to cytidine diphosphate choline.
Phosphocholine is transferred from the latter onto a
suitable acceptor to form the final product (Eq. 17-58,
step c). The polymerization pattern differs from that
for polysaccharide synthesis. When the sugar nucleo-
tides react, the entire nucleoside diphosphate is
eliminated (Eq. 17-56), but CDP-choline and CDP-
ethanolamine react with elimination of CMP (Eq.
17-58, step c), leaving one phospho group in the final
product. The same thing is true in the synthesis of
the bacterial teichoic acids (Chapter 8). Either CDP-
glycerol or CDP-ribitol is formed first and polymeriza-
tion takes place with elimination of CMP to form the
alternating phosphatesugar alcohol polymer.
220
5. Irreversible Modification and Catabolism
of Polymers
While polymers are being synthesized continuously
by cells, they are also being modified and torn down.
Nothing within a cell is static. As discussed in Chapters
O
O PO
3
2
ATP
ADP
Kinase a
b
Nucleotide triphosphate
PP
i
2 P
i
H
2
O
Glycopyranosyl ester of
nucleoside diphosphate,
e.g., UDP-glucose
O
O
H
H
One or more steps
O
O
Acceptor
Linked glycosyl unit
Nucleoside diphosphate
c
O
O P
O
O
O
H
Acceptor end of
polysaccharide
O
O Nucleoside

P
O
O

(17-56)
K. Biosynthesis and Modification of Polymers
PP
i
HO Nucleotide O P
O
O
O

P
O
O
O

P
O

O
OH
HO Nu O P
O
O
O

3' 5'
Polynucleotide
3'
O Polynucleotide
5'
5'
H
H
2
O
2 P
i
(17-57)
Ch 17IND page - E - 2/14/03, 11:18 AM 995
996 Chapter 17. The Organization of Metabolism
10 and 29, everything turns over at a slower or faster
rate. Hydrolases attack all of the polymers of which
cells are composed, and active catabolic reactions de-
grade the monomers formed. Membrane surfaces are
also altered, for example, by hydroxylation and glyco-
sylation of both glycoproteins and lipid head groups.
It is impossible to list all of the known modification
reactions of biopolymers. They include hydrolysis,
methylation, acylation, isopentenylation, phospho-
rylation, sulfation, and hydroxylation. Precursor
molecules are cut and trimmed and often modified
further to form functional proteins or nucleic acids.
Phosphotransferase reactions splice RNA transcripts
to form mRNA and a host of alterations convert pre-
cursors into mature tRNA molecules (Chapter 28).
Even DNA, which remains relatively unaltered,
undergoes a barrage of chemical attacks. Only because
of the presence of an array of repair enzymes (Chapter
27) does our DNA remain nearly unchanged so that
faithful copies can be provided to each cell in our
bodies and can be passed on to new generations.
L. Regulation of Biosynthesis
A simplified view of metabolism is to consider a
cell as a bag of enzymes. Indeed, much of metabo-
lism can be explained by the action of several thousand
enzymes promoting specific reactions of their substrates.
These reactions are based upon the natural chemical
reactivities of the substrates. However, the enzymes,
through the specificity of their actions and through
association with each other,
96,221223
channel the reactions
into a selected series of metabolic pathways. The
reactions are often organized as cycles which are
inherently stable. We have seen that biosynthesis
often involves ATP-dependent reductive reactions.
It is these reductive processes that produce the less reactive
nonpolar lipid groupings and amino acid side chains so
essential to the assembly of insoluble intracellular structures.
Oligomeric proteins, membranes, microtubules, and
filaments are all the natural result of aggregation
caused largely by hydrophobic interactions with
electrostatic forces and hydrogen bonding providing
specificity. A major part of metabolism is the creation
of complex molecules that aggregate spontaneously
to generate structure. This structure includes the
lipid-rich cytoplasmic membranes which, together
with embedded carrier proteins, control the entry of
substances into cells. Clearly, the cell is now much
more than a bag of enzymes, containing several com-
partments, each of which contains its own array
of enzymes and other components. Metabolite con-
centrations may vary greatly from one compartment
to another.
The reactions that modify lipids and glycoproteins
provide a driving force that assists in moving mem-
brane materials generated internally into the outer
surface of cells. Other processes, including the break-
down by lysosomal enzymes, help to recycle mem-
brane materials. Oxidative attack on hydrophobic
materials such as the sterols and the fatty acids of
membrane lipids results in their conversion into more
soluble substances which can be degraded and com-
pletely oxidized. The flow of matter within cells tends
to occur in metabolic loops and some of these loops
lead to formation of membranes and organelles and to
their turnover. This flow of matter, which is responsible
for growth and development of cells, is driven both
by hydrolysis of ATP coupled to biosynthesis and by
irreversible degradative alterations of polymers and
lipid materials. It also provides for transient formation
and breakup of complexes of macromolecules, which
may be very large, in response to varying metabolic
needs.
Anything that affects the rate of a reaction involved
in either biosynthesis or degradation of any component
of the cell will affect the overall picture in some way.
Thus, every chemical reaction that contributes to a
quantitatively significant extent to metabolism has
(17-58)
H
3
C N
+
H
3
C
H
3
C
CH
2
CH
2
OH
N
+
CH
2
CH
2
O PO
3
2
N
+
CH
2
CH
2
O P
O
O
O

P
O
O
O

cytidine
N
+
CH
2
CH
2
O P
O

O Y
O
Choline
ATP
ADP
CTP
PP
i
H
2
O
2 P
i
Cytidine diphosphate choline
YOH
CMP
Phospholipid
(phosphatidylcholine if Y is a diglyceride)
Phosphocholine acceptor
such as the hydroxyl
group of a diglyceride
c
b
a
Ch 17IND page - E - 2/14/03, 11:19 AM 996
997
their energy through oxidation of glucose.
226,227
For
this reason, the glucose level of blood cannot be allowed
to drop much below the normal 5 mM. The mechanism
of regulation of the blood glucose level is complex and
incompletely understood. A series of hormones are
involved.
Insulin. This 51-residue cross-linked polypeptide
(Fig. 7-17) is synthesized in the pancreatic islets of
Langerhans, a tissue specialized for synthesis and
secretion into the bloodstream of a series of small
peptide hormones. One type of islet cells, the cells,
forms primarily insulin which is secreted in response
to high (> 5mm) blood [glucose].
228
Insulin has a wide
range of effects on metabolism,
228a
which are discussed
in Chapter 11, Section G. Most of these effects are
thought to arise from binding to insulin receptors
(Figs. 11-11 and 11-12) and are mediated by cascades
such as that pictured in Fig. 11-13.
229232c
The end result
is to increase or decrease activities of a large number
of enzymes as is indicated in Table 17-3. Some of
those are also shown in Fig. 17-20, which indicates
interactions with the tricarboxylic acid cycle and lipid
metabolism. Binding of insulin to the extracellular
domain of its dimeric receptor induces a conforma-
tional change that activates the intracellular tyrosine
kinase domains of the two subunits. Recent studies
suggest that in the activated receptor the two trans-
membrane helices and the internal tyrosine kinase
domains move closer together, inducing the essential
autophosphosphorylation.
232b
The kinase domain of
the phosphorylated receptor, in turn, phosphorylates
several additional proteins, the most important of
which seem to be the insulin receptor substrates IRS-1
and IRS-2. Both appear to be essential in different
tissues.
232d,e
Phosphorylated forms of these proteins
initiate a confusing variety of signaling cascades.
232fi
One of the most immediate effects of insulin
is to stimulate an increased rate of uptake of glucose
by muscle and adipocytes (fat cells) and other insulin-
sensitive tissues. This uptake is accomplished largely
by movement of the glucose transporter GLUT4
(Chapter 8) from internal sequestered storage vesicles
located near the cell membrane into functioning
positions in the membranes.
232f,j m
Activation of this
translocation process apparently involves IRS-1 and
phosphatidylinositol (PI) 3-kinase, which generates
PI-3,4,5-P
3
(Fig. 11-9).
232c,n,o
The latter induces the
translocation. However, the mechanism remains
obscure. The process may also require a second
signaling pathway which involves action of the insulin
receptor kinase on an adapter protein known as CAP,
a transmembrane caveolar protein flotillin, and a
third protein Cbl, a known cellular protooncogene.
Phosphorylated Cbl forms a complex with CAP and
flotillin in a lipid raft which induces the exocytosis
of the sequestered GLUT4 molecules.
232o,p
some controlling influence. Since molecules interact
with each other in so many ways, reactions of metabolic
control are innumerable. Small molecules act on macro-
molecules as effectors that influence conformation and
reactivity. Enzymes act on each other to break covalent
bonds, to oxidize, and to crosslink. Transferases add
phospho, glycosyl, methyl, and other groups to various
sites. The resulting alterations often affect catalytic
activities. The number of such interactions significant
to metabolic control within an organism may be in the
millions. Small wonder that biochemical journals are
filled with a confusing number of postulated control
mechanisms.
Despite this complexity, some regulatory mecha-
nisms stand out clearly. The control of enzyme synthesis
through feedback repression and the rapid control of
activity by feedback inhibition (Chapter 11) have been
considered previously. Under some circumstances, in
which there is a constant growth rate, these controls
may be sufficient to ensure the harmonious and pro-
portional increase of all constitutents of a cell. Such
may be the case for bacteria during logarithmic growth
(Box 9-B) or for a mammalian embryo growing rapidly
and drawing all its nutrients from the relatively constant
supply in the maternal blood.
Contrast the situation in an adult. Little growth
takes place, but the metabolism must vary with time
and physiological state. The body must make drastic
readjustments from normal feeding to a starvation
situation and from resting to heavy exercise. The
metabolism needed for rapid exertion is different from
that needed for sustained work. A fatty diet requires
different metabolism than a high-carbohydrate diet.
The necessary control mechanisms must be rapid and
sensitive.
1. Glycogen and Blood Glucose
Two special features of glucose metabolism in
animals are dominant.
224
The first is the storage of
glycogen for use in providing muscular energy rapidly.
This is a relatively short-term matter but the rate of
glycolysis can be intense: The entire glycogen content
of muscle could be exhausted in only 20 s of anaerobic
fermentation or in 3.5 min of oxidative metabolism.
225
There must be a way to turn on glycolysis quickly and
to turn it off when it is no longer needed. At the same
time, it must be possible to reconvert lactate to glucose
or glycogen (gluconeogenesis). The glycogen stores of
the muscle must be repleted from glucose of the blood.
If insufficient glucose is available from the diet or from
the glycogen stores of the liver, it must be synthesized
from amino acids.
The second special feature of glucose metabolism
is that certain tissues, including brain, blood cells,
kidney medulla, and testis, ordinarily obtain most of
L. Regulation of Biosynthesis
Ch 17IND page - E - 2/14/03, 11:19 AM 997
998 Chapter 17. The Organization of Metabolism
A. Activity increased
Enzymes of glycolysis
Glucokinase Transcription induced
Phosphofructokinase via 2,6-fructose P
2
Pyruvate kinase Dephosphorylation
6-Phosphofructo-2-kinase Dephosphorylation
Enzymes of glycogen synthesis
Glucokinase Transcription
Glycogen synthase (muscle) Dephosphorylation
Enzymes of lipid synthesis
Pyruvate dehydrogenase (adipose) Dephosphorylation (Eq. 17-9)
Acetyl-CoA carboxylase Dephosphorylation
ATP-citrate lyase Phosphorylation
Fatty acid synthase
Lipoprotein lipase
Hydroxymethylglutaryl-CoA reductase
B. Activity decreased
Enzymes of gluconeogenesis
Pyruvate carboxylase
PEP carboxykinase Transcription inhibited
Fructose 1,6-bisphosphate
Glucose 6-phosphatase
Enzymes of lipolysis
Triglyceride lipase
(hormone-sensitive lipase) Dephosphorylation
Enzymes of glycogenolysis
Glycogen phosphorylase
C. Other proteins affected by insulin
Glucose transporter GLUT4 Redistribution
Ribosomal protein S6 Phosphorylation by p90
rsk
IGF-II receptor Redistribution
Transferrin receptor Redistribution
Calmodulin Phosphorylation
TABLE 17-3
Some Effects of Insulin on Enzymes
Name of Enzyme Type of Regulation
A clue to another possible unrecognized mecha-
nism of action for insulin comes from the observation
that urine of patients with non-insulin-dependent
diabetes contains an unusual isomer of inositol,
D-chiro-inositol.
233,234
Plasma of such individuals contains
an antagonist of insulin action, an
inositol phosphoglycan containing
myo-inositol as a cyclic 1,2-phosphate
ester and galactosamine and man-
nose in a 1:1:3 ratio.
235
This appears
to be related to the glycosyl phos-
phatidylinositol (GPI) membrane
anchors (Fig. 8-13). It has been
suggested that such a glycan, perhaps
containing chiro-inositol, is released
in response to insulin and serves as a
second messenger for insulin.
235236a
This hypothesis remains unproved.
237
However, insulin does greatly stim-
ulate a GPI-specific phospholipase
C, at least in yeast.
237a
Another
uncertainty surrounds the possible
cooperation of chromium (Chapter 16)
in the action of insulin.
How do the insulin-secreting
pancreatic cells sense a high
blood glucose concentration? Two
specialized proteins appear to be
involved. The sugar transporter
GLUT2 allows the glucose in blood
to equilibrate with the free glucose
in the cells,
237b
while glucokinase
(hexokinase IV or D) apparently
serves as the glucose sensor.
228,238
Despite the fact that glucokinase
is a monomer, it displays a coopera-
tive behavior toward glucose bind-
ing, having a low affinity at low
[glucose] and a high affinity at high
[glucose]. Mutant mice lacking the glucokinase gene
develop early onset diabetes which is mild in hetero-
zygotes but severe and fatal within a week of birth for
homozygotes.
239,240
These facts alone do not explain
how the sensor works and there are doubtless other
components to the signaling system.
HO
OH
HO
OH
OH
OH
1
OH
OH
OH
HO
4
3
6
myo-Inositol
(numbered as d)
1
2
3
4
5
6
d-chiro-Inositol
OH
2
HO
5
Ch 17IND page - E - 2/14/03, 11:19 AM 998
999
A current theory is that the increased rate of glucose
catabolism in the cells when blood [glucose] is high
leads to a high ratio of [ATP]/[ADP] which induces
closure of ATP-sensitive K
+
channels and opening of
voltage-gated Ca
2+
channels.
241
This could explain the
increase in [Ca
2+
] within cells which has been associ-
ated with secretion of insulin
242,243
and which is
thought to induce the exocytosis in insulin storage
granules. The internal [Ca
2+
] in pancreatic islet cells
is observed to oscillate in a characteristic way that is
synchronized with insulin secretion.
243
Glucagon. This 29-residue peptide is the principal
hormone that counteracts the action of insulin. Gluca-
gon acts primarily on liver cells (hepatocytes) and
adipose tissue and is secreted by the cells of the
islets of Langerhans in the pancreas, the same tissue
whose cells produce insulin, if the blood glucose
concentration falls much below 2 mM.
244 250
Like the
insulin-secreting cells, the pancreatic cells contain
glucokinase, which may be involved in sensing the
drop in glucose concentration. However, the carrier
GLUT2 is not present and there is scant information
on the sensing mechanism.
248
Glucagon promotes an increase in the blood glu-
cose level by stimulating breakdown of liver glycogen,
by inhibiting its synthesis, and by stimulating gluco-
neogenesis. All of these effects are mediated by cyclic
AMP through cAMP-activated protein kinase (Fig. 11-4)
and through fructose 2,6-P
2
(Fig. 11-4 and next section).
Glucagon also has a strong effect in promoting the
release of glucose into the bloodstream. Adrenaline
has similar effects, again mediated by cAMP. However,
glucagon affects the liver, while adrenaline affects
many tissues. Glucocorticoids such as cortisol
(Chapter 22) also promote gluconeogenesis and the
accumulation of glycogen in the liver through their
action on gene transcription.
The release of glucose from the glycogen stores
in the liver is mediated by glucose 6-phosphatase,
which is apparently embedded within the membranes
of the endoplasmic reticulum. A labile enzyme, it
consists of a 357-residue catalytic subunit,
251,252
which
may be associated with other subunits that participate
in transport.
252,253
A deficiency of this enzyme causes
the very severe type 1a glycogen storage disease
(see Box 20-D).
251,253
Only hepatocytes have significant
glucose 6-phosphatase activity.
2. Phosphofructo-1-Kinase in the Regulation
of Glycolysis
The metabolic interconversions of glucose 1-P,
glucose 6-P, and fructose 6-P are thought to be at or
near equilibrium within most cells. However, the
phosphorylation by ATP of fructose 6-P to fructose 1,6-P
2
catalyzed by phosphofructose-1-kinase (Fig. 11-2, step b;
Fig. 17-17, top center) is usually far from equilibrium.
This fact was established by comparing the mass
action ratio [fructose 1,6-P
2
] [ADP]/[fructose 6-P] [ATP]
measured within tissues with the known equilibrium
constant for the reaction. At equilibrium this mass
action ratio should be equal to the equilibrium constant
(Section I,2). The experimental techniques for deter-
mining the four metabolite concentrations that are
needed for evaluation of the mass action ratio in tissues
are of interest. The tissues must be frozen very rapidly.
This can be done by compressing them between large
liquid nitrogen-cooled aluminum clamps. For details
see Newsholme and Start,
225
pp. 30 32. Tissues can be
cooled to 80C in less than 0.1 s in this manner. The
frozen tissue is then powdered, treated with a frozen
protein denaturant such as perchloric acid, and analyzed.
From data obtained in this way, a mass action ratio of
0.03 was found for the phosphofructo-1-kinase reaction
in heart muscle.
225
This is much lower than the equi-
librium constant of over 3000 calculated from the
value of G (pH 7) = 20.1 kJ mol
1
. Thus, like other
biochemical reactions that are nearly irreversible thermo-
dynamically, this reaction is far from equilibrium in
tissues.
The effects of ATP, AMP, and fructose 2,6-bisphos-
phate on phosphofructokinase have been discussed
in Chapter 11, Section C. Fructose 2,6-P
2
is a potent
allosteric activator of phosphofructokinase and a strong
competitive inhibitor of fructose 1,6-bisphosphatase
(Fig. 11-2). It is formed from fructose 6-P and ATP
by the 90-kDa bifunctional phosphofructo-2-kinase/
fructose 2,6-bisphosphatase. Thus, the same protein
forms and destroys this allosteric effector. Since the
bifunctional enzyme is present in very small amounts,
the rate of ATP destruction from the substrate cycling
is small.
Glucagon causes the concentration of liver fructose
2,6-P
2
to drop precipitously from its normal value.
This, in turn, causes a rapid drop in glycolysis rate
and shifts metabolism toward gluconeogenesis. At the
same time, liver glycogen breakdown is inceased and
glucose is released into the bloodstream more rapidly.
The effect on fructose 2,6-P
2
is mediated by a cAMP-
dependent protein kinase which phosphorylates the
bifunctional kinase/phosphatase in the liver.
254
This
modification greatly reduces the kinase activity and
strongly activates the phosphatase, thereby destroying
the fructose 2,6-P
2
. The changes in activity appear
to be largely a result of changes in the appropriate
K
m
values which are increased for fructose 6-P and
decreased for fructose 2,6-P
2
.
255
L. Regulation of Biosynthesis
Ch 17IND page - E - 2/14/03, 11:19 AM 999
1000 Chapter 17. The Organization of Metabolism
3. Gluconeogenesis
If a large amount of lactate enters the liver, it is
oxidized to pyruvate which enters the mitochondria.
There, part of it is oxidized through the tricarboxylic
acid cycle. However, if [ATP] is high, pyruvate dehy-
drogenase is inactivated by phosphorylation (Eq. 17-9)
and the amount of pyruvate converted to oxaloacetate
and malate (Eq. 17-46) may increase. Malate may leave
the mitochondrion to be reoxidized to oxaloacetate,
which is then converted to PEP and on to glycogen
(heavy green arrows in Fig. 17-20). When [ATP] is
high, phosphofructokinase is also blocked, but the
fructose 1,6-bisphosphatase, which hydrolyzes one
phosphate group from fructose 1,6-P
2
(Fig. 11-2, step
d), is active. If the glucose content of blood is low, the
glucose 6-P in the liver is hydrolyzed and free glucose
is secreted. Otherwise, most of the glucose 6-P is
converted to glycogen. Muscle is almost devoid of
glucose 6-phosphatase, the export of glucose not being
a normal activity of that tissue.
Gluconeogenesis in liver is strongly promoted by
glucagon and adrenaline. The effects, mediated by
cAMP, include stimulation of fructose 1,6-bisphospha-
tase and inhibition of phosphofructo-1-kinase, both
caused by the drop in the level of fructose 2,6-P
2
.
254,256
The conversion of pyruvate to PEP via oxaloacetate is
also promoted by glucagon. This occurs primarily by
stimulation of pyruvate carboxylase (Eq. 14-3).
257,258
However, it has been suggested that the most impor-
tant mechanism by which glucagon enhances gluco-
neogenesis is through stimulation of mitochondrial
respiration, which in turn may promote gluconeo-
genesis.
257
The conversion of oxaloacetate to PEP by PEP-
carboxykinase (PEPCK, Eq. 14-43; Fig. 17-20) is
another control point in gluconeogenesis. Insulin
inhibits gluconeogenesis by decreasing transcription
of the mRNA for this enzyme.
259 261a
Glucagon and
cAMP stimulate its transcription. The activity of PEP
carboxykinase
262
is also enhanced by Mn
2 +
and by
very low concentrations of Fe
2+
. However, the enzyme
is readily inactivated by Fe
2 +
and oxygen.
263
Any
regulatory significance is uncertain.
Although the regulation of gluconeogenesis in
the liver may appear to be well understood, some
data indicate that the process can occur efficiently
in the presence of high average concentrations of
fructose 2,6-P
2
. A possible explanation is that liver
consists of several types of cells, which may contain
differing concentrations of this inhibitor of gluconeo-
genesis.
264
However, mass spectroscopic studies
suggest that glucose metabolism is similar throughout
the liver.
265
4. Substrate Cycles
The joint actions of phosphofructokinase and
fructose 1,6-bisphosphatase (Fig. 11-2, steps b and c;
see also Fig. 17-20) create a substrate cycle of the type
discussed in Chapter 11, Section F. Such cycles appar-
ently accomplish nothing but the cleavage of ATP to
ADP and P
i
(ATPase activity). There are many cycles
of this type in metabolism and the fact that they do not
ordinarily cause a disastrously rapid loss of ATP is a
consequence of the tight control of the metabolic path-
ways involved. In general, only one of the two enzymes
of Fig. 11-2, steps b and c, is fully activated at any time.
Depending upon the metabolic state of the cell, degra-
dation may occur with little biosynthesis or biosynthesis
with little degradation. Other obvious substrate cycles
involve the conversion of glucose to glucose 6-P and
hydrolysis of the latter back to glucose (Fig. 17-20,
upper left-hand corner), the synthesis and breakdown
of glycogen (upper right), and the conversion of PEP
to pyruvate and the reconversion of the latter to PEP
via oxaloacetate and malate (partially within the mito-
chondria).
While one might suppose that cells always keep
substrate cycling to a bare minimum, experimental
measurements on tissues in vivo have indicated sur-
prisingly high rates for the fructose 1,6-bisphosphatase
phosphofructokinase cycle in mammalian tissues
when glycolytic flux rates are low and also for the
pyruvate oxaloacetate PEP pyruvate cycle.
266
As pointed out in Chapter 11, by maintaining a low
rate of substrate cycling under conditions in which the
carbon flux is low (in either the glycolytic or gluco-
genic direction) the system is more sensitive to allos-
teric effectors than it would be otherwise. However,
when the flux through the glycolysis pathway is high
the relative amount of cycling is much less and the
amount of ATP formed approaches the theoretical 2.0
per glucose.
267
Substrate cycles generate heat, a property that is
apparently put to good use by cold bumblebees whose
thoracic temperature must reach at least 30C before
they can fly. The insects apparently use the fructose
bisphosphatasephosphofructokinase substrate cycle
(Fig. 11-2, steps b and c) to warm their flight muscles.
268
It probably helps to keep us warm, too.
5. Nuclear Magnetic Resonance, Isotopomer
Analysis, and Modeling of Metabolism
As as been pointed out in Boxes 3-C and 17-C,
the use of
13
C and other isotopic tracers together with
NMR and mass spectroscopy have provided powerful
tools for understanding the complex interrelationships
among the various interlocking pathways of metabo-
lism. In Box 17-C the application of
13
C NMR to the
Ch 17IND page - E - 2/14/03, 11:19 AM 1000
1001
Figure 17-20 The interlocking pathways of glycolysis, gluconeogenesis, and fatty acid oxidation and synthesis with indications
of some aspects of control in hepatic tissues. ( ) Reactions of glycolysis, fatty acid degradation, and oxidation by the citric
acid cycle. ( ) Biosynthetic pathways. Some effects of insulin via indirect action on enzymes + , , or on transcription + ,
. Effects of glucagon + , .
Oxaloacetate
OAA
Malate
OAA
(oxaloacetate)
2-Oxoglutarate
Glucose Glucose 6-P
H
2
O
P
i
Fructose 6-P
Fru-2,6-P
2
H
2
O
P
i
ATP
ADP
Fructose 1,6-P
2
Glucose 1-P
P
i
UTP
Glycogen
Triglycerides
Malonyl-CoA
Acetyl-CoA
Lactate,
Amino
acids
PEP
6 steps
GDP
GTP
Aspartate
Pyruvate
CO
2
Citrate
HCO
3

Cytosol
Pyruvate
CO
2
HCO
3

Acetyl-CoA
Citrate
Isocitrate Citric
acid
cycle
Asp
Malate
Fumarate
Succinate
Beta
oxidation
Mitochondrion
CO
2
CO
2
NADPH
NADP
+
Fatty acid
synthase
Fatty acids
NAD
+
ATP
H
2
O
Malate
+
+

Gluco-
neogenesis
pathway
+
Succinyl-
CoA
Porphyrins
+

+
+

HMG-CoA
Mevalonate
ATP
+
Citrate
+
+
+

PEPCK
+

+
L. Regulation of Biosysnthesis
citric acid cycle was described. Similar approaches
have been used to provide direct measurement of the
glucose concentration in human brain (1.0 0.1 mM;
4.7 0.3 mM in plasma)
226
and to study gluconeo-
genesis
269 271
as well as fermentation.
271a
Similar
investigations have been made using mass spectros-
copy.
272
The metabolism of acetate through the
glycoylate pathway in yeast has been observed by
13
C NMR.
200
Data obtained from such experiments
are being used in attempts to model metabolism and
to understand how flux rates through the various
pathways are altered in response to varying condi-
tions.
65,273 276
Ch 17IND page - E - 2/14/03, 11:19 AM 1001
1002 Chapter 17. The Organization of Metabolism
BOX 17-F LACTIC ACIDEMIA AND OTHER DEFICIENCIES IN CARBOHYDRATE METABOLISM
The lactate concentration in blood can rise from
its normal value of 1 2 mM to as much as 22 mM
after very severe exercise such as sprinting, but it
gradually returns to normal, requiring up to 68 h,
less if mild exercise is continued. However, continu-
ously high lactic acid levels are observed when
enzymes of the gluconeogenic pathway are deficient
or when oxidation of pyruvate is partially blocked.
a,b
Severe and often lethal deficiencies of the four key
gluconeogenic enzymes pyruvate carboxylase, PEP
carboxykinase, fructose 1,6-bisphosphatase, and
glucose 6-phosphatase are known.
b
Pyruvate car-
boxylase deficiency may be caused by a defective
carboxylase protein, by an absence of the enzyme
that attaches biotin covalently to the three mitochon-
drial biotin-containing carboxylases (Chapter 14,
Section C), or by defective transport of biotin from
the gut into the blood. The latter types of deficien-
cy can be treated successfully with 10 mg biotin
per day.
Deficiency of pyruvate dehydrogenase is the
most frequent cause of lactic acidemia.
a,c
Since
this enzyme has several components (Fig. 15-15), a
number of forms of the disease have been observed.
Patients are benefitted somewhat by a high-fat, low-
carbohydrate diet. Transient lactic acidemia may
result from infections or from heart failure. One
treatment is to administer dichloroacetate, which
stimulates increased activity of pyruvate dehydro-
genase, while action is also taken to correct the
underlying illness.
d
Another problem arises if a
lactate transporter is defective so that lactic acid
accumulates in muscles.
e
A different problem results from deficiency of
enzymes of glycolysis such as phosphofructokinase
(see Box 20-D), phosphoglycerate mutase, and pyru-
vate kinase. Lack of one isoenzyme of phospho-
glycerate mutase in muscle leads to intolerance to
strenuous exercise.
f
A deficiency in pyruvate kinase
is one of the most common defects of glycolysis in
erythrocytes and leads to a shortened erythrocyte
lifetime and hereditary hemolytic anemia.
g
Deficiency of the first enzyme of the pentose
phosphate pathway, glucose 6-phosphate dehydro-
genase, is widespread.
h
Its geographical distribu-
tion suggests that, like the sickle-cell trait, it confers
some resistance to malaria. A partial deficiency of
6-phosphogluconolactonase (Eq. 17-12, step b) has
also been detected within a family and may have
contributed to the observed hemolytic anemia.
i
a
Robinson, B. H. (1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly,
W. S., and Valle, D., eds), pp. 14791499, McGraw-Hill, New York
b
Robinson, B. H. (1982) Trends Biochem. Sci. 7, 151153
c
McCartney, R. G., Sanderson, S. J., and Lindsay, J. G. (1997)
Biochemistry 36, 68196826
d
Stacpoole, P. W., 17 other authors, and Dichloroacetate-Lactic
Acidosis Study Group (1992) N. Engl. J. Med. 327, 15641569
e
Fishbein, W. N. (1986) Science 234, 12541256
f
DiMauro, S., Mirando, A. F., Khan, S., Gitlin, K., and Friedman,
R. (1981) Science 212, 12771279
g
Tanaka, K. R., and Paglia, D. E. (1995) in The Metabolic and
Molecular Bases of Inherited Disease (Scriver, C. R., Beaudet, A. L.,
Sly, W. S., and Valle, D., eds), pp. 34853511, McGraw-Hill,
New York
h
Pandolfi, P. P., Sonati, F., Rivi, R., Mason, P., Grosveld, F., and
Luzzatto, L. (1995) EMBO J. 14, 52095215
i
Beutler, E., Kuhl, W., and Gelbart, J. (1985) Proc. Natl. Acad. Sci.
U.S.A. 82, 38763878
6. The Fasting State
During prolonged fasting, glycogen supplies are
depleted throughout the body and fats become the
principal fuels. Both glucose and pyruvate are in short
supply. While the hydrolysis of lipids provides some
glycerol (which is phosphorylated and oxidized to
dihydroxyacetone-P), the quantity of glucose precursors
formed in this way is limited. Since the animal body
cannot reconvert acetyl-CoA to pyruvate, there is a
continuing need for both glucose and pyruvate. The
former is needed for biosynthetic processes, and the
latter is a precursor of oxaloacetate, the regenerating
substrate of the citric acid cycle. For this reason, during
fasting the body readjusts its metabolism. As much as
75% of the glucose need of the brain can gradually be
replaced by ketone bodies derived from the breakdown
of fats (Section A,4).
277
Glucocorticoids (e.g., cortisol;
Chapter 22) are released from the adrenal glands. By
inducing enzyme synthesis, these hormones increase
the amounts of a variety of enzymes within the cells
of target organs such as the liver. Glucocorticoids also
appear to increase the sensitivity of cell responses to
cAMP and hence to hormones such as glucagon.
268
The overall effects of glucocorticoids include an
increased release of glucose from the liver (increased
activity of glucose 6-phosphatase), an elevated blood
glucose and liver glycogen, and a decreased synthesis
of mucopolysaccharides. The reincorporation of amino
acids released by protein degradation is inhibited
and synthesis of enzymes degrading amino acids is
enhanced. Among these enzymes are tyrosine and
alanine aminotransferases, enzymes that initiate amino
acid degradation which gives rise to the glucogenic
precursors fumarate and pyruvate.
The inability of the animal body to form the glucose
Ch 17IND page - E - 2/14/03, 11:19 AM 1002
1003
BOX 17-G DIABETES MELLITUS
The most prevalent metabolic problem affecting
human beings is diabetes mellitus.
ac
Out of a million
people about 400 develop type I (or juvenile-onset)
insulin-dependent diabetes mellitus (IDDM)
between the ages of 8 and 12. Another 33,000 (over
3%) develop diabetes by age 40 50, and by the late
70s over 7% are affected. A propensity toward
diabetes is partially hereditary, and recessive sus-
ceptibility genes are present in a high proportion
of the population. The severity of the disease varies
greatly. About half of the type I patients can be
treated by diet alone, while the other half must
receive regular insulin injections because of the
atrophy of the insulin-producing cells of the pan-
creas. Type I diabetes sometimes develops very
rapidly with only a few days of ravenous hunger
and unquenchable thirst before the onset of keto-
acidosis. Without proper care death can follow
quickly. This suggested that a virus infection might
cause the observed death of the insulin-secreting
cells of the pancreatic islets. However, the disease
appears to be a direct result of an autoimmune
response (Chapter 31). Antibodies directed against
such proteins as insulin, glutamate decarboxyl-
ase,
d,e
and a tyrosine phosphatase
f
of the patients
own body are present in the blood. There may also
be a direct attack on the cells by T cells of the
immune system (see Chapter 31).
g,h
The events
that trigger such autoimmune attacks are not clear,
but there is a strong correlation with susceptibility
genes, in both human beings
i,j
and mice.
k,l
Adults seldom develop type I diabetes but often
suffer from type II or non-insulin-dependent
diabetes mellitus (NIDDM). This is not a single
disease but a syndrome with many causes. There is
usually a marked decrease in sensitivity to insulin
(referred to as insulin resistance) and poor uptake
and utilization of glucose by muscles.
m
In rare
cases this is a result of a mutation in the gene for
the insulin molecule precursors (Eq. 10-8) or in the
gene regulatory regions of the DNA.
n,o
Splicing of
the mRNA
p
may be faulty or there may be defects in
the structure or in the mechanisms of activation of
the insulin receptors (Figs. 11-11 and 11-12).
q
The
number of receptors may be too low or they may be
degraded too fast to be effective. About 15% of
persons with NIDDM have mutations in the insulin
substrate protein IRS-1 (Chapter 11, Section G) but
the significance is not clear.
m,r
Likewise, the causes
of the loss of sensitivity of insulin receptors as well
as other aspects of insulin resistance are still poorly
understood.
s
In addition, prolonged high glucose
concentrations result in decreased insulin synthesis
or secretion, both of which are also complex processes.
After synthesis the insulin hexamer is stored as
granules of the hexamer (insulin)
6
Zn
2
(Fig. 7-18)
in vesicles at low pH. For secretion to occur the
vesicles must first dock at membrane sites and
undergo exocytosis. The insulin dissolves, releasing
the Zn
2+
, and acts in the monomeric form.
t
Because
the mechanisms of action of insulin are still not fully
understood, it is difficult to interpret the results of
the many studies of diabetes mellitus.
A striking symptom of diabetes is the high blood
glucose level which may range from 8 to 60 mM.
Lower values are more typical for mild diabetes
because when the glucose concentration exceeds the
renal threshold of ~ 8 mM the excess is secreted into
the urine. Defective utilization of glucose seems to
be tied to a failure of glucose to exert proper
L. Regulation of Biosysnthesis
precursors pyruvate or oxaloacetate from acetyl units
is sometimes a cause of severe metabolic problems.
Ketosis, which was discussed in Section A,4, develops
when too much acetyl-CoA is produced and not
efficiently oxidized in the citric acid cycle. Ketosis
occurs during starvation, with fevers, and in insulin-
dependent diabetes (see also Box 17-G). In cattle, whose
metabolism is based much more on acetate than is
ours, spontaneously developing ketosis is a frequent
problem.
7. Lipogenesis
A high-carbohydrate meal leads to an elevated
blood glucose concentration. The glycogen reserves
within cells are filled. The ATP level rises, blocking
the citric acid cycle, and citrate is exported from mito-
chondria (Fig. 17-20). Outside the mitochondria citrate
is cleaved by the ATP-requiring citrate lyase (Eq. 14-37)
to acetyl-CoA and oxaloacetate. The oxaloacetate can
be reduced to malate and the latter oxidized with
NADP
+
to pyruvate (Eq. 17-46), which can again enter
the mitochondrion. In this manner acetyl groups are
exported from the mitochondrion as acetyl-CoA which
can be carboxylated, under the activating influence of
citrate, to form malonyl-CoA, the precursor of fatty
acids. The NADPH formed from oxidation of the
malate provides part of the reducing equivalents
needed for fatty acid synthesis. Additional NADPH
is available from the pentose phosphate pathway.
Thus, excess carbohydrate is readily converted into
fat by our bodies. These reactions doubtless occur to
some extent in most cells, but they are quantitatively
Ch 17IND page - E - 2/14/03, 11:19 AM 1003
1004 Chapter 17. The Organization of Metabolism
feedback control. The result is that gluconeogenesis
is increased with corresponding breakdown of pro-
teins and amino acids. The liver glycogen is deplet-
ed and excess nitrogen from protein degradation
appears in the urine. In IDDM diabetes the products
of fatty acid degradation accumulate, leading to
ketosis. The volume of urine is excessive and tissues
are dehydrated.
Although the acute problems of diabetes, such
as coma induced by ketoacidosis, can usually be
avoided, it has not been possible to prevent long-
term complications that include cataract formation
and damage to the retina and kidneys. Most diabetics
eventually become blind and half die within 15 20
years. Many individuals with NIDDM develop
insulin-dependent diabetes in later life as a result of
damage to the pancreatic cells. The high glucose
level in blood appears to be a major cause of these
problems. The aldehyde form of glucose reacts
with amino groups of proteins to form Schiff bases
which undergo the Amadori rearrangement to form
ketoamines (Eq. 4-8). The resulting modified proteins
tend to form abnormal disulfide crosslinkages.
Crosslinked aggregates of lens proteins may be a
cause of cataract. The accumulating glucose-modified
proteins may also induce autoimmune responses
that lead to the long-term damage to kidneys and
other organs. Another problem results from reduction
of glucose to sorbitol (Box 20-A). Accumulation
of sorbitol in the lens may cause osmotic swelling,
another factor in the development of cataracts.
w,x
Excessive secretion of the 37-residue polypeptide
amylin, which is synthesized in the cells along
with insulin, is another frequent complication of
diabetes.
u,v
Amylin precipitates readily within islet
cells to form amyloid deposits which are character-
istic of NIDDM.
For many persons with diabetes regular injec-
tions of insulin are essential. Insulin was discovered
in 1921 in Toronto by Banting and Best, with a con-
troversial role being played by Professor J. J. R.
Macleod, who shared the Nobel prize with Banting
in 1923.
y,z
In 1922 the first young patients received
pancreatic extracts and a new, prolonged life.
z bb
Persons with IDDM are still dependent upon daily
injections of insulin, but attempts are being made
to treat the condition with transplanted cells from
human cadavers.
cc
Animal insulins are suitable for
most patients, but allergic reactions sometimes make
human insulin essential. The human hormone,
which differs from bovine insulin in three positions
(Thr in human vs Ala in bovine at positions 8 of the A
chain and 30 of the B chain and Ile vs Val at position
10 of the A chain), is now produced in bacteria using
recombinant DNA. Nonenzymatic laboratory syn-
thesis of insulin has also been achieved, but it is
difficult to place the disulfide crosslinks in the
proper positions. New approaches mimic the natu-
ral synthesis, in which the crosslinking takes place
in proinsulin (Fig. 10-7).
NIDDM is strongly associated with obesity,
dd
and dieting and exercise often provide adequate
control of blood glucose. Sulfonylurea drugs such
as the following induce an increase in the number of
insulin receptors formed and are also widely used in
treatment of the condition.
ee,ff
These drugs bind to
and inhibit ATP-sensitive K
+
channels in the cell
membranes. A defect in this sulfonylurea receptor
has been associated with excessive insulin secretion
H
2
N C
N
NH
NH
2
H
Aminoguanidine
a
Atkinson, M. A., and Maclaren, N. K. (1990) Sci. Am. 263(Jul), 6271
b
Taylor, S. I. (1995) in The Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 843896, McGraw-Hill, New York
c
Draznin, B., and LeRoith, D., eds. (1994) Molecular Biology of
Diabetes, Parts I and II, Humana Press, Totowa, New Jersey
d
Baekkeskov, S., Aanstoot, H.-J., Christgau, S., Reetz, A.,
Solimena, M., Cascalho, M., Folli, F., Richter-Olesen, H., and
Camilli, P.-D. (1990) Nature (London) 347, 151156
e
Nathan, B., Bao, J., Hsu, C.-C., Aguilar, P., Wu, R., Yarom, M.,
Kuo, C.-Y., and Wu, J.-Y. (1994) Proc. Natl. Acad. Sci. U.S.A. 91,
242246
f
Lu, J., Li, Q., Xie, H., Chen, Z.-J., Borovitskaya, A. E., Maclaren,
N. K., Notkins, A. L., and Lan, M. S. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 23072311
g
Solimena, M., Dirkx, R., Jr., Hermel, J.-M., Pleasic-Williams, S.,
Shapiro, J. A., Caron, L., and Rabin, D. U. (1996) EMBO J. 15,
21022114
h
MacDonald, H. R., and Acha-Orbea, H. (1994) Nature (London)
371, 283284
i
Todd, J. A. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 85608565
in infants.
17
New types of drugs are being tested.
ggkk
These include inhibitors of aldose reductase,
ii
which
forms sorbitol; compounds such as aminoguanidine,
which inhibit formation of advanced products of
glycation and newly discovered fungal metabolites
that activate insulin receptors.
jj
BOX 17-G DIABETES MELLITUS (continued)
H
3
C S N
O
O
C
O
N (CH
2
)
3
CH
3
H H
1-Butyl-p-tolylsulfonylurea
Ch 17IND page - E - 2/14/03, 11:19 AM 1004
1005
j
Davies, J. L., Kawaguchi, Y., Bennett, S. T., Copeman, J. B.,
Cordell, H. J., Pritchard, L. E., Reed, P. W., Gough, S. C. L.,
Jenkins, S. C., Palmer, S. M., Balfour, K. M., Rowe, B. R., Farrall,
M., Barnett, A. H., Bain, S. C., and Todd, J. A. (1994) Nature
(London) 371, 130136
k
Todd, J. A., Aitman, T. J., Cornall, R. J., Ghosh, S., Hall, J. R. S.,
Hearne, C. M., Knight, A. M., Love, J. M., McAleer, M. A., Prins,
J.-B., Rodrigues, N., Lathrop, M., Pressey, A., DeLarato, N. H.,
Peterson, L. B., and Wicker, L. S. (1991) Nature (London) 351,
542547
l
Leiter, E. H. (1989) FASEB J. 3, 22312241
m
Kim, J. K., Gavrilova, O., Chen, Y., Reitman, M. L., and
Shulman, G. I. (2000) J. Biol. Chem. 275, 84568460
n
Zhao, L., Cissell, M. A., Henderson, E., Colbran, R., and Stein,
R. (2000) J. Biol. Chem. 275, 1053210537
o
Catasti, P., Chen, X., Moyzis, R. K., Bradbury, E. M., and Gupta,
G. (1996) J. Mol. Biol. 264, 534545
p
Wang, J., Shen, L., Najafi, H., Kolberg, J., Matschinsky, F. M.,
Urdea, M., and German, M. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 43604365
q
Schmid, E., Hotz-Wagenblatt, A., Hack, V., and Drge, W. (1999)
FASEB J. 13, 14911500
r
Thomas, P. M., Cote, G. J., Wohllk, N., Haddad, B., Mathew, P.
M., Rabl, W., Aguilar-Bryan, L., Gagel, R. F., and Bryan, J. (1995)
Science 268, 426429
s
Nakajima, K., Yamauchi, K., Shigematsu, S., Ikeo, S., Komatsu,
M., Aizawa, T., and Hashizume, K. (2000) J. Biol. Chem. 275,
2088020886
t
Aspinwall, C. A., Brooks, S. A., Kennedy, R. T., and Lakey, J. R.
T. (1997) J. Biol. Chem. 272, 3130831314
u
Leighton, B., and Cooper, G. J. S. (1990) Trends Biochem. Sci. 15,
295299
v
Lorenzi, A., Razzaboni, B., Weir, G. C., and Yankner, B. A. (1994)
Nature (London) 368, 756760
w
De Winter, H. L., and von Itzstein, M. (1995) Biochemistry 34,
82998308
x
Wilson, D. K., Bohren, K. M., Gabbay, K. H., and Quiocho, F. A.
(1992) Science 257, 8184
y
Stevenson, L. G. (1979) Trends Biochem. Sci. 4, N158 N160
z
Broad, W. J. (1982) Science 217, 11201122
aa
Orci, L., Vassalli, J.-D., and Perrelet, A. (1988) Sci. Am. 259(Sep),
8594
bb
Marliss, E. B. (1982) N. Engl. J. Med. 306, 362364
cc
Lacy, P. E. (1995) Sci. Am. 273(Jul), 5058
dd
Simoneau, J.-A., Colberg, S. R., Thaete, F. L., and Kelley, D. E.
(1995) FASEB J. 9, 273278
ee
Aguilar-Bryan, L., Nichols, C. G., Wechsler, S. W., Clement, J. P.,
IV, Boyd, A. E., III, Gonzlez, G., Herrera-Sosa, H., Nguy, K.,
Bryan, J., and Nelson, D. A. (1995) Science 268, 423426
ff
Eliasson, L., Renstrm, E., mml, C., Berggren, P.-O., Ber-
torello, A. M., Bokvist, K., Chibalin, A., Deeney, J. T., Flatt, P. R.,
Gbel, J., Bromada, J., Larsson, O., Lindstrm, P., Rhodes, C. J.,
and Rorsman, P. (1996) Science 271, 813815
gg
Keen, H. (1994) N. Engl. J. Med. 331, 12261227
hh
Clark, C. M., Jr., and Lee, D. A. (1995) N. Engl. J. Med. 332,
12101216
ii
Bohren, K. M., Grimshaw, C. E., Lai, C.-J., Harrison, D. H.,
Ringe, D., Petsko, G. A., and Gabbay, K. H. (1994) Biochemistry
33, 20212032
jj
Qureshi, S. A., Ding, V., Li, Z., Szalkowski, D., Biazzo-Ashnault,
D. E., Xie, D., Saperstein, R., Brady, E., Huskey, S., Shen, X., Liu,
K., Xu, L., Salituro, G. M., Heck, J. V., Moller, D. E., Jones, A. B.,
and Zhang, B. B. (2000) J. Biol. Chem. 275, 3659036595
kk
Moler, D. E. (2001) Nature (London) 414, 821827
BOX 17-G (continued)
L. Regulation of Biosysnthesis
most important in the liver, in fat cells of adipose
tissue, and in mammary glands. The process is also
facilitated by insulin, which promotes the activation
of pyruvate dehydrogenase (Eq. 17-9) and of fatty acid
synthase of adipocytes.
277a
Activity of fatty acid syn-
thase seems to be regulated by the rate of transcription
of its gene, which is controlled by a transcription
factor designated either as adipocyte determination
and differentiation factor-1 (ADD-1) or sterol
regulatory element-binding protein-1c (SREBP-1c).
This protein (ADD-1/ SREBP-1c) may be a general
mediator of insulin action.
277b
The nuclear DNA-
binding protein known as peroxisome proliferator-
activated receptor gamma (PPAR

) is also involved
in the control of insulin action, a conclusion based
directly on discovery of mutations in persons with
type II diabetes.
277c
A newly discovered hormone
resistin, secreted by adipocytes, may also play a
role.
277d
Another adipocyte hormone, leptin, impairs
insulin action.
277e
Recent evidence suggests that both
insulin and leptin may have direct effects on the
brain which also influence blood glucose levels.
277f
Malonyl-CoA, which may also play a role in insulin
secretion,
278,279
inhibits carnitine palmitoyltransferase I
(CPT I; Fig. 17-2), slowing fatty acid catabolism.
280
Ch 17IND page - E - 2/14/03, 11:19 AM 1005
1006 Chapter 17. The Organization of Metabolism
1. Knoll, L. J., Schall, O. F., Suzuki, I., Gokel, G.
W., and Gordon, J. I. (1995) J. Biol. Chem. 270,
2009020097
2. Thorpe, C., and Kim, J.-J. P. (1995) FASEB J. 9,
718725
3. Vock, P., Engst, S., Eder, M., and Ghisla, S.
(1998) Biochemistry 37, 18481860
4. Aoyama, T., Ueno, I., Kamijo, T., and
Hashimoto, T. (1994) J. Biol. Chem. 269, 19088
19094
5. Eaton, S., Bartlett, K., and Pourfarzan, M.
(1996) Biochem. J. 320, 345357
5a. Dwyer, T. M., Mortl, S., Kemter, K., Bacher, A.,
Fauq, A., and Frerman, F. E. (1999) Biochemis-
try 38, 97359745
5b. Barycki, J. J., OBrien, L. I., Strauss, A. W.,
and Banaszak, L. J. (2001) J. Biol. Chem. 276,
3671836726
6. Ikeda, Y., Okamure-Ikeda, K., and Tanaka, K.
(1985) J. Biol. Chem. 260, 13111325
7. Yang, S.-Y., Bittman, R., and Schultz, H. (1985)
J. Biol. Chem. 260, 28622888
8. Yang, S.-Y., Yang, X.-Y. H., Healy-Louie, G.,
Schulz, H., and Elzinga, M. (1990) J. Biol.
Chem. 265, 1042410429
9. Wyatt, J. M. (1984) Trends Biochem. Sci. 9, 2023
10. Nada, M. A., Rhead, W. J., Sprecher, H.,
Schulz, H., and Roe, C. R. (1995) J. Biol. Chem.
270, 530535
11. Osumi, T., and Hashimoto, T. (1984) Trends
Biochem. Sci. 9, 317 319
12. Kaikaus, R. M., Sui, Z., Lysenko, N., Wu, N.
Y., Ortiz de Montellano, P. R., Ockner, R. K.,
and Bass, N. M. (1993) J. Biol. Chem. 268,
2686626871
13. Chu, R., Varanasi, U., Chu, S., Lin, Y., Usuda,
N., Rao, M. S., and Reddy, J. K. (1995) J. Biol.
Chem. 270, 49084915
14. Masters, C., and Crane, D. (1995) The
Peroxisome: A Vital Organelle, Cambridge Univ.
Press, London
15. Luo, Y., Karpichev, I. V., Kohanski, R. A., and
Small, G. M. (1996) J. Biol. Chem. 271, 12068
12075
16. Elgersma, Y., van Roermund, C. W. T.,
Wanders, R. J. A., and Tabak, H. F. (1995)
EMBO J. 14, 34723479
17. Gerhardt, B. (1993) in Lipid Metabolism in
Plants (Moore, T. S., Jr., ed), pp. 528565, CRC
Press, Boca Raton, Florida
18. Moore, T. S., Jr., ed. (1993) Lipid Metabolism in
Plants, CRC Press, Boca Raton, Florida
18a. Geisbrecht, B. V., Zhang, D., Schulz, H., and
Gould, S. J. (1999) J. Biol. Chem. 274, 21797
21803
19. Novikov, D. K., Vanhove, G. F., Carchon, H.,
Asselberghs, S., Eyssen, H. J., Van Veldhoven,
P. P., and Mannaerts, G. P. (1994) J. Biol. Chem.
269, 2712527135
19a. Lopez-Huertas, E., Charlton, W. L., Johnson,
B., Graham, I. A., and Baker, A. (2000) EMBO
J. 19, 67706777
20. Orci, L., Vassalli, J.-D., and Perrelet, A. (1988)
Sci. Am. 259(Sep), 8594
21. Luthria, D. L., Baykousheva, S. P., and
Sprecher, H. (1995) J. Biol. Chem. 270, 13771
13776
22. Filppula, S. A., Sormunen, R. T., Hartig, A.,
Kunau, W.-H., and Hiltunen, J. K. (1995) J.
Biol. Chem. 270, 2745327457
23. Smeland, T. E., Cuebas, D., and Schulz, H.
(1991) J. Biol. Chem. 266, 2390423908
24. Preisig-Mller, R., Ghnemann-Schfer, K.,
and Kindl, H. (1994) J. Biol. Chem. 269, 20475
20481
25. van Roermund, C. W. T., Elgersma, Y., Singh,
N., Wanders, R. J. A., and Tabak, H. F. (1995)
EMBO J. 14, 34803486
References
26. Malila, L. H., Siivari, K. M., Mkel, M. J.,
Jalonen, J. E., Latip, P. M., Kunau, W.-H.,
and Hiltunen, J. K. (1993) J. Biol. Chem. 268,
2157821585
27. Chen, L.-S., Jin, S.-J., and Tserng, K.-Y. (1994)
Biochemistry 33, 1052710534
28. Chen, L.-S., Jin, S.-J., Dejak, I., and Tserng, K.-
Y. (1995) Biochemistry 34, 442450
29. Zhang, D., Liang, X., He, X.-Y., Alipui, O. D.,
Yang, S.-Y., and Schulz, H. (2001) J. Biol. Chem.
276, 1362213627
29a. Mursula, A. M., van Aalten, D. M. F.,
Hiltunen, J. K., and Wierenga, R. K. (2001) J.
Mol. Biol. 309, 845853
29b. Fillgrove, K. L., and Anderson, V. E. (2001)
Biochemistry 40, 1241212421
29c. Henke, B., Girzalsky, W., Berteaux-Lecellier,
V., and Erdmann, R. (1998) J. Biol. Chem. 273,
37023711
30. Johnson, M. J. (1967) Science 155, 15151519
31. Eggink, G., Engel, H., Meijer, W. G., Otten, J.,
Kingma, J., and Witholt, B. (1988) J. Biol.
Chem. 263, 1340013405
32. Stumpf, P. K., ed. (1987) The Biochemistry of
Plants; A Comprehensive Treatise, Vol. 9,
Academic Press, Orlando, Florida
32a. Hamberg, M., Sanz, A., and Castresana, C.
(1999) J. Biol. Chem. 274, 2450324513
33. Singh, H., Beckman, K., and Poulos, A. (1994)
J. Biol. Chem. 269, 95149520
34. Vanhove, G. F., Van Veldhoven, P. P., Fransen,
M., Denis, S., Eyssen, H. J., Wanders, R. J. A.,
and Mannaerts, G. P. (1993) J. Biol. Chem. 268,
1033510344
34a. Foulon, V., Antonenkov, V. D., Croes, K.,
Waelkens, E., Mannaerts, G. P., Van
Veldhoven, P. P., and Casteels, M. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1003910044
35. Lazarow, P. B., and Moser, H. W. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2287
2324, McGraw-Hill, New York
36. Kaya, K., Ramesha, C. S., and Thompson, G.
A., Jr. (1984) J. Biol. Chem. 259, 35483553
37. Hardwick, J. P., Song, B.-J., Huberman, E., and
Gonzalez, F. J. (1987) J. Biol. Chem. 262, 801810
38. Cerdan, S., Knnecke, B., Dlle, A., and Seelig,
J. (1988) J. Biol. Chem. 263, 11664 11674
39. Tserng, K.-Y., and Jin, S.-J. (1991) J. Biol. Chem.
266, 29242929
40. Jin, S.-J., Hoppel, C. L., and Tserng, K.-Y.
(1992) J. Biol. Chem. 267, 119125
41. Bieber, L. L., Emaus, R., Valkner, K., and
Farrell, S. (1982) Fed. Proc. 41, 28582862
42. Bieber, L. L. (1988) Ann. Rev. Biochem. 57,
261283
43. Rebouche, C. J. (1992) FASEB J. 6, 33793386
44. Ramsay, R. R. (1994) in Essays in Biochemistry,
Vol. 28 (Tipton, K. F., ed), pp. 4762, Portland
Press, London and Chapel Hill, North
Carolina
45. Farrell, S. O., Fiol, C. J., Reddy, J. K., and Bieber,
L. L. (1984) J. Biol. Chem. 259, 1308913095
46. Brady, P. S., Ramsay, R. R., and Brady, L. J.
(1993) FASEB J. 7, 10391044
46a. Schmalix, W., and Bandlow, W. (1993) J. Biol.
Chem. 268, 2742827439
47. Brown, N. F., Weis, B. C., Husti, J. E., Foster,
D. W., and McGarry, J. D. (1995) J. Biol. Chem.
270, 89528957
47a. Yamazaki, N., Shinohara, Y., Kajimoto, K.,
Shindo, M., and Terada, H. (2000) J. Biol.
Chem. 275, 3173931746
48. Kelly, D. P., and Strauss, A. W. (1994) N. Engl.
J. Med. 330, 913 919
49. Marinetti, G. V. (1990) Disorders of Lipid
Metabolism, Plenum, New York
49a. Tamai, I., Ohashi, R., Nezu, J.-i, Sai, Y.,
Kobayashi, D., Oku, A., Shimane, M., and
Tsuji, A. (2000) J. Biol. Chem. 275, 4006440072
50. Roe, C. R., and Coates, P. M. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 1501
1533, McGraw-Hill, New York
50a. Wang, Y., Ye, J., Ganapathy, V., and Longo, N.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 2356
2360
51. Ford, D. A., Han, X., Horner, C. C., and Gross,
R. W. (1996) Biochemistry 35, 79037909
52. Requero, M. A., Goi, F. M., and Alonso, A.
(1995) Biochemistry 34, 1040010405
53. Treem, W. R., Stanley, C. A., Finegold, D. N.,
Hale, D. E., and Coates, P. M. (1988) N. Engl. J.
Med. 319, 13311336
54. Rhead, W. J., Amendt, B. A., Fritchman, K. S.,
and Felts, S. J. (1983) Science 221, 7375
55. Mosser, J., Douar, A.-M., Sarde, C.-O.,
Kioschis, P., Feil, R., Moser, H., Poustka, A.-
M., Mandel, J.-L., and Aubourg, P. (1993)
Nature (London) 361, 726730
56. Aubourg, P., Adamsbaum, C., Lavallard-
Rousseau, M.-C., Rocchiccioli, F., Cartier, N.,
Jambaqu, I., Jakobezak, C., Lemaitre, A.,
Boureau, F., Wolf, C., and Bougnres, P.-F.
(1993) N. Engl. J. Med. 329, 745752
57. Moser, H. W., Smith, K. D., and Moser, A. B.
(1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 2 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds),
pp. 23252349, McGraw-Hill, New York
58. Yahraus, T., Braverman, N., Dodt, G., Kalish,
J. E., Morrell, J. C., Moser, H. W., Valle, D., and
Gould, S. J. (1996) EMBO J. 15, 29142923
59. Street, J. M., Evans, J. E., and Natowicz, M. R.
(1996) J. Biol. Chem. 271, 35073516
60. Wolins, N. E., and Donaldson, R. P. (1994) J.
Biol. Chem. 269, 11491153
60a. Fujiwara, C., Imamura, A., Hashiguchi, N.,
Shimozawa, N., Suzuki, Y., Kondo, N.,
Imanaka, T., Tsukamoto, T., and Osumi, T.
(2000) J. Biol. Chem. 275, 3727137277
60b. Quant, P. A. (1994) in Essays in Biochemistry,
Vol. 28 (Tipton, K. F., ed), Portland Press,
Chapel Hill, North Carolina
61. Krebs, H. A., Williamson, D. H., Bates, M. W.,
Page, M. A., and Hawkins, R. A. (1971) Adv.
Enzyme Regul. 9, 387409
62. McGarry, J. D., and Foster, D. W. (1980) Ann.
Rev. Biochem. 49, 395420
63. Endemann, G., Goetz, P. G., Edmond, J., and
Brunengraber, H. (1982) J. Biol. Chem. 257,
3434 3440
64. Ohgaku, S., Brady, P. S., Schumann, W. C.,
Bartsch, G. E., Margolis, J. M., Kumaran, K.,
Landau, S. B., and Landau, B. R. (1982) J. Biol.
Chem. 257, 92839289
64a. Misra, I., and Miziorko, H. M. (1996)
Biochemistry 35, 96109616
64b. Chun, K. Y., Vinarov, D. A., and Miziorko, H.
M. (2000) Biochemistry 39, 1467014681
64c. Chun, K. Y., Vinarov, D. A., Zajicek, J., and
Miziorko, H. M. (2000) J. Biol. Chem. 275,
1794617953
65. Sato, K., Kashiwaya, Y., Keon, C. A., Tsuchiya,
N., King, M. T., Radda, G. K., Chance, B.,
Clarke, K., and Veech, R. L. (1995) FASEB J. 9,
651658
66. Casazza, J. P., Felver, M. E., and Veech, R. L.
(1984) J. Biol. Chem. 259, 231236
67. Argils, J. M. (1986) Trends Biochem. Sci. 11,
6163
68. Landau, B. R., and Brunengraber, H. (1987)
Trends Biochem. Sci. 12, 113114
Ch 17IND page - E - 2/14/03, 11:19 AM 1006
1007
References
69. Gavino, V. C., Somma, J., Philbert, L., David,
F., Garneau, M., Blair, J., and Brunengraber,
H. (1987) J. Biol. Chem. 262, 6735 6740
70. Vagelos, P. R. (1960) J. Biol. Chem. 235, 346350
70a. Sacksteder, K. A., Morrell, J. C., Wanders, R. J.
A., Matalon, R., and Gould, S. J. (1999) J. Biol.
Chem. 274, 2446124468
71. Hofmeister, A. E. M., and Buckel, W. (1992)
Eur. J. Biochem. 206, 547552
72. Buckel, W. (1992) FEMS Microbiol. Rev. 88,
211232
73. Vagelos, P. R., Earl, J. M., and Stadtman, E. R.
(1959) J. Biol. Chem. 234, 490497, 765 769
74. Kuchta, R. D., and Abeles, R. H. (1985) J. Biol.
Chem. 260, 13181 13189
75. Taylor, S. I. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 843896, McGrawHill,
New York
76. Weigand, E., Young, J. W., and McGilliard, A.
D. (1972) Biochem. J. 126, 201209
77. Baldwin, J. E., and Krebs, H. (1981) Nature
(London) 291, 381 385
78. Nishimura, J. S., and Grinnell, F. (1972) Adv.
Enzymol. 36, 183 202
79. Rolleson, F. S. (1972) Curr. Top. Cell. Regul. 5,
4775
79a. Jucker, B. M., Lee, J. Y., and Shulman, R. G.
(1998) J. Biol. Chem. 273, 1218712194
79b. Petersen, S., de Graaf, A. A., Eggeling, L.,
Mllney, M., Wiechert, W., and Sahm, H.
(2000) J. Biol. Chem. 275, 3593235941
80. Kornberg, H. L. (1966) Essays Biochem. 2, 131
81. Newsholme, E. A., and Start, C. (1973)
Regulation in Metabolism, Wiley, New York
(pp. 124145)
82. Lieber, C. S. (1976) Sci. Am. 234(Mar), 2533
83. Kalnitsky, G., and Tapley, D. F. (1958) Biochem.
J. 70, 2834
84. Rahmatullah, M., and Roche, T. E. (1987) J.
Biol. Chem. 262, 1026510271
85. Yudkoff, M., Nelson, D., Daikhin, Y., and
Erecinska, M. (1994) J. Biol. Chem. 269, 27414
27420
85a. Reed, L. J. (2001) J. Biol. Chem. 276, 38329
38336
86. Stueland, C. S., Eck, K. R., Stieglbauer, K. T.,
and LaPorte, D. C. (1987) J. Biol. Chem. 262,
1609516099
86a. Panisko, E. A., and McAlister-Henn, L. (2001)
J. Biol. Chem. 276, 12041210
87. Rashed, H. M., Waller, F. M., and Patel, T. B.
(1988) J. Biol. Chem. 263, 57005706
88. Chiang, P. K., and Sacktor, B. (1975) J. Biol.
Chem. 250, 3399 3408
89. Krebs, H. A. (1970) Adv. Enzyme Regul. 8,
335 353
90. Maeng, C.-Y., Yazdi, M. A., and Reed, L. J.
(1996) Biochemistry 35, 58795882
91. Harris, R. A., Bowker-Kinley, M. M., Wu, P.,
Jeng, J., and Popov, K. M. (1997) J. Biol. Chem.
272, 1974619751
92. Patel, M. S., and Roche, T. E. (1990) FASEB J.
4, 32243233
92a. Yang, D., Gong, X., Yakhnin, A., and Roche, T.
E. (1998) J. Biol. Chem. 273, 1413014137
92b. McCartney, R. G., Sanderson, S. J., and
Lindsay, J. G. (1997) Biochemistry 36, 6819
6826
93. Bessman, S. P., Mohan, C., and Zaidise, I.
(1986) Proc. Natl. Acad. Sci. U.S.A. 83, 5067
5070
94. Wan, B., LaNoue, K. F., Cheung, J. Y., and
Scaduto, R. C., Jr. (1989) J. Biol. Chem. 264,
1343013439
95. Srere, P. A. (1971) Adv. Enzyme Regul. 9, 221
233
96. Ovdi, J., and Srere, P. A. (1996) Cell Biochem.
Funct. 14, 249258
97. Sherry, A. D., and Malloy, C. R. (1996) Cell
Biochem. Funct. 14, 259268
97a. Vlot, C., Mixon, M. B., Teige, M., and Srere,
P. A. (1997) Biochemistry 36, 1427114276
98. Palmer, T. N., and Sugden, M. C. (1983) Trends
Biochem. Sci. 8, 161162
99. Loeber, G., Infante, A. A., Maurer-Fogy, I.,
Krystek, E., and Dworkin, M. B. (1991) J. Biol.
Chem. 266, 30163021
100. Mandella, R. D., and Sauer, L. A. (1975) J. Biol.
Chem. 250, 5877 5884
101. Macrae, A. R. (1971) Biochem. J. 122, 495501
102. Huh, T.-L., Casazza, J. P., Huh, J.-W., Chi, Y.-
T., and Song, B. J. (1990) J. Biol. Chem. 265,
1332013326
103. Nathan, B., Bao, J., Hsu, C.-C., Aguilar, P., Wu,
R., Yarom, M., Kuo, C.-Y., and Wu, J.-Y. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 242246
104. Nathan, B., Hsu, C.-C., Bao, J., Wu, R., and
Wu, J.-Y. (1994) J. Biol. Chem. 269, 72497254
105. Toney, M. D., Pascarella, S., and De Biase, D.
(1995) Protein Sci. 4, 23662374
106. Hearl, W. G., and Churchich, J. E. (1985) J.
Biol. Chem. 260, 1636116366
107. Baum, G., Lev-Yadum, S., Fridmann, Y.,
Arazi, T., Katsnelson, H., Zik, M., and Fromm,
H. (1996) EMBO J. 15, 29882996
108. Baum, G., Chen, Y., Arazi, T., Takatsuji, H.,
and Fromm, H. (1993) J. Biol. Chem. 268,
1961019617
109. Satyanarayan, V., and Nair, P. M. (1990)
Phytochemistry 29, 367 375
110. Streeter, J. G., and Thompson, J. F. (1972) Plant
Physiol. 49, 579 584
111. del Rio, R. M. (1981) J. Biol. Chem. 256, 9816
9819
112. Bartley, W., Kornberg, H. L., and Quayle, J. R.
(1970) Essays in Cell Metabolism, Wiley-
Interscience, New York (p. 125)
113. Quayle, J. R. (1963) Biochem. J. 89, 492503
114. Chang, Y.-Y., Wang, A.-Y., and Cronan, J. E., Jr.
(1993) J. Biol. Chem. 268, 39113919
115. Fothergill-Gilmore, L. A. (1986) Trends
Biochem. Sci. 11, 4751
115a. Bond, C. J., Jurica, M. S., Mesecar, A., and
Stoddard, B. L. (2000) Biochemistry 39, 15333
15343
115b. Valentini, G., Chiarelli, L., Fortin, R.,
Speranza, M. L., Galizzi, A., and Mattevi, A.
(2000) J. Biol. Chem. 275, 1814518152
115c. Rigden, D. J., Phillips, S. E. V., Michels, P. A.
M., and Fothergill-Gilmore, L. A. (1999) J.
Mol. Biol. 291, 615635
116. Horecker, B. L., Gibbs, M., Klenow, H., and
Smyrniotis, P. Z. (1954) J. Biol. Chem. 207,
393403
117. Wood, T. (1985) The Pentose Phosphate Pathway,
Academic Press, Orlando, Florida
117a. Vought, V., Ciccone, T., Davino, M. H.,
Fairbairn, L., Lin, Y., Cosgrove, M. S., Adams,
M. J., and Levy, H. R. (2000) Biochemistry 39,
1501215021
117b. Karsten, W. E., Chooback, L., and Cook, P. F.
(1998) Biochemistry 37, 1569115697
117c. Kopriva, S., Koprivova, A., and Sss, K.-H.
(2000) J. Biol. Chem. 275, 12941299
118. Bublitz, C., and Steavenson, S. (1988) J. Biol.
Chem. 263, 12849 12853
118a. Johnson, A. E., and Tanner, M. E. (1998)
Biochemistry 37, 57465754
118b. Chen, Y.-R., Larimer, F. W., Serpersu, E. H.,
and Hartman, F. C. (1999) J. Biol. Chem. 274,
21322136
119. Zubay, G. L., Parsons, W. W., and Vance, D. E.
(1995) Principles of Biochemistry, W.C.Brown,
Dubuque, Iowa (p. 276)
120. Reitzer, L. J., Wice, B. M., and Kennell, D.
(1980) J. Biol. Chem. 255, 5616 5626
121. Scofield, R. E., Kosugi, K., Chandramouli, V.,
Kumaran, K., Schumann, W. C., and Landau,
B. R. (1985) J. Biol. Chem. 260, 1543915444
122. Magnusson, I., Chandramouli, V., Schumann,
W. C., Kumaran, K., Wahren, J., and Landau,
B. R. (1988) Proc. Natl. Acad. Sci. U.S.A. 85,
46824685
122a. Kurland, I. J., Alcivar, A., Bassilian, S., and
Lee, W.-N. P. (2000) J. Biol. Chem. 275, 36787
36793
123. Longenecker, J. P., and Williams, J. F. (1980)
Biochem. J. 188, 847857
124. Williams, J. F. (1980) Trends Biochem. Sci. 5,
315320
125. Williams, J. F. (1983) Trends Biochem. Sci. 8,
275277
126. Flanigan, I., Collins, J. G., Arora, K. K.,
Macleod, J. K., and Williams, J. F. (1993) Eur. J.
Biochem. 213, 477485
127. Landau, B. R., and Wood, H. G. (1983) Trends
Biochem. Sci. 8, 292296; 312313
128. Casazza, J. P., and Veech, R. L. (1986) J. Biol.
Chem. 261, 690698
129. Bernacchia, G., Schwall, G., Lottspeich, F.,
Salamini, F., and Bartels, D. (1995) EMBO J.
14, 610618
130. Berthon, H. A., Kuchel, P. W., and Nixon, P. F.
(1992) Biochemistry 31, 1279212798
131. Horecker, B. L. (1965) J. Chem. Educ. 42, 244
253
132. Decker, K., Jungermann, K., and Thauer, R. K.
(1970) Angew. Chem. Int. Ed. Engl. 9, 138158
133. Mushegian, A. R., and Koonin, E. V. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1026810273
134. Bult, C. J., and 39 other authors. (1996) Science
273, 10581073
135. DiGirolamo, M., Newby, F. D., and Lovejoy, J.
(1992) FASEB J. 6, 24052412
136. Robinson, B. H. (1982) Trends Biochem. Sci. 7,
151153
136a. DiGirolamo, M., Newby, F. D., and Lovejoy, J.
(1992) FASEB J. 6, 24052412
136b. Shulman, R. G., and Rothman, D. L. (2001)
Proc. Natl. Acad. Sci. U.S.A. 98, 457461
136c. Gladden, L. B. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 395397
136d. Bouzier, A.-K., Goodwin, R., Macouillard-
Poulletier de Gannes, F., Valeins, H., Voisin,
P., Canioni, P., and Merle, M. (1998) J. Biol.
Chem. 273, 2716227169
137. Roberts, J. K. M., Callis, J., Jardetzky, O.,
Walbot, V., and Freeling, M. (1984) Proc. Natl.
Acad. Sci. U.S.A. 81, 60296033
138. Hake, S., Kelley, P. M., Taylor, W. C., and
Freeling, M. (1985) J. Biol. Chem. 260, 5050
5054
139. Shoubridge, E. A., and Hochachka, P. W.
(1980) Science 209, 308309
139a. Teusink, B., Walsh, M. C., van Dam, K., and
Westerhoff, H. V. (1998) Trends Biochem. Sci.
23, 162169
139b. Bakker, B. M., Mensonides, F. I. C., Teusink,
B., van Hoek, P., Michels, P. A. M., and
Westerhoff, H. V. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 20872092
139c. Manchester, K. L. (2000) Trends Biochem. Sci.
25, 8992
140. Tempest, D. W., and Neijssel, O. M. (1987) in
Escherichia coli and Salmonella typhimurium
(Neidhardt, F. C., ed), pp. 797806, Am. Soc.
for Microbiology, Washington, D.C.
141. Blackwood, A. C., Neish, A. C., and Ledingham,
G. A. (1956) J. Bacteriol. 72, 497499
142. Krebs, H. A. (1972) Essays Biochem. 8, 134
143. Tse, J. M.-T., and Schloss, J. V. (1993)
Biochemistry 32, 10398 10403
Ch 17IND page - E - 2/14/03, 11:19 AM 1007
1008 Chapter 17. The Organization of Metabolism
References
144. Bui, E. T. N., Bradley, P. J., and Johnson, P. J.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 9651
9656
145. Hochachka, P. W., and Mustafa, T. (1972)
Science 178, 10561060
146. Hochachka, P. W., and Somero, G. N. (1973)
Strategies of Biochemical Adaptation, Saunders,
Philadelphia, Pennsylvania (pp. 4661)
147. Hochachka, P. W. (1980) Living Without
Oxygen, Harvard Univ. Press, Cambridge,
Massachusetts
148. Zhang, M., Eddy, C., Deanda, K., Finkelstein,
M., and Picataggio, S. (1995) Science 267, 240
243
149. Stanier, R. Y., Doudoroff, M., and Adelberg, E.
A. (1970) Microbial World, 3rd ed., Prentice-
Hall, Englewood Cliffs, New Jersey (p. 191)
150. Switzer, R. L. (1974) in The Enzymes, 3rd ed.,
Vol. 10 (Boyer, P. D., ed), pp. 607629,
Academic Press, New York
150a. Baykov, A. A., Fabrichniy, I. P., Pohjanjoki, P.,
Zyryanov, A. B., and Lahti, R. (2000)
Biochemistry 39, 1193911947
150b. Ahn, S., Milner, A. J., Ftterer, K., Konopka,
M., Ilias, M., Young, T. W., and White, S. A.
(2001) J. Mol. Biol. 313, 797811
151. Wood, H. G., and Goss, N. H. (1985) Proc.
Natl. Acad. Sci. U.S.A. 82, 312315
152. Wood, H. G. (1977) Fed. Proc. 36, 21972205
153. Liu, C.-L., Hart, N., and Peck, H. D., Jr. (1982)
Science 217, 363 364
154. Weinbach, E. C. (1981) Trends Biochem. Sci. 6,
254257
155. Takeshige, K., and Tazawa, M. (1989) J. Biol.
Chem. 264, 3262 3266
156. Kornberg, A., Rao, N. N., and Ault-Rich, D.
(1999) Ann. Rev. Biochem. 68, 89125
156a. Rashid, M. H., Rumbaugh, K., Passador, L.,
Davies, D. G., Hamood, A. N., Iglewski, B. H.,
and Kornberg, A. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 96369641
156b. Bolesch, D. G., and Keasling, J. D. (2000) J.
Biol. Chem. 275, 3381433819
157. Mudd, S. H. (1973) The Enzymes, 3rd ed., Vol.
8, Academic Press, New York (pp. 121154)
158. Cornforth, J. W., Reichard, S. A., Talalay, P.,
Carrell, H. L., and Glusker, J. P. (1977) J. Am.
Chem. Soc. 99, 72927301
159. Peck, H. D., Jr. (1974) in The Enzymes, 3rd ed.,
Vol. 10 (Boyer, P. D., ed), pp. 651669,
Academic Press, New York
160. Geller, D. H., Henry, J. G., Belch, J., and
Schwartz, N. B. (1987)
J. Biol. Chem. 262, 73747382
161. Liu, C., Martin, E., and Leyh, T. S. (1994)
Biochemistry 33, 2042 2047
162. Li, H., Deyrup, A., Mensch, J. R., Jr.,
Domowicz, M., Konstantinidis, A. K., and
Schwartz, N. B. (1995) J. Biol. Chem. 270,
2945329459
163. Morra, S., Chiadmi, M., LeBras, G., Lascu, I.,
and Janin, J. (1995) Biochemistry 34, 11062
11070
163a. MacRae, I. J., Segel, I. H., and Fisher, A. J.
(2000) Biochemistry 39, 16131621
164. Wakil, S. J., Stoops, J. K., and Joshi, V. C.
(1983) Ann. Rev. Biochem. 52, 537579
165. White, H. B., III, Mitsuhashi, O., and Bloch, K.
(1971) J. Biol. Chem. 246, 47514754
166. Williamson, D. H., Lund, P., and Krebs, H. A.
(1967) Biophys. J. 103, 514527
167. Stubbs, M., Veech, R. L., and Krebs, H. A.
(1972) Biophys. J. 126, 5965
168. Chung, Y., and Jue, T. (1992) Biochemistry 31,
1115911165
169. Krebs, H. A., and Veech, R. L. (1969)
Mitochondria: Structure & Function, Academic
Press, New York (pp. 101109)
170. Veech, R. L., Guynn, R., and Veloso, D. (1972)
Biochem. J. 127, 387397
171. Lundquist, R., and Olivera, B. M. (1971) J.
Biol. Chem. 246, 11071116
172. Buchanan, B. B. (1969) J. Biol. Chem. 244,
42184223
173. Gehring, U., and Arnon, D. I. (1972) J. Biol.
Chem. 247, 6963 6969
173a. Furdui, C., and Ragsdale, S. W. (2002)
Biochemistry 41, 99219937
174. Stanier, R. Y., Doudoroff, M., and Adelberg, E.
A. (1970) The Microbial World, 3rd ed.,
Prentice-Hall, Englewood Cliffs, New Jersey
175. Stanier, R. Y., Ingraham, J. L., Wheelis, M. L.,
and Painter, P. R. (1986) The Microbial World,
5th ed., Prentice-Hall, Englewood Cliffs, New
Jersey
176. Brock, T. D., and Madigan, M. T. (1988)
Microbiology, Prentice-Hall, Englewood Cliffs,
New Jersey
177. Gottschalk, G. (1979) Bacterial Metabolism,
Springer-Verlag, New York
178. Buchanan, B. B., Schrmann, P., and
Shanmugam, K. T. (1972) Biochim. Biophys.
Acta. 283, 136145
179. Wood, H. G., Ragsdale, S. W., and Pezacka, E.
(1986) Trends Biochem. Sci. 11, 1417
180. Ferry, J. G., ed. (1993) Methanogenesis: Ecology,
Physiology, Biochemistry and Genetics,
Chapman & Hall, New York
181. Thauer, R. K., Hedderich, R., and Fischer, R.
(1993) in Methanogenesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
209252, Chapman & Hall, New York
182. Thauer, R. K., Schwrer, B., and Zirngibl, C.
(1993) in Microbial Growth on C1 Compounds
(Murrell, J. C., and Kelly, D. P., eds), pp. 151
162, Intercept Ltd., Andover, UK
183. Mller, V., Blaut, M., and Gottschalk, G.
(1993) in Methanogenesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
360 406, Chapman & Hall, New York
184. Simpson, P. G., and Whitman, W. B. (1993) in
Methanogenesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
445472, Chapman & Hall, New York
185. Ljungdahl, L., Irion, E., and Wood, H. G.
(1965) Biochemistry 4, 27712780
186. Wood, H. G. (1991) FASEB J. 5, 156163
187. Drake, H. L. (1993) in Microbial Growth on C1
Compounds (Murrell, J. C., and Kelly, D. P.,
eds), pp. 493507, Intercept Ltd., Andover,
UK
188. Wood, H. G., and Ljungdahl, L. G. (1991) in
Variations in Autotrophic Life (Shively, J. M.,
and Barton, L. L., eds), pp. 201 250,
Academic Press, San Diego, California
188a. Tan, X. S., Sewell, C., and Lindahl, P. A. (2002)
J. Am. Chem. Soc. 124, 62776284
189. Qiu, D., Kumar, M., Ragsdale, S. W., and Spiro,
T. G. (1996) J. Am. Chem. Soc. 118, 1042910435
189a. Doukov, T. I., Iverson, T. M., Seravalli, J.,
Ragsdale, S. W., and Drennan, C. L. (2002)
Science 298, 567572
189b. Peters, J. W. (2002) Science 298, 552553
190. Menon, S., and Ragsdale, S. W. (1996)
Biochemistry 35, 12119 12125
191. Hemming, A., and Blotevogel, K. H. (1985)
Trends Biochem. Sci. 10, 198200
192. Salem, A. R., Hacking, A. J., and Quayle, J. R.
(1973) Biochem. J. 136, 8996
193. Quayle, J. R. (1980) Biochem. Soc. Trans. 8, 110
194. Anthony, C. (1982) The Biochemistry of
Methylotrophs, Academic Press, New York
195. Colby, J., Dalton, H., and Whittenbury, R.
(1979) Ann. Rev. Microbiol. 33, 481517
196. Haber, C. L., Allen, L. N., Zhao, S., and
Hanson, R. S. (1983) Science 221, 1147 1153
197. Higgins, I. J., Best, D. J., and Hammond, R. C.
(1980) Nature (London) 286, 561567
198. Anthony, C. (1996) Biochem. J. 320, 697711
199. Stueland, C. S., Gorden, K., and LaPorte, D. C.
(1988) J. Biol. Chem. 263, 1947519479
200. Dickinson, J. R., Dawes, I. W., Boyd, A. S. F.,
and Baxter, R. L. (1983) Proc. Natl. Acad. Sci.
U.S.A. 80, 58475851
201. Liu, F., Thatcher, J. D., and Epstein, H. F.
(1997) Biochemistry 36, 255260
202. Cook, A. G., and Knowles, J. R. (1985)
Biochemistry 24, 5158
203. Pocalyko, D. J., Carroll, L. J., Martin, B. M.,
Babbitt, P. C., and Dunaway-Mariano, D.
(1990) Biochemistry 29, 1075710765
204. Xu, Y., Yankie, L., Shen, L., Jung, Y.-S.,
Mariano, P. S., and Dunaway-Mariano, D.
(1995) Biochemistry 34, 21812187
205. Ohlrogge, J. B. (1982) Trends Biochem. Sci. 7,
386387
205a. Heath, R. J., Su, N., Murphy, C. K., Rock, C. O.
(2000) J. Biol. Chem. 275, 4012840133
206. Holak, T. A., Kearsley, S. K., Kim, Y., and
Prestegard, J. H. (1988) Biochemistry 27, 6135
6142
207. Wood, W. I., Peterson, D. O., and Bloch, K.
(1978) J. Biol. Chem. 253, 26502656
208. Kolodziej, S. J., Penczek, P. A., Schroeter, J. P.,
and Stoops, J. K. (1996) J. Biol. Chem. 271,
2842228429
209. Smith, S. (1994) FASEB J. 8, 12481259
210. White, R. H. (1989) Arch. Biochem. Biophys.
270, 691697
211. Lipmann, F. (1971) Science 173, 875884
212. Kurahashi, K. (1974) Ann. Rev. Biochem. 43,
445459
213. Kleinkauf, H., and von Dhren, H. (1983)
Trends Biochem. Sci. 8, 281283
214. Stein, T., Vater, J., Kruft, V., Otto, A.,
Wittmann-Liebold, B., Franke, P., Panico, M.,
McDowell, R., and Morris, H. R. (1996)
J. Biol. Chem. 271, 1542815435
215. Pfeifer, E., Pavela-Vrancic, M., von Dhren,
H., and Kleinkauf, H. (1995) Biochemistry 34,
74507459
216. Pavela-Vrancic, M., Pfeifer, E., Schrder, W.,
von Dhren, H., and Kleinkauf, H. (1994) J.
Biol. Chem. 269, 1496214966
217. Haese, A., Pieper, R., von Ostrowski, T., and
Zocher, R. (1994)
J. Mol. Biol. 243, 116122
218. Kao, C. M., Pieper, R., Cane, D. E., and Khosla,
C. (1996) Biochemistry 35, 1236312368
219. Cortes, J., Wiesmann, K. E. H., Roberts, G. A.,
Brown, M. J. B., Staunton, J., and Leadlay, P. F.
(1995) Science 268, 14871489
220. Pooley, H. M., and Karamata, D. (1994) in
Bacterial Cell Wall (New Comprehensive
Biochemistry), Vol. 27 (Ghuysen, J.-M., and
Hakenbeck, R., eds), pp. 187198, Elsevier,
Amsterdam
221. Knull, H., and Minton, A. P. (1996) Cell
Biochem. Funct. 14, 237 248
222. Low, P. S., Rathinavelu, P., and Harrison, M.
L. (1993) J. Biol. Chem. 268, 1462714631
223. Hardin, C. D., and Roberts, T. M. (1995)
Biochemistry 34, 1323 1331
224. Milner, Y., and Wood, H. G. (1972) Proc. Natl.
Acad. Sci. U.S.A. 69, 24632468
225. Newsholme, E. A., and Start, C. (1973)
Regulation in Metabolism, Wiley, New York
226. Gruetter, R., Novotny, E. J., Boulware, S. D.,
Rothman, D. L., Mason, G. F., Shulman, G. I.,
Shulman, R. G., and Tamborlane, W. V. (1992)
Proc. Natl. Acad. Sci. U.S.A. 89, 11091112
227. Lapidot, A., and Gopher, A. (1994) J. Biol.
Chem. 269, 27198 27208
228. Efrat, S., Tal, M., and Lodish, H. F. (1994)
Trends Biochem. Sci. 19, 535538
Ch 17IND page - E - 2/14/03, 11:19 AM 1008
1009
References
228a. Saltiel, A. R., and Kahn, C. R. (2001) Nature
(London) 414, 799806
229. Paz, K., Voliovitch, H., Hadari, Y. R., Roberts,
C. T., Jr., LeRoith, D., and Zick, Y. (1996) J.
Biol. Chem. 271, 69987003
230. Moxham, C. M., Tabrizchi, A., Davis, R. J.,
and Malbon, C. C. (1996) J. Biol. Chem. 271,
3076530773
231. Guilherme, A., Klarlund, J. K., Krystal, G.,
and Czech, M. P. (1996) J. Biol. Chem. 271,
2953329536
232. Tsakiridis, T., Taha, C., Grinstein, S., and Klip,
A. (1996) J. Biol. Chem. 271, 1966419667
232a. Krook, A., Whitehead, J. P., Dobson, S. P.,
Griffiths, M. R., Ouwens, M., Baker, C.,
Hayward, A. C., Sen, S. K., Maassen, J. A.,
Siddle, K., Tavar, J. M., and ORahilly, S.
(1997) J. Biol. Chem. 272, 3020830214
232b. Ottensmeyer, F. P., Beniac, D. R., Luo, R. Z.-T.,
and Yip, C. C. (2000) Biochemistry 39, 12103
12112
232c. Aguirre, V., Uchida, T., Yenush, L., Davis, R.,
and White, M. F. (2000) J. Biol. Chem. 275,
90479054
232d. Alper, J. (2000) Science 289, 37,39
232e. Withers, D. J., Gutierrez, J. S., Towery, H.,
Burks, D. J., Ren, J.-M., Previs, S., Zhang, Y.,
Bernal, D., Pons, S., Shulman, G. I., Bonner-
Weir, S., and White, M. F. (1998) Nature
(London) 391, 900904
232f. Czech, M. P., and Corvera, S. (1999) J. Biol.
Chem. 274, 18651868
232g. Lavan, B. E., Fantin, V. R., Chang, E. T., Lane,
W. S., Keller, S. R., and Lienhard, G. E. (1997)
J. Biol. Chem. 272, 2140321407
232h. Inoue, G., Cheatham, B., Emkey, R., and
Kahn, C. R. (1998) J. Biol. Chem. 273, 11548
11555
232i. Qiao, L.-y, Goldberg, J. L., Russell, J. C., and
Sun, X. J. (1999) J. Biol. Chem. 274, 1062510632
232j. Pessin, J. E., Thurmond, D. C., Elmendorf, J.
S., Coker, K. J., and Okada, S. (1999) J. Biol.
Chem. 274, 25932596
232k. Shepherd, P. R., and Kahn, B. B. (1999) N.
Engl. J. Med. 341, 248257
232l. Rea, S., Martin, L. B., McIntosh, S., Macaulay,
S. L., Ramsdale, T., Baldini, G., and James, D.
E. (1998) J. Biol. Chem. 273, 1878418792
232m.Lee, W., Ryu, J., Souto, R. P., Pilch, P. F., and
Jung, C. Y. (1999) J. Biol. Chem. 274, 37755
37762
232n. Kanzaki, M., Watson, R. T., Artemyev, N. O.,
and Pessin, J. E. (2000) J. Biol. Chem. 275,
71677175
232o. Czech, M. P. (2000) Nature (London) 407, 147
148
232p. Baumann, C. A., Ribon, V., Kanzaki, M.,
Thurmond, D. C., Mora, S., Shigematsu, S.,
Bickel, P. E., Pessin, J. E., and Saltiel, A. R.
(2000) Nature (London) 407, 202207
233. Ostlund, R. E., Jr., Seemayer, R., Gupta, S.,
Kimmel, R., Ostlund, E. L., and Sherman, W.
R. (1996) J. Biol. Chem. 271, 1007310078
234. Ostlund, R. E., Jr., McGill, J. B., Herskowitz, I.,
Kipnis, D. M., Santiago, J. V., and Sherman,
W. R. (1993) Proc. Natl. Acad. Sci. U.S.A. 90,
99889992
235. Galasko, G. T. F., Abe, S., Lilley, K., Zhang, C.,
and Larner, J. (1996) J. Clin. Endocrinol. Metab.
81, 10511057
236. Romero, G., Luttrell, L., Rogol, A., Zeller, K.,
Hewlett, E., and Larner, J. (1988) Science 240,
509511
236a. Frick, W., Bauer, A., Bauer, J., Wied, S., and
Mller, G. (1998) Biochemistry 37, 1342113436
237. Saltiel, A. R. (1994) FASEB J. 8, 10341040
237a. Mller, G., Grey, S., Jung, C., and Bandlow, W.
(2000) Biochemistry 39, 14751488
237b. Thorens, B., Guillam, M.-T., Beermann, F.,
Burcelin, R., and Jaquet, M. (2000) J. Biol.
Chem. 275, 2375123758
238. Van Schaftingen, E., Detheux, M., and Veiga
da Cunha, M. (1994) FASEB J. 8, 414419
239. Terauchi, Y., Sakura, H., Yasuda, K., Iwamoto,
K., Takahashi, N., Ito, K., Kasai, H., Suzuki,
H., Ueda, O., Kamada, N., Jishage, K.,
Komeda, K., Noda, M., Kanazawa, Y.,
Taniguchi, S., Miwa, I., Akanuma, Y.,
Kodama, T., Yazaki, Y., and Kadowaki, T.
(1995) J. Biol. Chem. 270, 3025330256
240. Agius, L., Peak, M., Newgard, C. B., Gomez-
Foix, A. M., and Guinovart, J. J. (1996) J. Biol.
Chem. 271, 3047930486
241. Webb, D.-L., Islam, M. S., Efanov, A. M.,
Brown, G., Khler, M., Larsson, O., and
Berggren, P.-O. (1996) J. Biol. Chem. 271,
1907419079
242. Deeney, J. T., Cunningham, B. A., Chheda, S.,
Bokvist, K., Juntti-Berggren, L., Lam, K.,
Korchak, H. M., Corkey, B. E., and Berggren,
P.-O. (1996) J. Biol. Chem. 271, 1815418160
243. Bergsten, P., Grapengiesser, E., Gylfe, E.,
Tangholm, A., and Hellman, B. (1994) J. Biol.
Chem. 269, 87498753
244. Buggy, J. J., Livingston, J. N., Rabin, D. U.,
and Yoo-Warren, H. (1995) J. Biol. Chem. 270,
74747478
245. Walajtys-Rhode, E., Zapatero, J., Moehren, G.,
and Hoek, J. B. (1992) J. Biol. Chem. 267, 370
379
246. de Duve, C. (1994) FASEB J. 8, 979 981
247. Rubanyi, G. M., and Botelho, L. H. P. (1991)
FASEB J. 5, 2713 2720
248. Heimberg, H., De Vos, A., Moens, K.,
Quartier, E., Bouwens, L., Pipeleers, D., Van
Schaftingen, E., Madsen, O., and Schuit, F.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 7036
7041
249. Granner, D., and Pilkis, S. (1990) J. Biol. Chem.
265, 1017310176
250. Unson, C. G., Wu, C.-R., and Merrifield, R. B.
(1994) Biochemistry 33, 68846887
251. Lei, K.-J., Pan, C.-J., Liu, J.-L., Shelly, L. L., and
Chou, J. Y. (1995) J. Biol. Chem. 270, 11882
11886
252. Berteloo, A., St-Denis, J.-F., and van de Werve,
G. (1995) J. Biol. Chem. 270, 2109821102
253. Chen, Y.-T., and Burchell, A. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 935
965, McGraw-Hill, New York
254. Pilkis, S. J., El-Maghrabi, M. R., and Claus, T.
C. (1988) Ann. Rev. Biochem. 57, 755783
255. Tornheim, K. (1988) J. Biol. Chem. 263, 2619
2624
256. Scheffler, J. E., and Fromm, H. J. (1986)
Biochemistry 25, 6659 6665
257. Kraus-Friedmann, N. (1986) Trends Biochem.
Sci. 11, 276 279
258. Blackard, W. G., and Clore, J. N. (1988) J. Biol.
Chem. 263, 1672516730
259. Liu, J., Park, E. A., Gurney, A. L., Roesler, W.
J., and Hanson, R. W. (1991) J. Biol. Chem. 266,
1909519102
260. Beale, E. G., Chrapkiewicz, N. B., Scoble, H.
A., Metz, R. J., Quick, D. P., Noble, R. L.,
Donelson, J. E., Biemann, K., and Granner, D.
K. (1985) J. Biol. Chem. 260, 1074810760
261. Scott, D. K., Mitchell, J. A., and Granner, D. K.
(1996) J. Biol. Chem. 271, 31909 31914
261a. Yeagley, D., Moll, J., Vinson, C. A., and Quinn,
P. G. (2000) J. Biol. Chem. 275, 1781417820
262. Hppner, W., Beckert, L., Buck, F., and Seitz,
H.-J. (1991) J. Biol. Chem. 266, 1725717260
263. Punekar, N. S., and Lardy, H. A. (1987) J. Biol.
Chem. 262, 6714 6719
264. Kuwajima, M., Golden, S., Katz, J., Unger, R.
H., Foster, D. W., and McGarry, J. D. (1986) J.
Biol. Chem. 261, 26322637
265. Cline, G. W., and Shulman, G. I. (1995) J. Biol.
Chem. 270, 28062 28067
266. Rognstad, R., and Katz, J. (1972) J. Biol. Chem.
247, 60476054
267. Clark, M. G., Bloxham, D. P., Holland, P. C.,
and Lardy, H. A. (1973) Biochem. J. 134, 589
597
268. Exton, J. H., Friedmann, N., Wong, E. H.-A.,
Brineaux, J. P., Corbin, J. D., and Park, C. R.
(1972) J. Biol. Chem. 247, 35793588
269. Katz, J., Wals, P., and Lee, W.-N. P. (1993) J.
Biol. Chem. 268, 2550925521
270. Des Rosiers, C., Di Donato, L., Comte, B.,
Laplante, A., Marcoux, C., David, F.,
Fernandez, C. A., and Brunengraber, H. (1995)
J. Biol. Chem. 270, 1002710036
270a. Previs, S. F., Hallowell, P. T., Neimanis, K. D.,
David, F., and Brunengraber, H. (1998) J. Biol.
Chem. 273, 1685316859
271. Previs, S. F., Fernandez, C. A., Yang, D.,
Soloviev, M. V., David, F., and Brunengraber,
H. (1995) J. Biol. Chem. 270, 1980619815
271a. Zhang, B. L., Yunianta, and Martin, M. L.
(1995) J. Biol. Chem. 270, 1602316029
272. Neese, R. A., Schwarz, J.-M., Faix, D., Turner,
S., Letscher, A., Vu, D., and Hellerstein, M. K.
(1995) J. Biol. Chem. 270, 14452 14463
273. Martin, G., Chauvin, M.-F., Dugelay, S., and
Baverel, G. (1994) J. Biol. Chem. 269, 26034
26039
274. Fernandez, C. A., and Des Rosiers, C. (1995) J.
Biol. Chem. 270, 1003710042
275. Ni, T.-C., and Savageau, M. A. (1996) J. Biol.
Chem. 271, 7927 7941
276. Shiraishi, F., and Savageau, M. A. (1993) J.
Biol. Chem. 268, 1691716928
277. Wang, H.-C., Ciskanik, L., Dunaway-Mariano,
D., van der Saal, W., and Villafranca, J. J.
(1988) Biochemistry 27, 625633
277a. Moon, Y. S., Latasa, M.-J., Kim, K.-H., Wang,
D., and Sul, H. S. (2000) J. Biol. Chem. 275,
1012110127
277b. Flier, J. S., and Hollenberg, A. N. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1419114192
277c. Barroso, I., Gurnell, M., Crowley, V. E. F.,
Agostini, M., Schwabe, J. W., Soos, M. A., Li
Maslen, G., Williams, T. D. M., Lewis, H.,
Schafer, A. J., Chatterjee, V. K. K., and
ORahilly, S. (1999) Nature (London) 402,
880883
277d. Steppan, C. M., Bailey, S. T., Bhat, S., Brown,
E. J., Banerjee, R. R., Wright, C. M., Patel, H.
R., Ahima, R. S., and Lazar, M. A. (2001)
Nature (London) 409, 307312
277e. Mller, G., Ertl, J., Gerl, M., and Preibisch, G.
(1997) J. Biol. Chem. 272, 10585 10593
277f. Schwartz, M. W. (2000) Science 289, 20662067
278. Newgard, C. B., and McGarry, J. D. (1995)
Ann. Rev. Biochem. 64, 689719
279. Corkey, B. E., Glennon, M. C., Chen, K. S.,
Deeney, J. T., Matschinsky, F. M., and Prentki,
M. (1989) J. Biol. Chem. 264, 2160821612
280. Kudo, N., Barr, A. J., Barr, R. L., Desai, S., and
Lopaschuk, G. D. (1995) J. Biol. Chem. 270,
1751317520
281. Munir, E., Yoon, J. J., Tokimatsu, T., Hattori,
T., and Shimada, M. (2001) Proc. Natl. Acad.
Sci. U.S.A. 98, 1112611130
Ch 17IND page - E - 2/14/03, 11:19 AM 1009
1010 Chapter 17. The Organization of Metabolism
Study Questions
1. Write out a complete step-by-step mechanism for
the reactions by which citrate can be synthesized
from pyruvate and then exported from mitochon-
dria for use in the biosynthesis of fatty acids.
Include a chemically reasonable mechanism for
the action of ATPcitrate lyase, which catalyzes
the following reaction:
ATP + citrate + CoA-SH Acetyl-CoA +
oxaloacetate + ADP + P
i
Show how this reaction can be incorporated into
an ATP-driven cyclic pathway for generating
NADPH from NADH.
2. Show which parts (if any) of the citric acid cycle
are utilized in each of the following reactions and
what, if any, additional enzymes are needed in
each case.
a. Oxidation of acetyl-CoA to CO
2
b. Catabolism of glutamate to CO
2
c. Biosynthesis of glutamate from pyruvate
d. Formation of propionate from pyruvate
3. Here is a possible metabolic reaction for a fungus.
L-Leucine + 2-oxoglutarate
2
+ 2
1
/
2
O
2

L-glutamate

+ citrate
3
+ H
2
O + 2H
+
G (pH 7) = 1026 kJ/mol
Suggest a metabolic pathway for this reaction.
Is it thermodynamically feasible?
4. It has been suggested that in Escherichia coli pyru-
vate may act as the regenerating substrate for a
catalytic cycle by which glyoxylate, OHCCOO

,
is oxidized to CO
2
. Key enzymes in this cycle are
thought to be a 2-oxo-4-hydroxyglutarate aldolase
and 2-oxoglutarate dehydrogenase. Propose a
detailed pathway for this cycle.
5. Some bacteria use a dicarboxylic acid cycle
to oxidize glyoxylate OHC COO

to CO
2
. The
regenerating substrate for this cycle is acetyl-CoA.
It is synthesized from glyoxylate by a complex
pathway that begins with conversion of two
molecules of glyoxylate to tartronic semialdehyde:

OOC CHOH CHO. The latter is then dehydro-


genated to D-glycerate.
Write out a detailed scheme for the dicarboxylate
cycle. Also indicate how glucose and other cell
constituents can be formed from intermediates
created in this biosynthetic pathway.
6. Some bacteria that lack the usual aldolase produce
ethanol and lactic acid in a 1:1 molar ratio via the
heterolactic fermentation. Glucose is converted
to ribulose 5-phosphate via the pentose phosphate
pathway enzymes. A thiamin diphosphate-
dependent phosphoketolase cleaves xylulose
5-phosphate in the presence of inorganic phosphate
to acetyl phosphate and glyceraldehyde 3-phosphate.
Propose a mechanism for the phosphoketolase
reaction and write a balanced set of equations for
the fermentation.
7. Bacteria of the genera Aerobacter and Serratia
ferment glucose according to the following
equation:
1
1
/
2
Glucose 2,3-butanediol + 3 CO
2
+ H
2
+
ethanol
Write out a detailed pathway for the reactions.
Use the pyruvate formatelyase reaction. What
yield of ATP do you expect per molecule of
glucose fermented?
8. Some Clostridia ferment glucose as follows:
2 Glucose + 2 H
2
O butyrate

+ 2 acetate

+
4 CO
2
+ 6 H
2
+ 3 H
+
G (pH 7) = 479 kJ/ mol of butyrate formed
Write out detailed pathways. How much ATP do
you think can be formed per glucose molecule
fermented?
9. Propionic acid bacteria use the following fermen-
tation:
1
1
/
2
Glucose 2 propionate + acetate

+ 3 H
+
+
CO
2
+ H
2
O
G = 464 kJ/ mol of acetate formed
Write out a detailed pathway for the reactions.
How much ATP can be formed per molecule of
glucose?
10. Consider the following reaction which can occur
in the animal body:
2 Lactate

(C
3
H
5
O
3

) + CoASH + 2 NAD
+

butyryl-CoA (C
4
H
7
OSCoA) +
2 CO
2
+ 2 NADH + H
2
O
G (pH 7) 70 kJ/ mol of butyryl-CoA formed
Outline the sequence of reactions involved in this
Ch 17IND page - E - 2/14/03, 11:19 AM 1010
1011
Study Questions
transformation. Do you think that any ATP will
either be used or generated in the reaction?
Explain.
11. Leucine, an essential dietary constituent for
human beings, is synthesized in many bacteria
and plants using pyruvate as a starting material.
Outline this pathway of metabolism and illustrate
the chemical reaction mechanisms involved in
each step.
12. Write a step-by-step sequence for all of the
chemical reactions involved in the biosynthesis
of l-leucine from 2-oxoisovalerate:
Notice that this compound contains one carbon
atom less than leucine. Start by condensing
2-oxoisovalerate with acetyl-CoA in a reaction
similar to that of citrate synthase. Use structural
formulas. Show all intermediate structures and
indicate what coenzymes are needed. Use curved
arrows to indicate the flow of electrons in each step.
13. Some fungi synthesize lysine from 2-oxoglutarate
by elongating the chain using a carbon atom
derived from acetyl-CoA to form the 6-carbon
2-oxoadipate. The latter is converted by an ATP-
dependent reduction to the -aldehyde. Write
out reasonable mechanisms for the conversion
of 2-oxoglutarate to the aldehyde. The latter is
converted on to L-lysine by a nonPLP-dependent
transamination via saccharopine (Chapter 24).
14. Outline the pathway for biosynthesis of L-leucine
from glucose and NH
4
+
in autotrophic organisms.
In addition, outline the pathways for degradation
of leucine to CO
2
, water, and NH
4
+
in the human
body. For this overall pathway or metabolic
loop, mark the locations (one or more) at which
each of the following processes occurs.
a. Synthesis of a thioester by dehydrogenation
b. Substrate-level phosphorylation
c. Thiamin-dependent condensation
d. Oxoacid chain-elongation process
e. Transamination
f. Oxidative decarboxylation of an -oxoacid
g. Partial oxidation
h. Thiolytic cleavage
i. Claisen condensation
j. Biotin-dependent carboxylation
15. A photosynthesizing plant is exposed to
14
CO
2
.
On which carbon atoms will the label first appear
in glucose?
16. The Calvin Benson cycle and the pentose phos-
phate pathway (Eq. 17-12) have many features in
common but run in opposite directions. Since the
synthesis of glucose from CO
2
requires energy,
the energy expenditure for the two processes
will obviously differ. Describe the points in each
pathway where a Gibbs energy difference is used
to drive the reaction in the desired direction.
17. Draw the structure of ribulose 1(
14
C), 5-bisphosphate.
Enter an asterisk (*) next to carbon 1 to show that
this position is
14
C-labeled.
Draw the structures of the products of the ribulose
bisphosphate carboxylase reaction, indicating the
radioactive carbon position with an asterisk.
18. A wood-rotting fungus is able to convert glucose to
oxalate approximately according to the following
equation:
Glucose + 5 O
2
2 Oxalic acid (C
2
O
4
H
2
) +
2 CO
2
+ 4 H
2
O
Propose a mechanism. See Munir et al.
281
for details.
H
3
C CH C COO

CH
3
O
Ch 17IND page - E - 2/14/03, 11:19 AM 1011
1012 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
A. The Architecture of the Mitochondrion
1. The Mitochondrial Membranes and Compartments
2. The Chemical Activities of Mitochondria
3. The Mitochondrial Genome
4. Growth and Development
B. Electron Transport Chains
1. The Composition of the Mitochondrial Electron
Transport System
2. The Sequence of Electron Carriers
Submitochondrial particles and complexes
Mitochondrial electron transport in plants and fungi
Electron transport chains of bacteria
3. Structures and Functions of the Individual
Complexes I - IV and Related Bacterial Assemblies
Complex I, NADH-ubiquinone oxidoreductase
Complex II, succinate-ubiquinone oxidoreductase
Complex III (ubiquinol-cytochrome c oxidoreductase
or cytochrome bc
1
complex)
Complex IV. Cytochrome c oxidase (Ubiquinol-
cytochrome c oxidoreductase)
C. Oxidative Phosphorylation
1. The Stoichiometry (P/O Ratio) and Sites of Oxidative
Phosphorylation
Respiratory control and uncoupling
States of mitochondria and spectrophotometric
observation
2. Thermodynamics and Reverse Electron Flow
3. The Mechanism of Oxidative Phosphorylation
Search for chemical intermediates
Peter Mitchells chemiosmotic theory
Observed values of E
m
and pH
Proton pumps driven by electron transport
4. ATP Synthase
The synthase structure
How is ATP made?
Paul Boyers binding change mechanism
Rotational catalysis
5. ATP-driven Proton Pumps
6. Uncouplers and Energy-linked Processes
in Mitochondria
D. Transport and Exchange across Mitochondrial
Membranes
E. Energy from Inorganic Reactions
1. Reduced Inorganic Compounds as Substrates
for Respiration
The hydrogen-oxidizing bacteria
Nitrifying bacteria
The sulfur-oxidizing bacteria
2. Anaerobic Respiration
Nitrate as an electron acceptor
Reduction of nitrite: denitrification
Sulfate-reducing and sulfur-reducing bacteria
Methane bacteria
1013
1014
1015
1016
1018
1019
1019
1019
1020
1023
1025
1026
1026
1027
1027
1028
1032
1033
1033
1033
1034
1036
1036
1037
1039
1039
1041
1041
1043
1044
1044
1045
1046
1047
1050
1051
1051
1051
1052
1054
1054
1054
1056
1057
F. Oxygenases and Hydroxylases
1. Dioxygenases
2. Monooxygenases
Flavin-containing monooxygenases
Reduced pteridines as cosubstrates
Hydroxylation-induced migration
2-oxoglutarate as a decarboxylating
cosubstrate
Copper-containing hydroxylases
Hydroxylation with cytochrome P450
Other iron-containing oxygenases
Nitric oxide and NO synthases
G. Biological Effects of Reduced Oxygen
Compounds
1. The Respiratory Burst of Neutrophils
2. Oxidative Damage to Tissues
Antioxidant systems
Transcriptional regulation of antioxidant proteins
References
Study Questions
1022
1024
1048
1066
1070
1016
1018
1019
1021
1025
1034
1035
1050
Boxes
Box 18-A Historical Notes on Respiration
Box 18-B Defects of Mitochondrial DNA
Box 18-C Using Metabolism to Generate Heat:
Thermogenic Tissues
Box 18-D Vitamin C: Ascorbic Acid
Box 18-E The Toxicity of Acetaminophen
Tables
Table 18-1 Catalog of Mitochondrial Genes
Table 18-2 Alterations in the Genetic Code in the
DNA of Animal Mitochondria
Table 18-3 Ratios of Components in the Electron
Transport Chain of Mitochondria
Table 18-4 Some Well-Known Respiratory
Inhibitors
Table 18-5 Some Artificial Electron Acceptors
Table 18-6 Wavelengths of Light Used to
Measure States of Oxidation of
Carriers in the Electron Transport
Chain of Mitochondria
Table 18-7 Electrode Potentials of Mitochondrial
Electron Carriers and Gibbs Energy
Changes Associated with Passage of
Electrons
Table 18-8 Some Mitochondrial Membrane
Transporters
1057
1057
1059
1059
1061
1062
1063
1064
1065
1068
1069
1072
1072
1074
1074
1075
1076
1085
Contents
Electrons flowing through the electron transport chains in the membranes of the mitochondria,
(at the left) in this thin section through the retina of a kangaroo rat (Dipodomys ordi) generates
ATP. The ATP provides power for the synthesis and functioning of the stacked photoreceptor
disks seen at the right. The outer segment of each rod cell (See Fig. 23-40), which may be
1520 mm in length, consists of these disks, whose membranes contain the photosensitive
protein pigment rhodopsin. Absorption of light initiates an electrical excitation which is sent
to the brain. Micrograph from Porter and Bonneville (1973) Fine Structure of Cells and Tissues,
Lea and Febiger, Philadelphia, Pennsylvania. Courtesy of Mary Bonneville.
Ch 18IND page - E 2/14/03, 11:07 AM 1012
1013
Electron Transport, Oxidative
Phosphorylation, and Hydroxylation
18
In this chapter we will look at the processes by
which reduced carriers such as NADH and FADH
2
are oxidized within cells. Most familiar to us, because
it is used in the human body, is aerobic respiration.
Hydrogen atoms of NADH, FADH
2
, and other reduced
carriers appear to be transferred through a chain of
additional carriers of increasingly positive reduction
potential and are finally combined with O
2
to form
H
2
O. In fact, the hydrogen nuclei move freely as
protons (or sometimes as H

ions); it is the electrons


that are deliberately transferred. For this reason, the
chain of carriers is often called the electron transport
chain. It is also referred to as the respiratory chain.
Because far more energy is available to cells from
oxidation of NADH and FADH
2
than can be obtained
by fermentation, the chemistry of the electron trans-
port chain and of the associated reactions of ATP
synthesis assumes great importance. A central ques-
tion becomes How is ATP generated by flow of elec-
trons through this series of carriers? Not only is most
of the ATP formed in aerobic and in some anaerobic
organisms made by this process of oxidative phos-
phorylation, but the solar energy captured during
photosynthesis is used to form ATP in a similar man-
ner. The mechanism of ATP generation may also be
intimately tied to the function of membranes in the
transport of ions. In a converse manner, the mecha-
nism of oxidative phosphorylation may be related to
the utilization of ATP in providing energy for the
contraction of muscles.
In some organisms, especially bacteria, energy
may be obtained through oxidation of H
2
, H
2
S, CO, or
Fe
2+
rather than of the hydrogen atoms removed from
organic substrates. Furthermore, some bacteria use
anaerobic respiration in which NO
3

, SO
4
2
, or CO
2
act as oxidants either of reduced carriers or of reduced
inorganic substances. In the present chapter, we will
consider these energy-yielding processes as well as the
chemistry of reactions of oxygen that lead to incorpo-
ration of atoms from O
2
into organic compounds.
The oxidative processes of cells have been hard
to study, largely because the enzymes responsible are
located in or on cell membranes. In bacteria the sites
of electron transport and oxidative phosphorylation
are on the inside of the plasma membrane or on mem-
branes of mesosomes. In eukaryotes they are found
in the inner membranes of the mitochondria and, to a
lesser extent, in the endoplasmic reticulum. For this
reason we should probably start with a closer look at
mitochondria, the power plants of the cell.
A. The Architecture of the Mitochondrion
Mitochondria are present in all eukaryotic cells
that use oxygen in respiration, but the number per cell
and the form and size vary.
1 4
Certain tiny trypano-
somes have just one mitochondrion but some oocytes
have as many as 3 x 10
5
. Mammalian cells typically
contain several hundred mitochondria and liver cells
5
more than 1000. Mammalian sperm cells may contain
50 75 mitochondria,
6
but in some organisms only one
very large helical mitochondrion, formed by the fusion
of many individual mitochondria, wraps around the
base of the tail. Typical mitochondria appear to be
about the size of cells of E. coli. However, study of
ultrathin serial sections of a single yeast cell by elec-
tron microscopy has shown that, under some growth
conditions, all of the mitochondria are interconnected.
7
In every case a mitochondrion is enclosed by two
1.0 m
Ch 18IND page - E 2/14/03, 11:07 AM 1013
1014 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
concentric membranes, an outer and an inner mem-
brane, each ~ 5 7 nm thick (Figs. 18-1, 18-2). The inner
membrane is folded to form the cristae. The number
of cristae, the form of the cristae, and the relative
amount of the internal matrix space are variable. In
liver there is little inner membrane and a large matrix
space, while in heart mitochondria there are more
folds and a higher rate of oxidative phosphorylation.
The enzymes catalyzing the tricarboxylic acid cycle are
also unusually active in heart mitochondria. A typical
heart mitochondrion has a volume of 0.55 m
3
; for
every cubic micrometer of mitochondrial volume there
are 89 m
2
of inner mitochondrial membranes.
9
Mitchondria can swell and contract, and forms
other than that usually seen in osmium-fixed electron
micrographs have been described. In some mitochon-
dria the cristae are swollen, the matrix volume is much
reduced, and the intermembrane space between the
membranes is increased. Rapidly respiring mitochon-
dria fixed for electron microscopy exhibit forms that
have been referred to as energized and energized-
twisted.
10
The micrograph (Fig. 18-1) and drawing
(Fig. 18-2) both show a significant amount of inter-
membrane space. However, electron micrographs of
mitochondria from rapidly frozen aerobic tissues show
almost none.
11
Recent studies by electron microscopic
tomography show cristae with complex tubular struc-
tures. The accepted simple picture of mitochondrial
structure (Fig. 18-1) is undergoing revision.
1212b
The
isolated mitochondria that biochemists have studied
may be fragments of an interlinked mitochondrial
reticulum that weaves its way through the cell.
12b
However, this reticulum may not be static but may
break and reform. The accepted view that the mito-
chondrial matrix space is continuous with the internal
space in the cristae is also the subject of doubts. Perhaps
they are two different compartments.
12a
1. The Mitochondrial Membranes and
Compartments
The outer membranes of mitochondria can be
removed from the inner membranes by osmotic rup-
ture.
13
Analyses on separated membrane fractions
show that the outer membrane is less dense (density
~ 1.1 g / cm
3
) than the inner (density ~ 1.2 g / cm
3
). It
is highly permeable to most substances of molecular
mass 10 kDa or less because of the presence of pores of
~ 2 nm diameter. These are formed by mitochondrial
porins,
14 17
which are similar to the outer membrane
porins of gram-negative bacteria (Fig. 8-20). The ratio
of phospholipid to protein (~ 0.82 on a weight basis) is
much higher than in the inner membrane. Extraction
of the phospholipids by acetone destroys the mem-
brane. Of the lipids present, there is a low content of
cardiolipin, a high content of phosphatidylinositol and
cholesterol, and no ubiquinone.
The inner membrane is impermeable to many sub-
stances. Neutral molecules of <150 Da can penetrate
the membranes, but the permeability for all other
materials including small ions such as H
+
, K
+
, Na
+
,
and Cl

is tightly controlled. The ratio of phospholipid


to protein in the inner membrane is ~ 0.27, and cardio-
lipin makes up ~ 20% of the phospholipid present.
Cholesterol is absent. Ubiquinone and other compo-
nents of the respiratory chain are all found in the inner
membrane. Proteins account for 75% of the mass of
the membrane.
Another characteristic of the inner mitochondrial
membrane is the presence of projections on the inside
surface, which faces the mitochondrial matrix. See
Fig. 18-14. These spherical 85-kDa particles, discovered
by Fernandez Moran in 1962 and attached to the mem-
brane through a stalk, display ATP-hydrolyzing
(ATPase) activity. The latter was a clue that the knobs
might participate in the synthesis of ATP during oxida-
tive phosphorylation. In fact, they are now recognized
as a complex of proteins called coupling factor 1 (F
1
)
or ATP synthase.
In addition to bacterialike mitochondrial ribosomes
and small circular molecules of DNA, mitochondria
may contain variable numbers of dense granules of
calcium phosphate, either Ca
3
(PO
4
)
2
or hydroxylapa-
tite (Fig. 8-34),
4,18
as well as of phospholipoprotein.
4
Figure 18-1 Thin section of mitochondria of a cultured
kidney cell from a chicken embryo. The small, dark, dense
granules within the mitochondria are probably calcium
phosphate. Courtesy of Judie Walton.
Ch 18IND page - E 2/14/03, 11:07 AM 1014
1015
Quantitatively the major constituents of the matrix
space are a large number of proteins. These account
for about 56% by weight of the matrix material and
exist in a state closer to that in a protein crystal than in
a true solution.
1920a
Mitochondrial membranes also
contain proteins, both tightly bound relatively non-
polar intrinsic proteins and extrinsic proteins bound
Figure 18-2 (A) Schematic diagram of mitochondrial structure. (B) Model
showing organization of particles in mitochondrial membranes revealed by
freeze-fracture electron microscopy. The characteristic structural features seen
in the four half-membrane faces (EF and PF) that arise as a result of fracturing
of the outer and inner membranes are shown. The four smooth membrane
surfaces (ES and PS) are revealed by etching. From Packer.
8
Outer membrane
P fracture face
(2806 particles/m
2
)
Outer membrane
E fracture face
(770 particles/m
2
)
Inner membrane
E fracture face
(2120 particles/m
2
)
Inner membrane
P fracture face
(4208 particles/m
2
)
Matrix Cytosol
Outer membrane
Intermembrane space
Inner membrane
Outer membrane
Inner membrane
5-7 nm membrane thickness
When negatively stained
with phosphotungstate
the matrix face of the
inner membrane appears
covered with knobs of
coupling factor F
1
(ATP
synthase, Fig. 18-14)
attached by stalks
Cytochromes, ubiquinone,
and nonheme iron carriers
are embedded in inner
membrane
8.5 nm
~7 nm
Intermembrane space
Inner membrane
is not freely
permeable
Submitochondrial particles
usually have the matrix
side of the inner membrane
on the outside
Outer membrane is
permeable to ATP,
ADP, sucrose,
salts, etc.
Matrix
Cristae
A
B
to the membrane surfaces. The other mitochondrial
compartment, the intermembrane space, may normally
be very small but it is still home for a few enzymes.
2. The Chemical Activities of Mitochondria
Mention of mitochondria usually
brings to the mind of the biochemist
the citric acid cycle, the oxidation
pathway of fatty acid metabolism,
and oxidative phosphorylation.
In addition to these major processes,
many other chemical events also
occur. Mitochondria concentrate
Ca
2+
ions and control the entrance
and exit of Na
+
, K
+
, dicarboxylates,
amino acids, ADP, P
i
and ATP, and
many other substances.
16
Thus, they
exert regulatory functions both on
catabolic and biosynthetic sequences.
The glycine decarboxylase system
(Fig. 15-20) is found in the mito-
chondrial matrix and is especially
active in plant mitochondria (Fig.
23-37). Several cytochrome P450-
dependent hydroxylation reactions,
important to the biosynthesis and
catabolism of steroid hormones and
to the metabolism of vitamin D, take
place within mitochondria. Mito-
chondria make only a few of their
own proteins but take in several
hundred other proteins from the
cytoplasm as they grow and multiply.
Where within the mitochondria
are specific enzymes localized? One
approach to this question is to see
how easily the enzymes can be dis-
sociated from mitochondria. Some
enzymes come out readily under
hypotonic conditions. Some are
released only upon sonic oscillation,
suggesting that they are inside the
matrix space. Others, including the
cytochromes and the flavoproteins
that act upon succinate and NADH,
are so firmly embedded in the inner
mitochondrial membranes that they
can be dissociated only through the
use of non-denaturing detergents.
Because the enzymes of the citric
acid cycle
20a
(with the exception of
succinate dehydrogenase) and
oxidation are present in the matrix,
the reduced electron carriers must
approach the inner membrane from
A. The Architecture of the Mitochondrion
Ch 18IND page - E 2/14/03, 11:07 AM 1015
1016 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
the matrix side (the M side). Thus, the embedded
enzymes designed to oxidize NADH, succinate, and
other reduced substrates must be accessible from
the matrix side. However, sn-glycerol 3-phosphate
dehydrogenase, a flavoprotein, is accessible from the
outside of the cytoplasmic (C side) of the inner
membrane.
21
Fluorescent antibodies to cytochrome c
bind only to the cytoplasmic (intermembrane) side of
the inner membrane, but antibodies to cytochrome
oxidase label both sides, which suggested that this
protein complex spans the membrane.
22
However,
oxidation of cytochrome c by cytochrome oxidase
occurs only on the cytoplasmic surface.
22
Antibodies
to the ATP synthase that makes up the knobs bind
strictly to the matrix side.
The outer mitochondrial membrane contains
monoamine oxidase, cytochrome b
5
, fatty acyl-CoA
synthase, and enzymes of cardiolipin synthesis
22a
as
well as other proteins. Cardiolipin (diphosphatidyl-
glycerol; Fig. 21-4) is found only in the inner mito-
chondrial membrane and in bacteria. It is functionally
important for several mitochondrial enzymes including
cytochrome oxidase and cytochome bc
1
.
22ac
It is also
essential to photosynthetic membranes for which an
exact role in an interaction between the lipid mem-
brane and the associated protein has been revealed by
crystallography.
22d
In other respects the composition
of the inner mitochondrial membrane resembles that
of the membranes of the endoplasmic reticulum.
Isoenzyme III of adenylate kinase, a key enzyme in-
volved in equilibrating ATP and AMP with ADP (Eq.
6-65), is one of the enzymes present in the intermem-
brane space. A number of other kinases, as well as
sulfite oxidase, are also present between the mem-
branes.
4
As mentioned in Box 6-D, mitochondria sometimes
take up calcium ions. The normal total concentration
of Ca
2+
is ~1 mM and that of free Ca
2+
may be only
~0.1 M.
22e,f
However, under some circumstances
mitochondria accumulate large amounts of calcium,
perhaps acting as a Ca
2+
buffer.
22g,f
The so called
ryanodine receptors (Fig. 19-21), prominent in the
endoplasmic reticulum, have also been found in heart
mitochondria, suggesting a function in control of calcium
oscillations.
22i,j
On the other hand, accumulation of
calcium by mitochondria may be pathological and
the activation of Ca
2+
-dependent
proteases may be an initial step in
apoptosis.
22h,22k
3. The Mitochondrial Genome
Each mitochondrion contains
several molecules of DNA (mtDNA),
usually in a closed, circular form,
as well as the ribosomes, tRNA
molecules, and enzymes needed for
protein synthesis.
1,23 26
With rare
exceptions almost all of the mito-
chondrial DNA in a human cell is
inherited from the mother.
6,26a
The
size of the DNA circles varies from
16 19 kb in animals
27
to over 200 kb
in many higher plants. Complete
sequences of many mitochondrial
DNAs are known.
28,28a
Among
these are the 16,569 bp human
mtDNA,
29
the 16,338 bp bovine
mtDNA, the 16,896 bp mtDNA of
the wallaroo Macropus robustus,
30
and the 17,533 bp mtDNA of the
amphibian Xenopus laevis.
31,32
The
sea urchin Paracentotus lividus has
a smaller 15,697 bp genome. How-
ever, the order of the genes in this
and other invertebrate mtDNA is
different from that in mammalian
mitochondria.
33
Protozoal mtDNAs
vary in size from ~ 5900 bp for the
TABLE 18-1
Catalog of Mitochondrial Genes
a
a
Data from Palmer, J. D. (1997) Nature (London) 387, 454 455.
1
b
One for each amino acid of the genetic code but two each for serine and leucine.
Name and symbol Homo
sapiens
Reclino-
monas
americana
Saccharo-
myces
cerevisiae
Arabi-
dopsis
thaliana
Ribosomal RNA
s rRNA (small, 12s) 1 1 1 1
l rRNA (large, 16s) 1 1 1 1
5 S RNA 1 1
Transfer RNAs 22
b
26 24 22
NADH dehydrogenase
Subunits ND1 6, ND4L 7 12 0 9
Cytochrome b 1 1 1 1
Cytochrome oxidase
Subunits I, II, III 3 3 3 3
ATP synthase
Subunits 6, 8, others 2 5 3 4
Total protein coding genes 13 62 8 27
Total genes 37 92 35 53
Size of DNA (kbp) 16.596 69 75 367
Ch 18IND page - E 2/14/03, 11:07 AM 1016
1017
parasitic malaria organism Plasmodium falciparum
34,35
to
41,591 bp for Acanthamoeba castellani
36
and 69,034 bp for
the fresh water flagellate Reclinomonas americana.
26,37
All of the mammalian mtDNAs are organized as
shown in Fig. 18-3. The two strands of the DNA can
be separated by virtue of their differing densities. The
heavy (H) strand has a 5'3' polarity in a counter-
clockwise direction in the map of Fig. 18-3, while the
light (L) strand has a clockwise polarity. From the
sequences 13 genes for specific proteins, 2 genes for
ribosomal RNA molecules, and 22 genes for transfer
RNAs have been identified. The genes are listed in
Table 18-1 and have also been marked on the map
in Fig. 18-3. The map also shows the tRNA genes,
labeled with standard one-letter amino acid abbrevia-
tions, and the directions of transcription. Most of the
protein genes are on the H-strand. One small region,
the D-loop, contains an origin of replication and control
signals for transcription (see Chapters 27 and 28).
The genes in mammalian mtDNA are closely
packed with almost no nucleotides between them.
However, the 19.5-kb mtDNA of Drosophila contains an
(A+T)-rich region, which varies among species.
38
In
the much larger 78-kb genome of yeast Saccharomyces
cerevisiae many (A+T)-rich spacer regions are present.
39
The yeast mitochondrial genome also contains genes
for several additional proteins. Mitochondria of
Reclinomonas americana contain 97 genes, including
those for 5S RNA, the RNA of ribonuclease P as well
as a variety of protein coding genes. Perhaps this
organism is primitive, resembling the original progeni-
tor of eukaryotic life.
26
The mtDNA of trypanosomes
is present in the large mitochondrion or kinetoplast as
40 50 maxicircles ~ 20 35 kb in size, together with
5000 10,000 minicircles, each of 645 2500 bp (see
Fig. 5-16). The latter encode guide RNA for use in
RNA editing (Chapter 28). The large mitochondrial
DNAs of higher plants, e.g., Arabidopsis (Table 18-1),
also contain additional protein genes as well as large
segments of DNA between the genes. The genome of
the turnip (Brassica campestris) exists both as a 218-kb
master chromosome and smaller 83- and 135-kb
incomplete chromosomes, a pattern existing for most
land plants.
40 42
The muskmelon contains 2500 kb of
mitochondrial DNA (mtDNA). On
the other hand, most mtDNA of the
liverwort Marchantia polymorpha
consists of 186-kb linear duplexes.
42a
The compact size of the mam-
malian genome is dependent, in
part, on alterations in the genetic
code, as shown in Table 18-2, and a
modification of tRNA structures that
permits mitochondria to function
with a maximum of 22 tRNAs
(Chapter 28).
43 45
However, the
more primitive Reclinomonas utilizes
the standard genetic code in its
mitochondria.
26
The mammalian
mitochondrial genes contain no
introns, but some yeast mitochon-
drial genes do. Furthermore, some
of the introns contain long open
reading frames. At least two of
these genes-within-genes encode
enzymes that excise the introns.
Why does mtDNA contain any
protein genes, or why does mtDNA
even exist? It seems remarkable that
the cells of our bodies make the 100
or so extra proteins (encoded in the
nucleus) needed for replication,
transcription, amino acid activation,
and mitochondrial ribosome forma-
tion and bring these into the mito-
chondria for the sole purpose of
permitting the synthesis there of
13 proteins. The explanation is not
evident. What are the 13 proteins?
Figure 18-3 Genomic map of mammalian mitochondrial DNA. The stippled
areas represent tRNA genes which are designated by the single-letter amino
acid code; polarity is counterclockwise except for those marked by green
arrow heads. All protein-coding genes are encoded on the H strand (with
counterclockwise polarity), with the exception of ND6, which is encoded on
the L strand. COI, COII, and COIII: cytochrome oxidase subunits I, II, and III;
Cyt b: cytochrome b; ND: NADH dehydrogenase; ATP:ATP synthase. O
H
and
O
L
: the origins of H and L strand replication, respectively. After Wallace
46
and
Shoffner and Wallace.
45
Positions of a few of many known mutations that
cause serious diseases are marked using abbrevations defined in Box 18-B.
Direction of
transcription
from the
H strand
O
H
Direction of
replication
of H strand
Transcription
from L strand
Human
mitochondrial
DNA
16,596 bp
N
D
5
N
D
6
C
y
t
b
1
2
S
rR
N
A
1
6
S
r
R
N
A
N
D
1
C
O
II
N
D
2
N
D
4
C
O
II
I
C
O
I
ND4
ATP8
ND3
K
S
D
W
A
N
C
Y
I
Q
M
L
V
F
P
T
E
H
S
L
Direction of replication
of L strand
O
L
(D-loop)
ATP6
H strand
L strand
MERRF
NARP
MELAS
LHON
LHON
LHON
LHON
A. The Architecture of the Mitochondrion
Ch 18IND page - E 2/14/03, 11:07 AM 1017
1018 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Three are the large functional subunits of cytochrome
oxidase, one is cytochrome b, and seven are subunits
of the NADH dehydrogenase system (Complex I).
Two are subunits of ATP synthase. These are all vitally
involved in the processes of electron transport and
oxidative phosphorylation, but so are other proteins
that are imported from the cytoplasm.
One gene in yeast mtDNA is especially puzzling.
The var 1 gene encodes a mitochondrial ribosomal
protein, whose sequence varies with the strain of
yeast. The gene is also involved in unusual recombi-
national events.
47
Another unusual aspect of yeast
mitochondrial genetics is the frequent appearance of
petite mutants, which grow on an agar surface as
very small colonies. These have lost a large fraction
of their mitochondrial DNA and, therefore, the ability
to make ATP by oxidative phosphorylation. The
remaining petite mtDNA may sometimes become
integrated into nuclear DNA.
48
A few eukaryotes
that have no aerobic metabolism also have no mito-
chondria.
49
4. Growth and Development
Mitochondria arise by division and growth of
preexisting mitochondria. Because they synthesize
only a few proteins and RNA molecules, they must
import many proteins and other materials from the
cytoplasm. A mitochondrion contains at least 100
proteins that are encoded by nuclear genes.
50,50a
The
mechanisms by which proteins are taken up by mito-
chondria are complex and varied. Many of the newly
synthesized proteins carry, at the N terminus, pre-
sequences that contain mitochondrial targeting
signals
51 53
(Chapter 10). These amino acid sequences
often lead the protein to associate with receptor pro-
teins on the outer mitochondrial membrane and sub-
sequently to be taken up by the mitochondria. While
the targeting sequences are usually at the N terminus
of a polypeptide, they are quite often internal. The
N-terminal sequences are usually removed by action
of the mitochondrial processing peptidase (MPP) in
the matrix, but internal targeting sequences are not
removed.
52
Targeting of proteins to mitochondria may
be assisted by mRNA binding proteins that guide
appropriate mRNAs into the vicinity of mitochondria
or other organelles.
53
In addition to targeting signals, polypeptides
destined for the inner mitochondrial membrane contain
additional topogenic signals that direct the polypep-
tide to its destination. These topogenic signals are
distinct from the targeting signals, which they some-
times follow. Topogenic signals are usually hydro-
phobic sequences, which may become transmembrane
segments of the protein in its final location.
52,54
The
uptake of many proteins by mitochondria requires
the electrical potential that is usually present across
the inner membrane (Section E). The fact that mito-
chondrial proteins usually have higher isoelectric
points and carry more positive charges at neutral pH
favors uptake.
55
In addition, chaperonins assist in
TABLE 18-2
Alterations in the Genetic Code in the DNA of
Animal Mitochondria
Codons Nuclear DNA
a
Mitochondrial DNA
AGA, ACG Arg Termination
AUA Ile Met
UGA Termination Trp
a
See Table 5-5 for the other standard codons.
Figure 18-4 Schematic diagram of the protein transport
machinery of mitochondrial membranes labeled according to
the uniform nomenclature.
57
Subunits of outer membrane
receptors and translocase are labeled Tom (translocase of
outer membrane) and those of the inner membrane Tim
(translocase of inner membrane). They are designated
Tom70, etc., according to their sizes in kilodaltons (kDa).
Preproteins are recognized by receptor Tom70Tom 37 and /
or by Tom22Tom20. Clusters of negative charges on many
components help guide the preprotein through the uptake
pores in one or both membranes.
50,58
See Pfanner et al.,
57
Schatz,
50,50a
and Gabriel et al.
50b
70
37
Outer
membrane
6
7
5
20
22
40
23
17
44
Preprotein
+

+ +

Inner
membrane
Tim
Tom

+ + + + + +
+
+ + +
+ + +
Ch 18IND page - E 2/14/03, 11:07 AM 1018
1019
unfolding the protein to be taken up, assist in trans-
port of some proteins,
50a
and may help the imported
proteins to assemble into oligomeric structures.
51 53,56
Protein uptake also requires a set of special pro-
teins described as the translocase of the outer mito-
chondrial membrane (Tom) and translocase of the
mitochondrial inner membrane (Tim). Subunits
that form the receptor targets and transport pores are
designated, according to their approximate molecular
masses in kD as Tom70, Tim23, etc. (Fig. 18-4).
57
Pre-
proteins are recognized by the receptor complexes
Tom70 Tom37 and / or Tom22 Tom20 on the mito-
chondrial surface. They then enter the general import
pore formed by Tom40, Tom6, and Tom7 with the
assistance of a small integral membrane protein Tom5,
which has a positively charged C-terminal membrane
anchor segment and a negatively charged N-terminal
portion that may bind to the positively charged mito-
chondrial targeting sequences.
50,59
A number of other
translocase components, including Tom20 and Tom22
of the outer membrane and Tim23 of the inner mem-
brane, also have acidic extramembranous domains.
58
This suggested an acid chain hypothesis according
to which the targeting signal interacts consecutively
with a series of acidic protein domains that help to
guide it across the two membranes.
50a,58,59
A series of
small proteins, Tim8, 9, 10, 12, 13, function in yeast
mitochondria to mediate the uptake of metabolite
transporters. A defect in the human nuclear gene for a
protein that resembles Tim8 causes deafness dystonia,
a recessive X-linked neurodegenerative disorder.
59a,b
B. Electron Transport Chains
During the 1940s, when it had become clear that
formation of ATP in mitochondria was coupled to
electron transport, the first attempts to pick the system
apart and understand the molecular mechanism began.
This effort led to the identification and at least partial
characterization of several flavoproteins, iron-sulfur
centers, ubiquinones, and cytochromes, most of which
have been described in Chapters 15 and 16. It also
led to the picture of mitochondrial electron transport
shown in Fig. 10-5 and which has been drawn in a
modern form in Fig. 18-5.
1. The Composition of the Mitochondrial
Electron Transport System
Because of the difficulty of isolating the electron
transport chain from the rest of the mitochondrion, it
is easiest to measure ratios of components (Table 18-3).
Cytochromes a, a
3
, b, c
1
, and c vary from a 1:1 to a 3:1
ratio while flavins, ubiquinone, and nonheme iron
occur in relatively larger amounts. The much larger
B. Electron Transport Chains
TABLE 18-3
Ratios of Components in the Electron Transport
Chain of Mitochondria
a,b
Electron carrier
a
From Wainio, W. W. (1970) The Mammalian Mitochondrial Respiratory
Chain, Academic Press, New York, and references cited therein.
b
Molecular ratios are given. Those for the cytochromes refer to the
relative numbers of heme groups.
Rat liver
mitochondria
Beef heart
mitochondria
Cytochrome a
3
1.0 1.1
Cytochrome a 1.0 1.1
Cytochrome b 1.0 1.0
Cytochrome c
1
0.63 0.330.51
Cytochrome c 0.78 0.660.85
Pyridine nucleotides 24
Flavins 3 1
Ubiquinone 36 7
Copper 2.2
Nonheme iron 5.5
amount of pyridine nucleotides is involved in carrying
electrons from the various soluble dehydrogenases of
the matrix to the immobile carriers in the inner mem-
brane, while ubiquinone has a similar function within
the lipid bilayer of mitochondrial membranes.
What are the molar concentrations of the electron
carriers in mitochondrial membranes? In one experi-
ment, cytochrome b was found in rat liver mitochon-
dria to the extent of 0.28 mol/g of protein. If we take
a total mitochondrion as about 22% protein, the average
concentration of the cytochrome would be ~ 0.06 mM.
Since all the cytochromes are concentrated in the inner
membranes, which may account for 10% or less of the
volume of the mitochondrion, the concentration of
cytochromes may approach 1 mM in these membranes.
This is sufficient to ensure rapid reactions with sub-
strates.
2. The Sequence of Electron Carriers
Many approaches have been used to deduce the
sequence of carriers through which electron flow takes
place (Fig. 18-5). In the first place, it seemed reason-
able to suppose that the carriers should lie in order of
increasing oxidationreduction potential going from
left to right of the figure. However, since the redox
potentials existing in the mitochondria may be some-
what different from those in isolated enzyme prepara-
tions, this need not be strictly true.
Ch 18IND page - E 2/14/03, 11:07 AM 1019
1020 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Figure 18-5 A current concept of the electron transport chain of mitochondria. Complexes I, III, and IV pass electrons from
NADH or NADPH to O
2
, one NADH or two electrons reducing one O to H
2
O. This electron transport is coupled to the trans-
fer of about 12 H
+
from the mitochondrial matrix to the intermembrane space. These protons flow back into the matrix
through ATP synthase (V), four H
+
driving the synthesis of one ATP. Succinate, fatty acyl-CoA molecules, and other substrates
are oxidized via complex II and similar complexes that reduce ubiquinone Q, the reduced form QH
2
carrying electrons to
complex III. In some tissues of some organisms, glycerol phosphate is dehydrogenated by a complex that is accessible from
the intermembrane space.
4 H
+
2 in dimensions
Cu
A
Cu
A
a a
3
Cu
B
Cytochrome c
oxidase
IV
III
Q
Q
QH
2
I
[FeS]
[FeS]
II
Fumarate
Succinate
Q
QH
2
[FeS]
ETF
Fatty acyl-CoA
Choline
CN

CO
N
3

FAD

H
2
FAD

Other
complexes
FAD
Fe
c
1
FeS
Q
Fe
c
Fe
b
L
Fe
b
H
[FeS]
[FeS]
[FeS]
FMN
1e

NADH
or
NADPH
Q?
2e

4 H
+
4 H
+
4 H
+
4 H
+
Rotenone
Amytal
Piericidin A
Progesterone
Ascorbate
TMPD Cytochrome c
Respiratory
inhibitors
Glycerol-P
Q
FAD
Matrix
I
Intermembrane space
4 H
+
O
2
(from below)
+ + + + + + + + + + + + +

2H
2
O
H
+
H
+
V
ATP
synthase
(see Fig. 18-14)
Antimycin A
The development by Chance of a dual wavelength
spectrophotometer permitted easy observation of the
state of oxidation or reduction of a given carrier within
mitochondria.
60
This technique, together with the study
of specific inhibitors (some of which are indicated in
Fig. 18-5 and Table 18-4), allowed some electron trans-
port sequences to be assigned. For example, blockage
with rotenone and amytal prevented reduction of the
cytochrome system by NADH but allowed reduction
by succinate and by other substrates having their own
flavoprotein components in the chain. Artificial elec-
tron acceptors, some of which are shown in Table 18-5,
were used to bypass parts of the chain as indicated in
Fig. 18-5.
Submitochondrial particles and complexes.
Many methods have been employed to break mito-
chondrial membranes into submitochondrial particles
that retain an ability to catalyze some of the reactions
of the chain.
61
For example, the KeilinHartree prepa-
ration of heart muscle is obtained by homogenizing
mitochondria and precipitation at low pH.
62
The
resulting particles have a low cytochrome c content
and do not carry out oxidative phosphorylation.
Ch 18IND page - E 2/14/03, 11:07 AM 1020
1021 B. Electron Transport Chains
However, they do transport electrons and react with
O
2
. Other electron transport particles have been pre-
pared by sonic oscillation. Under the electron micro-
scope such particles appear to be small membranous
vesicles resembling mitochondrial cristae.
Many detergents are strong denaturants of pro-
teins, but some of them disrupt mitochondrial mem-
branes without destroying enzymatic activity. A
favorite is digitonin (Fig. 22-12), which causes disinte-
gration of the outer membrane. The remaining frag-
ments of inner membrane retain activity for oxidative
phosphorylation. Such submitochondrial particles can
be fractionated further by chemical treatments. Sepa-
rate complexes can be obtained by treating the inner
membranes with the nondenaturing detergent cholate
(Fig. 22-10) and isolating the complexes by differential
salt fractionation using ammonium sulfate. The isolated
complexes I IV catalyze reactions of four different
portions of the electron transport process
63 65
as indi-
cated in Eq. 18-1:
These complexes are usually named as follows: I,
NADH-ubiquinone oxidoreductase; II, succinate-
ubiquinone oxidoreductase; III, ubiquinol-
cytochrome c oxidoreductase; IV, cytochrome c
oxidase. The designation complex V is sometimes
applied to ATP synthase (Fig. 18-14). Chemical analy-
sis of the electron transport complexes verified the
probable location of some components in the intact
chain. For example, a high iron content was found in
both complexes I and II and copper in complex IV.
We now recognize not only that these complexes
are discrete structural units but also that they are
functional units. Complete X-ray crystallographic
structures are available for complexes III and IV and
for much of the ATP synthase complex. As is indicated
in Fig. 18-5, complexes I IV are linked by two soluble
electron carriers, ubiquinone and cytochrome c.
The lipid-soluble ubiquinone (Q) is present in both
bacterial and mitochondrial membranes in relatively
large amounts compared to other electron carriers
(Table 18-2). It seems to be located at a point of con-
vergence of the NADH, succinate, glycerol phosphate,
and choline branches of the electron transport chain.
Ubiquinone plays a role somewhat like that of NADH,
which carries electrons between dehydrogenases in
the cytoplasm and from soluble dehydrogenases in
the aqueous mitochondrial matrix to flavoproteins
embedded in the membrane. Ubiquinone transfers
electrons plus protons between proteins within the
Succinate
NADH Q cytochrome c O
2
I
II
III IV
(18-1)
O
O
O
H
C CH
2
CH
3
H
3
CO
OCH
3
H
H
O
N
CH
3
CH
3
CH
3
CH
3
CH
3
OH
CH
3
OH
H
3
CO
H
3
CO
HN
N
H
O
O O
CH
3
CH
3
CH
2
CH
3
HN
C
OH
N
H
O
O CH
3
O
H
3
C
O
O CH
3
C
O
R
O CH
3
S
F
3
C
O O
R = n-butyl or n-hexyl
H
O
TABLE 18-4
Some Well-Known Respiratory Inhibitors
a
Rotenone, an insecticide and
fish poison from a plant root
a
See Fig. 18-5 for sites of inhibition.
Progesterone (See Fig. 22-11)
Antimycin A, a Streptomyces
antibiotic
Piericidin A, a structural
analog of ubiquinone
Thenoyltrifluoroacetone
(TTB, 4,4,4-trifluoro-
1-(2-thienyl)-1,3-
butadione
Cyanide

C N
Azide N N N

Carbon monoxide CO
Amytal
(amobarbital)
Ch 18IND page - E 2/14/03, 11:07 AM 1021
1022 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
BOX 18-A HISTORICAL NOTES ON RESPIRATION
Animal respiration has been of serious interest
to scientists since 1777, when Lavoisier concluded
that foods undergo slow combustion within the body,
supposedly in the blood. In 18031807, Spallanzani
established for the first time that the tissues were
the actual site of respiration, but his conclusions
were largely ignored. In 1884, MacMunn discovered
that cells contain the heme pigments, which are
now known as cytochromes. However, the leading
biochemists of the day dismissed the observations
as experimental error, and it was not until the present
century that serious study of the chemistry of bio-
logical oxidations began.
a,b
Recognition that substrates are oxidized
by dehydrogenation is usually attributed to H.
Wieland. During the years 19121922 he showed
that synthetic dyes, such as methylene blue, could
be substituted for oxygen and would allow res-
piration of cells in the absence of O
2
. Subsequent
experiments (see Chapter 15) led to isolation of the
soluble pyridine nucleotides and flavoproteins and
to development of the concept of an electron trans-
port chain.
Looking at the other end of the respiratory
chain, Otto Warburg
c,d
noted in 1908 that all aerobic
cells contain iron. Moreover, iron-containing char-
coal prepared from blood catalyzed nonenzymatic
oxidation of many substances, but iron-free charcoal
prepared from cane sugar did not. Cyanide was
found to inhibit tissue respiration at low concentra-
tions similar to those needed to inhibit nonenzymat-
ic catalysis by iron salts. On the basis of these
investigations, Warburg proposed in 1925 that aerobic
cells contain an iron-based Atmungsferment (respira-
tion enzyme), which was later called cytochrome
oxidase. It was inhibited by carbon monoxide.
Knowing that carbon monoxide complexes of
hemes are dissociated by light, Warburg and Negelein,
in 1928, determined the photochemical action
spectrum (see Chapter 23) for reversal of the carbon
monoxide inhibition of respiration of the yeast
Torula utilis. The spectrum closely resembled the
absorption spectrum of known heme derivatives
(Fig. 16-7). Thus, it was proposed that O
2
, as well as
CO, combines with the iron of the heme group in
the Atmungsferment.
Meanwhile, during 19191925, David Keilin,
while peering through a microscope equipped with
a spectroscopic ocular at thoracic muscles of flies
and other insects, observed a pigment with four
distinct absorption bands. At first he thought it
was derived by some modification of hemoglobin,
but when he found the same pigment in fresh
bakers yeast, he recognized it as an important new
substance. In his words:
e
One day while I was examing a suspension of yeast
freshly prepared from a few bits of compressed yeast
shaken vigorously with a little water in a test-tube, I
failed to find the characteristic four-banded absorption
spectrum, but before I had time to remove the suspension
from the field of vision of the microspectroscope the four
absorption bands suddenly reappeared. This experiment
was repeated time after time and always with the same
result: the absorption bands disappeared on shaking the
suspension with air and reappeared within a few seconds
on standing.
I must admit that this first visual perception of an
intracellular respiratory process was one of the most
impressive spectacles I have witnessed in the course of
my work. Now I have no doubt that cytochrome is not
only widely distributed in nature and completely inde-
pendent of haemoglobin, but that it is an intracellular
respiratory pigment which is much more important than
haemoglobin.
Keilin soon realized that three of the absorption
bands, those at 604, 564, and 550 nm (a, b, and c),
represented different pigments, while the one at 521
nm was common to all three. Keilin proposed the
names cytochromes a, b, and c. The idea of an elec-
tron transport or respiratory chain followed
e
quickly
as the flavin and pyridine nucleotide coenzymes
were recognized to play their role at the dehydro-
genase level. Hydrogen removed from substrates
by these carriers could be used to oxidize reduced
cytochromes. The latter would be oxidized by
oxygen under the influence of cytochrome oxidase.
In 1929, Fiske and Subbarow,
d,fh
curious about
the occurrence of purine compounds in muscle
extracts, discovered and characterized ATP. It
was soon shown (largely through the work of
Lundsgaard and Lohman)
f
that hydrolysis of ATP
provided energy for muscular contraction. At about
the same time, it was learned that synthesis of ATP
accompanied glycolysis. That ATP could also be
formed as a result of electron transport became clear
following an observation of Engelhardt
h,i
in 1930,
that methylene blue stimulated ATP synthesis by
tissues.
The study of electron transport chains and of
oxidative phosphorylation began in earnest after
Kennedy and Lehninger,
j
in 1949, showed that
mitochondria were the site not only of ATP synthe-
sis but also of the operation of the citric acid cycle
and fatty acid oxidation pathways. By 1959, Chance
had introduced elegant new techniques of spectro-
photometry that led to formulation of the electron
Ch 18IND page - E 2/14/03, 11:07 AM 1022
1023
BOX 18-A (continued)
transport chain as follows:
Substrate pyridine nucleotides flavoprotein
cyt b cyt c cyt a cyt a
3
O
2
Since that time, some new components have
been added, notably the ubiquinones and iron-
sulfur proteins, but the basic form proposed for the
chain was correct.
a
Kalckar, H. M. (1969) Biological Phosphorylations, Prentice-Hall,
Englewood Cliffs, New Jersey
b
Kalckar, H. M. (1991) Ann. Rev. Biochem. 60, 137
c
Edsall, J. T. (1979) Science 205, 384385
d
Fiske, C. H., and Subbarow, Y. (1929) Science 70, 381382
e
Keilin, D. (1966) The History of Cell Respiration and Cytochrome,
Cambridge Univ. Press, London and New York
f
Kalckar, H. (1980) Trends Biochem. Sci. 5, 5657
g
Schlenk, F. (1987) Trends Biochem. Sci. 12, 367368
h
Saraste, M. (1998) Science 283, 14881493
i
Slater, E. C. (1981) Trends Biochem. Sci. 6, 226227
j
Talalay, P., and Lane, M. D. (1986) Trends Biochem. Sci. 11, 356358
membrane bilayer. Membranes also contain ubiqui-
none-binding proteins,
66,67
which probably hold the
ubiquinone that is actively involved in electron trans-
port. Perhaps some ubiquinone molecules function
as fixed carriers. There is also uncertainty about the
number of sites at which ubiquinone functions in the
chain.
Mitochondrial electron transport in plants and
fungi. Plant mitochondria resemble those of mammals
in many ways, but they contain additional dehydro-
genases and sometimes utilize alternative pathways of
electron transport,
68 73
as do fungi.
74
Mitochondria are
impermeable to NADH and NAD
+
. Animal mitochon-
dria have shuttle systems (see Fig. 18-16) for bringing
the reducing equivalents of NADH into mitochondria
and to the NADH dehydrogenase that faces the matrix
side of the inner membrane. However, plant mito-
chondria also have an NADH dehydrogenase on
the outer surface of the inner membrane (Fig. 18-6).
This enzyme transfers electrons to ubiquinone, is not
inhibited by rotenone (see Fig. 18-5), and also acts on
NADPH. Inside the mitochondria a high-affinity
NADH dehydrogenase resembles complex I of animal
mitochondria and is inhibited by rotenone.
73
There is
also a low-affinity NADH dehydrogenase, which is
insensitive to rotenone. Some plant mitochondria
respire slowly in the presence of cyanide. They utilize
an alternative oxidase that replaces complex III and
cytochome c oxidase and which is not inhibited by
antimycin or by cyanide (Fig. 18-6).
68,71,75
It is especially
active in thermogenic plant tissues (Box 18-C). A
D
I III IV
Q QH
2
Succinate
Fumarate
b
FeS
c
NADH
NAD
+
NADH
NAD
+
Glycerol-P
Dihydroxy-
acetone-P
NAD(P)H
NAD(P)
+
O
2
H
2
O
O
2
H
2
O
Matrix
C
B
c
1
Cytosol (Intermembrane)
II
A
Figure 18-6 Schematic diagram of some mitochondrial dehydrogenase and oxidase complexes of plants and also the glycerol
phosphate dehydrogenase of animals, which is embedded in the inner membrane. Complexes IIV are also shown. (A) The
glycerol phosphate dehydrogenase of some animal tissues. It is accessible from the intermembrane space on the cytosolic
side. (B) The rotenone-insensitive NAD(P)H dehydrogenase of the external membrane surface of plants. (C) The rotenone-
insensitive NADH dehydrogenase facing the matrix side in some plants. (D) The plant alternative oxidase. Ubiquinone, Q.
The three green stippled dehydrogenases are not coupled to proton pumps or ATP synthesis. After Hoefnagel et al.
73
B. Electron Transport Chains
Ch 18IND page - E 2/14/03, 11:07 AM 1023
1024 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
BOX 18-B DEFECTS OF MITOCHONDRIAL DNA
A mutation in any of the 13 protein subunits, the
22 tRNAs, or the two rRNAs whose genes are carried
in mitochondrial DNA may possibly cause disease.
The 13 protein subunits are all involved in electron
transport or oxidative phosphorylation. The syn-
dromes resulting from mutations in mtDNA fre-
quently affect oxidative phosphorylation (OXPHOS)
causing what are often called OXPHOS diseases.
a g
Mitochondrial oxidative phosphorylation also de-
pends upon ~ 100 proteins encoded in the nucleus.
Therefore, OXPHOS diseases may result from defects
in either mitochondrial or nuclear genes. The former
are distinguished by the fact that they are inherited
almost exclusively maternally. Most mitochondrial
diseases are rare. However, mtDNA is subject to
rapid mutation, and it is possible that accumulating
mutants in mtDNA may be an important component
of aging.
h k
The first recognition of mitochondrial disease
came in 1959. A 30-year old Swedish woman was
found to have an extremely high basal metabolic rate
(180% of normal), a high caloric intake (>3000 kcal /
day), and an enormous perspiration rate. She had
developed these symptoms at age seven. Examina-
tion of her mitochondria revealed that electron trans-
port and oxidative phosphorylation were very
loosely coupled. This explains the symptoms. How-
ever, the disease (Luft disease) is extremely rare and
the underlying cause isnt known.
i
Its recognition
did focus attention on mitochondria, and by 1988,
120 different mtDNA defects had been described.
e,i
Some OXPHOS disorders, including Luft dis-
ease, result from mutations in nuclear DNA. A sec-
ond group arise from point mutations in mtDNA and
a third group involve deletions, often very large, in
mtDNA. Persons with these deletions survive be-
cause they have both mutated and normal mtDNA, a
condition of heteroplasmy of mtDNA. As these
persons age their disease may become more severe
because they lose many normal mitochondria.
d,e
The names of mitochondrial diseases are often
complex and usually are described by abbreviations.
Here are a few of them: LHON, Lebers hereditary
optical neuropathy; MERRF, myoclonic epilepsy
and ragged-red-fiber disease; MELAS, mitochondri-
al encephalomyopathy, lactic acidosis, and strokelike
episodes; NARP, neurological muscle weakness,
ataxia, and retinitis pigmentosa; Leigh disease
SNE, subacute necrotizing encephalomyelopathy;
KSS, KearnsSayre syndrome; CPEO, chronic
progressive external ophthalmoplegia. LHON is a
hereditary disease that often leads to sudden blind-
ness from death of the optic nerve especially among
males. Any one of several point mutations in sub-
units ND1, 2, 4, 5, and 6 of NADH dehydrogenase
(complex I; Figs. 18-3 and 18-5), cytochrome b of
complex II, or subunit I of cytochrome oxidase may
cause this syndrome. Most frequent is an R340H
mutation of the ND4 gene at position 11,778 of mtD-
NA (Fig. 18-3).
e,l,m
It may interfere with reduction of
ubiquinone.
n
Mutations in the ND1 gene at position
3460 and in the ND6 gene at position 14484 or in the
cytochrome b gene at position 15257 cause the same
disease.
l
The most frequent (80 90%) cause of MER-
RF, which is characterized by epilepsy and by the
appearance of ragged red fibers in stained sections of
muscle, is an A G substitution at position 8344 of
mtDNA in the TC loop (Fig. 5-30) of mitochondrial
tRNA
Lys
. A similar disease, MELAS, is accompanied
by strokes (not seen in MERRF) and is caused in 80%
of cases by an A G substitution in the dihydrouri-
dine loop (Fig. 5-30) of mitochondrial tRNA
Leu
.
o
CPEO, Leigh disease, and KSS often result from large
deletions of mtDNA.
p
NARP and related conditions
have been associated with an L156R substitution in
the ATPase 6 gene of ATP synthase.
q
Can mitochondrial diseases be treated? At-
tempts are being made to improve the function of
impaired mitochondria by adding large amounts of
ubiquinone, vitamin K, thiamin, riboflavin, and
succinate to the diet.
e
One report suggests that mito-
chondrial decay during aging can be reversed by
administration of N-acetylcarnitine.
k
a
Palca, J. (1990) Science 249, 11041105
b
Capaldi, R. A. (1988) Trends Biochem. Sci. 13, 144148
c
Darley-Usmar, V., ed. (1994) Mitochondria: DNA, Proteins and
Disease, Portland Press, London
d
Wallace, D. C. (1999) Science 283, 14821488
e
Shoffner, J. M., and Wallace, D. C. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 15351609,
McGraw-Hill, New York
f
Schon, E. A. (2000) Trends Biochem. Sci. 25, 555560
g
Tyler, D. D. (1992) The Mitochondrion in Health and Disease, VCH
Publ., New York
h
Wallace, D. C. (1992) Science 256, 628632
i
Luft, R. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 87318738
j
Tanhauser, S. M., and Laipis, P. J. (1995) J. Biol. Chem. 270,
2476924775
k
Shigenaga, M. K., Hagen, T. M., and Ames, B. N. (1994) Proc.
Natl. Acad. Sci. U.S.A. 91, 1077110778
l
Hofhaus, G., Johns, D. R., Hurko, O., Attardi, G., and Chomyn,
A. (1996) J. Biol. Chem. 271, 1315513161
m
Brown, M. D., Trounce, I. A., Jun, A. S., Allen, J. C., and Wallace,
D. C. (2000) J. Biol. Chem. 275, 3983139836
n
Zickermann, V., Barquera, B., Wikstrm, M., and Finel, M.
(1998) Biochemistry 37, 1179211796
o
Hayashi, J.-I., Ohta, S., Kagawa, Y., Takai, D., Miyabayashi, S.,
Tada, K., Fukushima, H., Inui, K., Okada, S., Goto, Y., and
Nonaka, I. (1994) J. Biol. Chem. 269, 1906019066
p
Moraes, C. T., and 19 other authors. (1989) N. Engl. J. Med. 320,
12931299
q
Hartzog, P. E., and Cain, B. D. (1993) J. Biol. Chem. 268, 1225012252
Ch 18IND page - E 2/14/03, 11:07 AM 1024
1025
N N
CH
3
CH
3
H
3
C
H
3
C
N O
Cl
Cl
OH
N
N
CH
3
S
N
N
CH
3
H
3
C
+ +
1/2 SO
4
2
N
CH
3
CH
3
O
CH
3
Fe(CN)
6
3
C
N
N
N
N
+
+
2
O

+
TABLE 18-5
Some Artificial Electron Acceptors
a,b
Compound Structure E(pH 7)
30C
a
From Wainio, W. W. (1970) The Mammalian Mitochondrial Respiratory Chain, Academic
Press, New York, pp. 106111.
b
See Fig. 18-5 for sites of action.
Tetrazolium
salts, e.g.,
neotetrazolium
chloride
Menadione
Methylene blue
Ascorbate
Phenazine
methosulfate (PMS)
2,6-Dichlorophenol-
indophenol (DCIP)
Oxidized form
of tetramethyl-p-
phenylenediamine
Ferricyanide
0.125 V
+0.008 V
(25C)
+0.011 V
+0.058 V
+0.080 V
+0.217 V
+0.260 V
+0.36 V
(25C)
(See Box 18-D)
similar oxidase is present in try-
panosomes.
72
Neither the rotenone-
insensitive dehydrogenases nor the
alternative oxidases are coupled to
synthesis of ATP.
Electron transport chains of
bacteria. The bacterial electron
transport systems are similar to that
of mitochondria but simpler. Bacte-
ria also have a variety of alternative
pathways that allow them to adapt
to various food sources and envi-
ronmental conditions.
76,77
The
gram-negative soil bacterium Para-
coccus denitrificans, which has been
called a free-living mitochondrion,
has a mammalian-type respiratory
system. Its complexes IIV resemble
those of animals and of fungi,
7879
but Paracoccus has fewer subunits in
each complex. Complex I of E. coli
is also similar to that of our own
bodies.
7980
However, other major
flavoprotein dehydrogenases in E.
coli act on D-lactate and sn-3-glycerol
phosphate.
81
Pyruvate is oxidized
by a membrane-bound flavoprotein
(Fig. 14-2). All of these enzymes
pass electrons to ubiquinone-8 (Q
8
).
82
Succinate dehydrogenase of E. coli
resembles that of mitochondria,
83
and the ubiquinol oxidase of Para-
coccus resembles complexes III + IV
of mitochondria. It can be resolved
into a three-subunit bc
1
complex, a
three-subunit c
1
aa
3
complex, and
another 57-kDa peptide.
84
The last
contains a 22-kDa cytochrome c
552
,
which is considerably larger than
mitochondrial cytochrome c.
The cytochrome aa
3
terminal
oxidase is produced constitutively,
i.e., under all conditions. However,
when cells are grown on succinate
or H
2
another set of enzymes is
produced with the b-type cyto-
chrome o
3
as the terminal quinol
oxidase (Eq. 18-2).
85
(18-2)
H
2
Succinate Q cytochrome b
cytochrome o
3
O
2

B. Electron Transport Chains


Ch 18IND page - E 2/14/03, 11:08 AM 1025
1026 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
unidentified cofactors.
79
Complex I from E. coli is
smaller, containing only 14 subunits. These are encoded
by a cluster of 14 genes, which can be directly related
by their sequences to subunits of mitochondrial com-
plex I and also to the corresponding genes of Paracoc-
cus denitrificans.
80,91
Complex I of Neurospora contains
at least 35 subunits.
89
The 14 subunits that are present
both in bacteria and in mitochondria probably form
the structural core of the complex. The other subunits
thicken, strengthen, and rigidify the arms. Some of
the extra subunits have enzymatic activities that are
not directly related to electron transport. Among these
are a 10-kDa prokaryotic type acyl carrier protein
(ACP), which may be a relic of a bacterial fatty acid
synthase, reflecting the endosymbiotic origin of mito-
chondria.
92
Also present is a 40-kDa NAD(P)H depen-
dent reductase / isomerase, which may be involved in
a biosynthetic process, e.g., synthesis of a yet unknown
redox group.
79,92
In all cases, FMN is apparently the immediate
acceptor of electrons from NADH. From the results
of extrusion of the Fe S cores (Chapter 16) and EPR
measurements it was concluded that there are three
tetranuclear (Fe
4
S
4
) ironsulfur centers and at least
two binuclear (Fe
2
S
2
) centers
93,94
as well as bound
ubiquinone.
95
Chemical analysis of iron and sulfide
suggested up to eight Fe S clusters per FMN, while
gene sequences reveal potential sites for formation of
six Fe
4
S
4
clusters and two Fe
2
S
2
clusters.
78
Treatment
of complex I with such chaotropic agents as 2.5 M
urea or 4 M sodium trichloroacetate followed by frac-
tionation with ammonium sulfate
95
gave three fractions:
Two terminal quinol oxidase systems, both related to
cytochrome c oxidase, are utilized by E. coli to oxidize
ubiquinol-8. When cultured at high oxygen tensions,
cytochrome bo
3
(also called cytochrome bo) is the major
oxidase. It utilizes heme o (Fig. 16-5) instead of heme
a. However, at low oxygen tension, e.g., in the late
logarithmic stage of growth, the second oxidase, cyto-
chrome bd, is formed.
76,86 88a
It contains two molecules
of the chlorin heme d (Fig. 16-5), which appear to be
involved directly in binding O
2
. This terminal oxidase
system is present in many bacteria and can utilize
either O
2
or nitrite as the oxidant. A simpler electron
transport chain appears to be involved in the oxidation
of pyruvate by E. coli. The flavoprotein pyruvate oxi-
dase passes electrons to Q
8
, whose reduced form can
pass electrons directly to cytochrome d. Incorporation
of these two pure protein complexes and ubiquinone-8
into phospholipid vesicles has given an active recon-
stituted chain.
82
Other bacteria utilize a variety of
quinol oxidase systems, which contain various combi-
nations of cytochromes: aa
3
, caa
3
, cao, bo
3
, and ba
3
.
88b,c
3. Structures and Functions of the Individual
Complexes I IV and Related Bacterial
Assemblies
What are the structures of the individual electron
transport complexes? What are the subunit composi-
tions? What cofactors are present? How are electrons
transferred? How are protons pumped? We will con-
sider these questions for each of complexes I IV, as
found in both prokaryotes and eukaryotes.
88d,e
Complex I, NADH-ubiquinone oxidoreductase.
Complex I oxidizes NADH, which is generated within
the mitochondrial matrix by many dehydrogenases.
Among these are the pyruvate, 2-oxoglutarate, malate,
and isocitrate dehydrogenases, which function in the
tricarboxylic acid cycle; the -oxoacyl-CoA dehydro-
genase of the oxidation system for fatty acids; and
2-hydroxybutyrate, glutamate, and proline dehydro-
genases. All produce NADH, which reacts with the
flavoprotein component of complex I. Whether from
bacteria,
79
fungal mitochondria,
89
or mammalian
mitochondria
89a,90
complex I exists as an L-shaped
object, of which each of the two arms is ~ 23 nm long.
One arm projects into the matrix while the other lies
largely within the inner mitochondrial membrane
(Fig. 18-7). The mitochondrial complex, which has a
mass of ~ 1 MDa, has the same basic structure as the
530-kDa bacterial complex. However, the arms are
thicker in the mitochondrial complex. Analysis of the
denatured proteins by gel electrophoresis revealed at
least 43 peptides.
78,90
Bound to some of these are the
electron carriers FMN, Fe
2
S
2
, and Fe
4
S
4
clusters, ubi-
quinone or other quinones, and perhaps additional
Figure 18-7 Three-dimensional image of bovine NADH-
Ubiquinone oxidoreductase (complex I) reconstructed from
individual images obtained by electron cyro-microscopy.
The resolution is 2.2 nm. The upper portion projects into the
mitochondrial matrix while the horizontal part lies within
the membrane as indicated. Courtesy of N. Grigorieff.
90
Ch 18IND page - E 2/14/03, 11:08 AM 1026
1027
(1) A soluble NADH dehydrogenase consisting of a
51-kDa peptide that binds both the FMN and also one
tetranuclear Fe S cluster (designated N3) and a 24-kDa
peptide that carries a binuclear Fe S center designated
N1b. (2) A 75-kDa peptide bearing two binuclear Fe S
centers, one of which is called N1a and also 47-, 30-,
and 13-kDa peptides. One of these carries tetranuclear
center N4. (3) A group of insoluble, relatively nonpolar
proteins, one of which carries tetranuclear cluster N2.
It may be the immediate donor of electrons to a ubiqui-
none held by a ubiquinone-binding protein designated
QP-N. In bacteria seven of these are homologs of the
seven NADH dehydrogenase subunits encoded by
mtDNA (Fig. 18-3). A 49-kDa subunit of complex I in
the yeast Yarrowia lipolytica is strikingly similar to the
hydrogen reactive subunit of NiFe hydrogenases (Fig.
16-26).
95a
These proteins are thought to lie within the
membrane arm and to form ~ 55 transmembrane
helices. Ubiquinone may also function as a carrier
within complex I,
96,97
and there may be a new redox
cofactor as well.
79
The following tentative sequence
(Eq. 18-3) for electron transfer within complex I (with
apparent E

values of carriers) has been suggested.


By equilibration with external redox systems, the redox
potentials of these centers within the mitochondria
have been estimated and are given (in V) in Eq. 18-3.
The presence of a
large fraction of the bound ubiquinone as a free radical
suggests that the quinone functions as a one-electron
acceptor rather than a two-electron acceptor. A charac-
teristic of complex I is inhibition by rotenone or pier-
icidin, both of which block electron transport at the site
indicated in Fig. 18-5.
Complex II, succinate-ubiquinone oxido-
reductase. Complex II, which carries electrons from
succinate to ubiquinone, contains covalently linked
8

-(N-histidyl)-FAD (Chapter 15) as well as Fe S


centers and one or more ubiquinone-binding sites.
There are four subunits whose structures and proper-
ties have been highly conserved among mitochondria
and bacteria and also in fumarate reductases. The
latter function in the opposite direction during anaero-
bic respiration with fumarate as the terminal oxidant,
both in bacteria
9899a
and in parasitic helminths and
other eukaryotes that can survive prolonged anaerobic
conditions (Chapter 17, Section F,2).
100
Complex II
from E. coli consists of 64-, 27-, 14-, and 13-kDa sub-
units, which are encoded by genes sdhCDAB of a single
operon.
101 103
The two larger hydrophilic subunits
associate to form the readily soluble succinate dehy-
drogenase. The 64-kDa subunit carries the covalently
bound FAD while the 27-kDa subunit carries three
Fe S centers. The two small 13- and 14-kDa subunits
form a hydrophobic anchor and contain a ubiquinol-
binding site (QD-S)
103
as well as a heme that may
bridge the two subunits
102
to form cytochrome b
556
.
The functions of the heme is uncertain. The soluble
mammalian succinate dehydrogenase resembles closely
that of E. coli and contains three Fe S centers: binuclear
S1 of E 0 V, and tetranuclear S2 and S3 of 0.25 to 0.40
and + 0.065 V, respectively. Center S3 appears to
operate between the 2 and 1 states of Eq. 16-17 just
as does the cluster in the Chromatium high potential
iron protein. The function of the very low potential S2
is not certain, but the following sequence of electron
transport involving S1 and S3 and the bound ubiqui-
none QD S
66
has been proposed (Eq. 18-4).
Succinate (FAD*, S1) S3 QP S Q
In addition to complexes I and II several other
membrane-associated FAD-containing dehydrogenase
systems also send electrons to soluble ubiquinone.
These include dehydrogenases for choline, sn-glycerol
3-phosphate, and the electron-transfer protein (ETF) of
the fatty acyl-CoA oxidation system (Fig. 18-5). The
last also accepts electrons from dehydrogenases for
sarcosine (N-methylglycine), dimethylglycine, and
other substrates. The sn-glycerol 3-phosphate dehy-
drogenase is distinguished by its accessibility from the
intermembrane (cytosolic) face of the inner mitochon-
drial membrane (Fig. 18-6).
Complex III (ubiquinol-cytochrome c oxido-
reductase or cytochrome bc
1
complex). Mitochon-
drial complex III is a dimeric complex, each subunit
of which contains 11 different subunits with a total
molecular mass of ~ 240 kDa per monomer.
104 107
However, in many bacteria the complex consists of
only three subunits, cytochrome b, cytochrome c
1
,
and the high potential (~ 0.3 V) Rieske iron-sulfur
protein, which is discussed in Chapter 16, Section A,7.
These three proteins are present in all bc
1
complexes.
In eukaryotes the 379-residue cytochrome b is mito-
chondrially encoded. Although there is only one
cytochrome b gene in the mtDNA, two forms of cyto-
chrome b can be seen in absorption spectra: b
H
(also
called b
562
or b
K
) and lower potential b
L
(also called b
566
or b
T
).
107a,b
X-ray diffraction studies have revealed the complete
11-subunit structure of bovine bc
1
complex
104,106107
as
well as a nearly complete structure of the chicken bc
1
complex (Fig. 18-8).
105
The bovine complex contains
2166 amino acid residues per 248-kDa monomer and
B. Electron Transport Chains
NADH FMN N1a (N3,
Fe
2
S
2
0.32
Fe
4
S
4
Fe
2
S
2
Fe
4
S
4
Fe
4
S
4
0.38 0.24 0.21
N1b, N4) N2 QP-N Q
V
rotenone

(18-3)
(18-4)
Ch 18IND page - E 2/14/03, 11:08 AM 1027
1028 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
exists in crystals as a 496-kDa dimer and probably func-
tions as a dimer.
106-107
The two hemes of cytochrome b
are near the two sides of the membrane, and the Fe S
and cytochrome c
1
subunits are on the surface next to
the intermembrane space (Fig. 18-8). On the matrix
side (bottom in Fig. 18-8A) are two large ~ 440 residue
core subunits that resemble subunits of the mito-
chondrial processing protease. They may be evolu-
tionary relics of that enzyme.
106,108,108a
Mitochondrial
cytochrome b
H
has an E value of +0.050 V, while that
of b
L
is 0.090 V at pH 7.
109
That of the Rieske Fe S
protein is + 0.28 V.
110
The sequence of electron transport within complex
III has been hard to determine in detail. For reasons
discussed in Section C, the Q-cycle shown in Fig. 18-9
has been proposed.
111 114a
As is indicated in Fig. 18-9,
complex II accepts electrons from QH
2
and passes
them consecutively to the Fe S protein, cytochrome c
1
,
and the external cytochrome c. However, half of the
electrons are recycled through the two heme groups of
cytochrome b, as is indicated in the figure and explained
in the legend. The X-ray structure (Fig. 18-8) is consis-
tent with this interpretation. Especially intriguing is
the fact that the Fe
2
S
2
cluster of the Rieske protein
subunit has been observed in two or three different
conformations.
105 107,114a-c
In Fig. 18-8C the structures
of two conformations are superimposed. The position
of the long helix at the right side is unchanged but the
globular domain at the top can be tilted up to bring
the Fe
2
S
2
cluster close to the heme of cytochrome c
1
, or
down to bring the cluster close to heme b
L
. Movement
between these two positions is probably part of the
catalytic cycle.
115
The simpler cytochrome bc
1
complexes of bacteria
such as E. coli,
102
Paracoccus dentrificans,
116
and the
photosynthetic Rhodobacter capsulatus
117
all appear to
function in a manner similar to that of the large mito-
chondrial complex. The bc
1
complex of Bacillus subtilis
oxidizes reduced menaquinone (Fig. 15-24) rather
than ubiquinol.
118
In chloroplasts of green plants
photochemically reduced plastoquinone is oxidized by
a similar complex of cytochrome b, c-type cytochrome
f, and a Rieske Fe S protein.
119120a
This cytochrome
b
6
f complex delivers electrons to the copper protein
plastocyanin (Fig. 23-18).
The electron acceptor for complex III is cytochrome
c, which, unlike the other cytochromes, is water soluble
and easily released from mitochondrial membranes.
Nevertheless, it is usually present in a roughly 1:1 ratio
with the fixed cytochromes, and it seems unlikely that
it is as free to diffuse as are ubiquinone and NAD
+
.
121,122
However, a small fraction of the cytochrome c may
diffuse through the intermembrane space and accept
electrons from cytochrome b
5
, which is located in the
outer membrane.
123
Cytochrome c forms a complex
with cardiolipin (diphosphatidylglycerol), a character-
istic component of the inner mitochondrial membrane.
124
Complex IV. Cytochrome c oxidase (ubiquinol-
cytochrome c oxidoreductase). Complex IV from
mammalian mitochondria contains 13 subunits. All of
them have been sequenced, and the three-dimensional
structure of the complete complex is known (Fig. 18-
10).
125 127
The simpler cytochrome c oxidase from
Paracoccus denitrificans is similar but consists of only
three subunits. These are homologous in sequence to
those of the large subunits I, II, and III of the mito-
chondrial complex. The three-dimensional structure
of the Paracoccus complex is also known. Its basic
structure is nearly identical to that of the catalytic core
of subunits I, II, and III of the mitochondrial complex
(Fig. 18-10,A).
128
All three subunits have transmem-
brane helices. Subunit III seems to be structural in
function, while subunits I and II contain the oxido-
reductase centers: two hemes a (a and a
3
) and two
different copper centers, Cu
A
(which contains two
Cu
2+
) and a third Cu
2+
(Cu
B
) which exists in an EPR-
silent exchange coupled pair with a
3
. Bound Mg
2+
and Zn
2+
are also present in the locations indicated in
Fig. 18-10.
The Cu
A
center has an unusual structure.
130 132
It
was thought to be a single atom of copper until the
three-dimensional structure revealed a dimetal center,
whose structure follows. The Cu
B
-cytochrome a
3
center is also unusual. A histidine ring is covalently
attached to tyrosine.
133 135a
Like the tyrosine in the
active site of galactose oxidase (Figs. 16-29, 16-30),
which carries a covalently joined cysteine, that of
cytochrome oxidase may be a site of tyrosyl radical
formation.
135
Cytochrome c oxidase accepts four electrons, one
at a time from cytochrome c, and uses them to reduce
O
2
to two H
2
O. Electrons enter the oxidase via the Cu
A
center and from there pass to the cytochrome a and on
to the cytochrome a
3
Cu
B
center where the reduction
of O
2
takes place. A possible sequence of steps in the
catalytic cycle is given in Fig. 18-11. Reduction of O
2
to
two H
2
O requires four electrons and also four protons.
An additional four protons are evidently pumped
across the membrane for each catalytic cycle.
136 138
The overall reaction is:
The reaction of O
2
with cytochrome c oxidase to form
the oxygenated species A (Fig. 18-11) is very rapid,
occurring with apparent lifetime (Eq. 9-5) of ~ 8 10
s.
139
Study of such rapid reactions has depended
upon a flow-flash technique developed by Greenwood
and Gibson.
136,140,141
Fully reduced cytochrome oxidase
is allowed to react with carbon monoxide, which binds
to the iron in cytochrome a
3
just as does O
2
. In fact, it
was the spectroscopic observation that only half of the
(18-5)
4 e

+ 8 H
+
IN
+ O
2
2 H
2
O + 4 H
+
OUT
Ch 18IND page - E 2/14/03, 11:08 AM 1028
1029
Figure 18-8 Stereoscopic ribbon diagrams of the chicken
bc
1
complex (A) The native dimer. The molecular twofold
axis runs vertically between the two monomers. Quinones,
phospholipids, and detergent molecules are not shown for
clarity. The presumed membrane bilayer is represented by
a gray band. (B) Isolated close-up view of the two confor-
mations of the Rieske protein (top and long helix at right)
in contact with cytochrome b (below), with associated heme
groups and bound inhibitors, stigmatellin, and antimycin.
The isolated heme of cytochrome c
1
(left, above) is also shown.
(C) Structure of the intermembrane (external surface) domains
of the chicken bc
1
complex. This is viewed from within the
membrane, with the transmembrane helices truncated at
roughly the membrane surface. Ball-and-stick models repre-
sent the heme group of cytochrome c
1
, the Rieske ironsulfur
cluster, and the disulfide cysteines of subunit 8. SU, subunit;
cyt, cytochrome. From Zhang et al.
105
A
B
C
B. Electron Transport Chains
Ch 18IND page - E 2/14/03, 11:08 AM 1029
1030 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Figure 18-9 Proposed routes of
electron transfer in mitochondrial
complex III according to Peter
Mitchells Q cycle. Ubiquinone (Q)
is reduced to QH
2
by complex I (left
side of diagram) using two H
+
taken
up from the matrix (leaving nega-
tive charges on the inner membrane
surface). After diffusing across the
bilayer (dashed line) the QH
2
is
oxidized in the two steps with
release of the two protons per QH
2
on the positive (P) side of the membrane. In the two-step oxidation via anionic radical Q

one electron flows via the Rieske Fe


S protein and the cytochrome c
1
heme to external cytochrome c. The other electron is transferred to heme b
L
of cytochrome b,
then across the membrane to heme b
H
which now reduces Q to Q

. A second QH
2
is dehydrogenated in the same fashion and
the electron passed through the cytochrome b centers is used to reduce Q

to QH
2
with uptake from the matrix of 2 H
+
. The
resulting QH
2
diffuses back across the membrane to function again while the other Q diffuses back to complex I. The net
result is pumping of 4 H
+
per 2 e

passed through the complex. Notice that in the orientation used in this figure the matrix is
at the bottom, not the top as in Figs. 18-4 and 18-5.
Intermembrane space
Matrix
P (positive) side
+ + + + +
4H+
FeS c
1
c
2e

from
Complex I
QH
2
2QH
2
QH
2
Q
2e

2Q

b
L
b
H
Antimycin
Q

Q
1e

To
cytochrome c
oxidase
2Q
2H+ 2H+

N (negative) side

+ + + +

+
cytochrome a combined with CO that led Keilin to
designate the reactive component a
3
. This CO com-
plex is mixed with O
2
-containing buffer and irradiated
with a laser pulse to release the CO and allow O
2
to
react. The first rapid reaction observed is the binding
of O
2
(step c in Fig. 18-11). Formation of a peroxy
intermediate from the initial oxygenated form (A in
Fig. 18-11) is very fast. The O O bond of O
2
has already
been cleaved in form P (Fig. 18-11), which has until
recently been thought to be the peroxy intermediate.
In fact, spectroscopic measurements indicate that form
P contains an oxo-ferryl ion with the second oxygen of
the original O
2
converted to an OH ion and probably
coordinated with Cu
B
.
136a,136c,142,142a c
P may also con-
tain an organic radical, perhaps formed from tyrosine
244 as indicated in Fig. 18-11.
A second relaxation time of = 32 45 s has been
assigned
139
to the conversion of the peroxide interme-
diate P to P. A third relaxation time ( = 100 140 s)
is associated with the oxidation of Cu
A
by a (not shown
in Fig. 18-11).
143
This electron transfer step limits the
rate of step f of Fig. 18-11. Another reduction step
with ~ 1.2 ms is apparently associated with electron
transfer in step h. This slowest step still allows a first-
order reaction rate of ~ 800 s
1
.
When O
2
reacts with cytochrome c oxidase, it may
be bound initially to either the a
3
iron or to Cu
B
, but in
the peroxy intermediate P it may bind to both atoms.
Oxyferryl compound F (Fig. 8-11) as well as radical
species, can also be formed by treatment of the oxidized
Cu
S
Cu
S
S
CH
3
O
C
C216
N
N
M227
C220
N N
H224
H181
E218
N
H
H376
N
Fe
a
3
Cu
O
O
N
N
Y244
O
H240
N
N
H291
H290
B
H
The Cu
A
center of cytochrome oxidase
The Cu
B
A
3
center of cytochrome oxidase
Ch 18IND page - E 2/14/03, 11:08 AM 1030
1031
Figure 18-10 Structure of
mitochondrial cytochrome c
oxidase. (A) Stereoscopic C

backbone trace for one mono-


meric complex of the core sub-
units I, II, and III. (B) Stereo-
scopic view showing all 13 sub-
units. The complete complex is
a dimer of this structure. From
Tsukihara et al.
125
(C) MolScript
ribbon drawing of one mono-
meric unit. The horizontal lines
are drawn at distances of 1.0
and 2.0 nm from the center of
the membrane bilayer as estimat-
ed from eight phospholipid
molecules bound in the struc-
ture. From Wallin et al.
127
Courtesy of Arne Elofsson.
(D) Schematic drawing of the
same complex showing positions
of the Cu
A
dimetal center, bound
Mg
2+
, heme a, the bimetal heme
a
3
- Cu
B
center, and bound Zn
2+
.
The location of an 0.48-nm mem-
brane bilayer is marked. From
Tsukihara et al.
129
(A), (B), and (D)
courtesy of Shinya Yoshikawa.
A
B
C D
B. Electron Transport Chains
Ch 18IND page - E 2/14/03, 11:08 AM 1031
1032 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
enzyme O with hydrogen peroxide.
143a-144
Use of
various inhibitors has also been important in studying
this enzyme. Cyanide, azide, and sulfide ions, as well
as carbon monoxide, are powerful inhibitors. Cyanide
specifically binds to the Fe
3+
form of cytochrome a
3
preventing its reduction,
145
while CO competes with
O
2
for its binding site. A much-used reagent that
modifies carboxyl groups in proteins, and which
inhibits many proton translocating proteins, is dicyclo-
hexyl carbodiimide (Eq. 3-10).
146
The step-by-step
flow of electrons through cytochrome c oxidase seems
quite well defined. However, one of the most impor-
tant aspects is unclear. How is the pumping of pro-
tons across the membrane coupled to electron
transport?
137,138,142,147,147a
Many recent studies have
employed directed mutation of residues in all four
subunits to locate possible proton pathways or chan-
nels.
148 152
Most ideas involve movement through
Figure 18-11 Possible catalytic cycle of cyto-
chrome c oxidase at the cytochrome a
3
Cu
B
site. The fully oxidized enzyme (O; left center)
receives four electrons consecutively from the
cyt c Cu
A
cyt a chain. In steps a and b both
heme a and Cu
B
, as well as the Cu
A
center and
cyt a
3
, are reduced to give the fully reduced
enzyme (R). In the very fast step c the cyt a
3
heme becomes oxygenated and in step d is
converted to a peroxide with oxidation of both
the Fe and Cu. Intermediate P was formerly
thought to be a peroxide but is now thought to
contain ferryl iron and an organic radical. This
radical is reduced by the third electron in step f
to give the ferryl form F, with Cu
2+
participating
in the oxidation. The fourth electron reduces
Cu
B
again (step g) allowing reduction to the
hydroxy form H in step h. Protonation to form
H
2
O (step i) completes the cycle which utilizes
4 e

+ 4 H
+
+ O
2
to form 2 H
2
O. Not shown is
the additional pumping of 4 H
+
across the mem-
brane from the matrix to the intermembrane
space.
e

a
b
d
e
f
g
Fe (IV)
O
e

H
+
, e

Fe (III)
+
Fe (II)
O
O
Fe (II)
O
2
c
Fe (III)
O
O

O Oxidized
E One-electron reduced
R Two-electron reduced
(completely reduced)
A Oxygenated
P Peroxy
F Oxyferryl
H Hydroxy
Fe (III)
+
Fe (III) Fe (IV)
i
h
P
F H
O
E
R
Fe (IV)
O
810 s
H
2
O
e

H
+
H
+
O OH
HOY (Tyrosine 244)
Cu
B
+
A
Cu
B
2+
30 s,
100 s
HOY
Cu
B
+
HOY
HOY
Cu
B
+
Cu
B
2+
HOY
Cu
B
2+
HOY
Cu
B
+
H
+
H
2
O
HOY
Very fast

OY
H
O Y
Cu
B
+
OH
Cu
B
+
OH
hydrogen bonded chains (Eq. 9-94), which may include
the carboxylate groups of the bound hemes.
153
Con-
formational changes may be essential to the gating of
proton flow by electron transfers.
143
The surface of the matrix side of cytochrome
oxidase contains histidine and aspartate side chains
close together. It has been suggested that they form a
proton collecting antenna that contains groups basic
enough to extract protons from the buffered matrix
and guide them to a proton conduction pathway.
154
Calcium ions also affect proton flow.
153a,b
We will
return to this topic in Section C,3 (p. 1040).
C. Oxidative Phosphorylation
During the 1940s when it had become clear that
formation of ATP from ADP and inorganic phosphate
Net charge on Fe(II)a
3
is shown as zero
because the charge 2
+
shown by some
authors is balanced by 2

from nitrogen
atoms in heme ring.
Structure originally proposed for form P.
Movement of one electron to form tyrosinate
radical is indicated by single-headed arrows
(see p. 1030)
Ch 18IND page - E 2/14/03, 11:08 AM 1032
1033
was coupled to electron transport in mitochondria,
intensive efforts were made to discover the molecular
mechanisms. However, nature sometimes strongly
resists attempts to pry out her secrets, and the situation
which prevailed was aptly summarized by Ephraim
Racker: Anyone who is not confused about oxidative
phosphorylation just doesnt understand the situa-
tion.
155
The confusion is only now being resolved.
1. The Stoichiometry (P/O Ratio) and Sites of
Oxidative Phosphorylation
Synthesis of ATP in vitro by tissue homogenates
was demonstrated in 1937 by Kalckar, who has written
a historical account.
156
In 1941, Ochoa
157
obtained the
first reliable measurement of the P/O ratio, the number
of moles of ATP generated per atom of oxygen utilized in
respiration. The P/O ratio is also equal to the number
of moles of ATP formed for each pair of electrons
passing through an electron transport chain. Ochoa
established that for the oxidation of pyruvate to acetyl-
CoA and CO
2
, with two electrons passed down the
mitochondrial electron transport chain, the P/O ratio
was ~ 3. This value has since been confirmed many
times.
158 160
However, experimental difficulties in
measuring the P/O ratio are numerous.
161
Many
errors have been made, even in recent years, and some
investigators
162
have contended that this ratio is closer
to 2.5 than to 3. One method for measuring the P/O
ratio is based on the method of determining the amount
of ATP used that is described in the legend to Fig. 15-2.
The experimental observation of a P/O ratio of ~ 3
for oxidation of pyruvate and other substrates that
donate NADH to the electron transport chain led to
the concept that there are three sites for generation of
ATP. It was soon shown that the P/ O ratio was only
2 for oxidation of succinate. This suggested that one
of the sites (site I) is located between NADH and
ubiquinone and precedes the diffusion of QH
2
formed
in the succinate pathway to complex III.
In 1949, Lehninger used ascorbate plus tetramethyl-
phenylene-diamine (TMPD, Table 18-4) to introduce
electrons into the chain at cytochrome c. The sequence
ascorbate TMPD cytochrome c was shown to
occur nonenzymatically. Later, it became possible to
use cytochrome c as an electron donor directly. In
either case only one ATP was generated, as would be
anticipated if only site III were found to the right of
cytochrome c. Site I was further localized by Lardy,
who used hexacyanoferrate (III) (ferricyanide) as an
artificial oxidant to oxidize NADH in the presence of
antimycin a. Again a P/ O ratio of one was observed.
Finally, in 1955, Slater showed that passage of elec-
trons from succinate to cytochrome c also gave only
one ATP, the one generated at site II. The concept of
three sites of ATP formation became generally accepted.
C. Oxidative Phosphorylation
However, as we shall see, these sites are actually
proton-pumping sites, and there may be more than
three of them.
Respiratory control and uncoupling. With
proper care relatively undamaged mitochondria can
be isolated. Such mitochondria are said to be tightly
coupled. By this we mean that electrons cannot pass
through the electron transport chain without generation
of ATP. If the concentration of ADP or of P
i
becomes
too low, both phosphorylation and respiration cease.
This respiratory control by ADP and P
i
is a property
of undamaged mitochondria. It may seem surprising
that damaged mitochondria or submitochondrial
particles are often able to transfer electrons at a faster
rate than do undamaged mitochondria. However,
electron transfer in damaged mitochondria occurs
without synthesis of ATP and with no slowdown as
the ADP concentration drops. A related kind of
uncoupling of electron transport from ATP syn-
thesis is brought about by various lipophilic anions
called uncouplers, the best known of which is
2,4-dinitrophenol. Even before the phenomenon
of uncoupling was discovered, it had been known that
dinitrophenol substantially increased the respiration
rates of animals. The compound had even been used
(with some fatal results) in weight control pills. The
chemical basis of uncoupling will be considered in
Section D.
States of mitochondria and spectrophoto-
metric observation. Chance and Williams defined
five states of tightly coupled mitochondria
60,163
; of
these, states 3 and 4 are most often mentioned. If no
oxidizable substrate or ADP is added the mitochon-
dria have a very low rate of oxygen uptake and are in
state 1. If oxidizable substrate and ADP are added
rapid O
2
uptake is observed, the rate depending upon
the rate of flow of electrons through the electron trans-
port chain. This is state 3. As respiration occurs the
coupled phosphorylation converts ADP into ATP,
exhausting the ADP. Respiration slows to a very low
value and the mitochondria are in state 4. If the sub-
strate is present in excess, addition of more ADP will
return the mitochondria to state 3.
Chance and associates employed spectrophoto-
metry on intact mitochondria or submitochondrial
particles to investigate both the sequence of carriers
and the sites of phosphorylation. Using the dual
wavelength spectrophotometer, the light absorption
at the absorption maximum (
max
) of a particular
component was followed relative to the absorption at
some other reference wavelength (
ref
). The principal
wavelengths used are given in Table 18-6. From these
measurements the state of oxidation or reduction of
each one of the carriers could be observed in the
various states and in the presence of inhibitors. The
Ch 18IND page - E 2/14/03, 11:08 AM 1033
1034 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
experiments served to establish that electrons passing
down the chain do indeed reside for a certain length
of time on particular carriers. That is, in a given state
each carrier exists in a defined ratio of oxidized to
reduced forms ([ox] / [red]). Such a result would not
be seen if the entire chain functioned in a cooperative
manner with electrons passing from the beginning to
the end in a single reaction. By observing changes in
the ratio [ox] / [red] under different conditions, some
localization of the three phosphorylation sites could be
made. In one experiment antimycin a was added to
block the chain ahead of cytochrome c
1
. Then tightly
coupled mitochondria were allowed to go into state
4 by depletion of ADP. Since the concentration of
oxygen was high and cytochrome a
3
has a low K
m
for
O
2
(~ 3 M) cytochrome a
3
was in a highly oxidized
state. Cytochrome a was also observed to be oxidized,
while cytochrome c
1
and c remained reduced. The
presence of this crossover point suggested at the
time that cytochrome c might be at or near one of the
energy conservation sites. Accounts of more recent
experiments using the same approach are given by
Wilson et al.
164
2. Thermodynamics and Reverse Electron Flow
From Table 6-8 the value of G for oxidation of
one mole of NADH by oxygen (1 atm) is 219 kJ. At a
pressure of ~ 10
2
atm O
2
in tissues the value is 213 kJ.
However, when the reaction is coupled to the synthesis
of three molecules of ATP (G = +34.5 kJ mol
1
) the net
Gibbs energy change for the overall reaction becomes
G = 110 kJ mol
1
. This is still very negative. How-
ever, we must remember that the concentrations of
ATP, ADP, and P
i
can depart greatly from the 1:1:1
ratio implied by the G value.
An interesting experiment is to allow oxidative
phosphorylation to proceed until the mitochondria
reach state 4 and to measure the phosphorylation
state ratio R
p
, which equals the value of [ATP] /
[ADP][P
i
] that is attained. This mass action ratio,
which has also been called the phosphorylation ratio
or phosphorylation potential (see Chapter 6 and Eq.
6-29), often reaches values greater than 10
4
10
5
M
1
in
the cytosol.
164
An extrapolated value for a zero rate
of ATP hydrolysis of log R
p
= 6.9 was estimated. This
corresponds (Eq. 6-29) to an increase in group transfer
potential (G of hydrolysis of ATP) of 39 kJ/mol. It
follows that the overall value of G for oxidation of
NADH in the coupled electron transport chain is less
negative than is G. If synthesis of three molecules
of ATP is coupled to electron transport, the system
should reach an equilibrium when R
p
= 10
6.4
at 25C,
the difference in G and G being 3RT ln Rp = 3 x
5.708 x 6.4 = 110 kJ mol
1
. This value of Rp is, within
experimental error, the same as the maximum value
observed.
165
There apparently is an almost true equi-
librium among NADH, O
2,
and the adenylate system
if the P/O ratio is 3.
Within more restricted parts of the chain it is
possible to have reversed electron flow. Consider the
passage of electrons from NADH, partway through
the chain, and back out to fumarate, the oxidized form
of the succinatefumarate couple. The Gibbs energy
change G (pH 7) for oxidation of NADH by fumarate
is 67.7 kJ mol
1
. In uncoupled mitochondria electron
flow would always be from NADH to fumarate.
However, in tightly coupled mitochondria, in which
ATP is being generated at site I, the overall value of
G becomes much less negative. If R
p
= 10
4
M
1
, G
for the coupled process becomes approximately zero
( 67.7 + 68 kJ mol
1
). Electron flow can easily be
reversed so that succinate reduces NAD
+
. Such ATP-
driven reverse flow occurs under some physiological
conditions within mitochondria of living cells, and
some anaerobic bacteria generate all of their NADH
by reversed electron flow (see Section E).
Another experiment involving equilibration with
the electron transport chain is to measure the observed
potential of a carrier in the chain as a function of the
concentrations of ATP, ADP, and P
i
. The observed
TABLE 18-6
Wavelengths of Light Used to Measure States of
Oxidation of Carriers in the Electron Transport
Chain of Mitochondria
a
NADH 340 374
Flavins 465 510
Cytochromes
b
2+
564() 575
530()
430()
c
1
2+
534()
523()
418()
c
2+
550() 540
521()
416()
a
2+
605() 630(590)
450()
a
a
3
2+
600()
a
445() 455
Carrier
max
(nm)
b

ref
(nm)
a
After Chance, B. and Williams, G. R. (1955) J. Biol. Chem, 217, 409
427; (1956) Adv. Enzymol. 17, 409427.
b
The wavelengths used for each carrier in dual wavelength spec-
troscopy appear opposite each other in the two columns. Some
positions of other absorption bands of cytochromes are also given.
Ch 18IND page - E 2/14/03, 11:08 AM 1034
1035
potential E is obtained by measuring log([ox] / [red])
and applying Eq. 18-6 in which E is the known mid-
point potential of the couple (Table 6-8) and n is the
number of electrons required to reduce one molecule
of the carrier. If the system is equilibrated with a
C. Oxidative Phosphorylation
redox buffer (Chapter 6), E can be fixed at a pre-
selected value. For example, a 1:1 mixture of succinate
and fumarate would fix E at +0.03 V while the couple
3-hydroxybutyrate-acetoacetate in a 1:1 ratio would
fix it at E = 0.266 V. Consider the potential of cyto-
chrome b
562
(b
H
), which has an E value of 0.030 V.
Substituting this in Eq. 18-7 and using E = 0.266 V
(as obtained by equilibration with 3-hydroxybutyrate
acetoacetate), it is easy to calculate that at equilibrium
the ratio [ox] / [red] for cytochrome b
562
is about 10
5
.
In other words, in the absence of O
2
this cytochrome will be kept almost
completely in the reduced form in
an uncoupled mitochondrion.
However, if the electron trans-
port between 3-hydroxybutyrate
and cytochrome b
562
is tightly coupled
to the synthesis of one molecule of
ATP, the observed potential of the
carrier will be determined not only
by the imposed potential E
i
of the
equilibrating system but also by the
phosphorylation state ratio of the
adenylate system (Eq. 18-7). Here
G
ATP
is the group transfer poten-
tial (G of hydrolysis) of ATP at
pH 7 (Table 6-6), and n is the num-
ber of electrons passing through the
chain required to synthesize one
ATP. In the upper part of the equa-
tion n is the number of electrons
required to reduce the carrier, namely
one in the case of cytochrome b
562
.
From Eq. 18-7 it is clear that in
the presence of a high phosphoryla-
tion state ratio a significant fraction
of cytochrome b
562
may remain in
the reduced form at equilibrium.
Thus, if R
p
= 10
4
, if E for cyto-
chrome b
562
is 0.030 V, if n = 2, and
the potential E is fixed at 0.25 V
using the hydroxybutyrateaceto-
acetate couple, we calculate, from
Eq. 18-7, that the ratio [ox] / [red] for
cytochrome b
562
will be 1.75. Now, if
R
p
is varied the observed potential
of the carrier should change as pre-
dicted by Eq. 18-7. This variation
has been observed.
164
For a tenfold
change in R
p
the observed potential
of cytochrome b
562
changed by 0.030
V, just that predicted if n = 2. On
the other hand, the observed poten-
tial of cytochrome c varied by 0.059
V for every tenfold change in the
ratio. This is just as expected if n = 2,
and if synthesis of two molecules of
E
G
nF
E
n
=

= +
=
0 0592 .
log
[ ]
[ ]
ox
red
observed potential of carrier
(18-6)
TABLE 18-7
Electrode Potentials of Mitochondrial Electron Carriers and
Gibbs Energy Changes Associated with Passage of Electrons
a
Electron carrier
E (pH 7)
isolated
E (pH 7.2)
in mito-
chondria
G (kJ mol
-1
)
for 2 e

flow to
O
2
at 10
2
atm,
carriers at pH 7
NADH / NAD+ 0.320 213
Group I Flavoprotein ~ 0.30
~ 0.30 V FeS protein ~ 0.305
-Hydroxybutyrate
acetoacetate 0.266 203
Lactatepyruvate 0.185 187
Succinate-fumarate 0.031 146
Group II Flavoprotein ~ 0.045
~ 0 V Cytochrome b
T
0.030
Cu 0.001
FeS protein 0.030
Cytochrome b
K
0.030
Ubiquinone 0.10 0.045 132
Cytochrome a
3
+ ATP 0.155
Group III Cytochrome c
1
0.215
Cytochrome c 0.254 0.235 102
Cytochrome b
T
+ ATP 0.245
Cytochrome a 0.29 0.210
Cu 0.245
FeS protein 0.28
Group IV Cytochrome a
3
0.385 77
O
2
(10
2
atm) 0.785 0.00
1 atm 0.815
a
Data from Wilson, D. F., Dutton, P. L., Erecinska, M., Lindsay, J. G., and Soto, N. (1972)
Acc. Chem. Res. 5, 234241 and Wilson, D. F., Erecinska, M., and Sutoon, P. L. (1974)
Ann. Rev. Biophys. Bioeng. 3, 203230.
Ch 18IND page - E 2/14/03, 11:08 AM 1035
1036 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
ATP is coupled to the electron transport to cytochrome
c. Thus, we have experimental evidence that when
one-electron carriers such as the cytochromes are
involved, the passage of two electrons is required to
synthesize one molecule of ATP. Furthermore, from
experiments of this type it was concluded that the sites
of phosphorylation were localized in or related to
complexes I, III, and IV.
Another kind of experiment is to equilibrate the
electron transport chain with an external redox pair
of known potential using uncoupled mitochondria.
The value of E of a particular carrier can then be
measured by observation of the ratio [ox] / [red] and
applying Eq. 18-7. While changes in the equilibrating
potential E will be reflected by changes in [ox] / [red]
the value of E will remain constant. The E values
of Fe S proteins and copper atoms in the electron
transport chain have been obtained by equilibrating
mitochondria, then rapidly freezing them in liquid
nitrogen, and observing the ratios [ox] / [red] by EPR
at 77K (Table 18-7).
The values of E of the mitochondrial carriers fall
into four isopotential groups at ~ 0.30, ~ 0, ~ + 0.22,
and ~ + 0.39 V (Table 18-7). When tightly coupled
mitochondria are allowed to go into state 4 (low ADP,
high ATP, O
2
present but low respiration rate), the
observed potentials change. That of the lowest iso-
potential group (which includes NAD
+
/ NADH) falls
to ~ 0.38 V, corresponding to a high state of reduction
of the carriers to the left of the first phosphorylation
site in Fig. 18-4. Groups 2 and 3 remain close to their
midpoint potentials at ~ 0.05 and + 0.26 V. In this
condition the potential difference between each suc-
cessive group of carriers amounts to ~ 0.32 V, just
enough to balance the formation of one molecule of
ATP for each two electrons passed at a ratio R
p
10
4
M
1
(Eq. 18-7).
Two cytochromes show exceptional behavior and
appear twice in Table 18-7. The midpoint potential E
of cytochrome b
566
(b
L
) changes from 0.030 V in the
absence of ATP to + 0.245 V in the presence of a high
concentration of ATP. On the other hand, E for cyto-
chrome a
3
drops from + 0.385 to 0.155 V in the presence
of ATP. These shifts in potential must be related to the
coupling of electron transport to phosphorylation.
3. The Mechanism of Oxidative
Phosphorylation
It was natural to compare mitochondrial ATP
synthesis with substrate-level phosphorylations, in
which high-energy intermediates are generated by
the passage of electrons through the substrates. The best
known example is oxidation of the aldehyde group
of glyceraldehyde 3-phosphate to an acyl phosphate,
which, after transfer of the phospho group to ADP,
becomes a carboxylate group (Fig. 15-6). The Gibbs
energy of oxidation of the aldehyde to the carboxylate
group provides the energy for the synthesis of ATP.
However, this reaction differs from mitochondrial
electron transport in that the product, 3-phosphoglycerate,
is not reconverted to glyceraldehyde 3-phosphate. Electron
carriers of the respiratory chain must be regenerated in
some cyclic process. Because of this, it was difficult to
imagine practical mechanisms for oxidative phospho-
rylation that could be related to those of substrate
level phosphorylation. Nevertheless, many efforts
were made over a period of several decades to find
such high-energy intermediates.
Search for chemical intermediates. An early
hypothetical model, proposed by Lipmann,
166
is
shown in Fig. 18-12. Here A, B, and C are three electron
carriers in the electron transport chain. Carrier C is a
better oxidizing agent than B or A. Carrier B has some
special chemistry that permits it, in the reduced state,
to react with group Y of a protein (step b) to form Y BH
2
.
The latter, an unidentified adduct, is converted by
oxidation with carrier C (step c) to a high energy
oxidized form indicated as Y ~ B. Once the possibility
of generating such an intermediate is conceded, it is
easy to imagine plausible ways in which the energy of
this intermediate could be transferred into forms with
which we are already familiar. For example, another
protein X could react (step d) to form X ~ Y in which
the X ~ Y linkage could be a thioester, an acyl phos-
phate, or other high-energy form. Furthermore, it
might not be necessary to have two proteins; X and
Y could be different functional groups of the same
protein. They might be nonprotein components, e.g.,
Y might be a phospholipid.
Generation of ATP by the remaining reactions
(steps e and f of Fig. 18-12) is straightforward. For
example, if X ~ Y were a thioester the reactions would
be the reverse of Eq. 12-48. These reaction steps would
also be responsible for observed exchange reactions,
for example, the mitochondrially catalyzed exchange
of inorganic phosphate (H
32
PO
4
2
) into the terminal
position of ATP. Mitochondria and submitochondrial
particles also contain ATP-hydrolyzing (ATPase)
activity, which is thought to depend upon the same
machinery that synthesizes ATP in tightly coupled
mitochondria. In the scheme of Fig. 18-12, ATPase
E E
n
E
G
n
RT
n F
E
n n
R
i
i
( )
.
log
[ ]
[ ]
.
ln
[ ]
[ ][ ]
. .
log
observed
ox
red
ATP
ADP P
ATP
i
P
= +
= +

= +

0 0592
96 5
0 358 0 0592
10
10

(18-7)
Ch 18IND page - E 2/14/03, 11:08 AM 1036
1037
activity would be observed if hydro-
lysis of X ~ Y were to occur. Partial
disruption of the system would lead
to increased ATPase reactivity, as is
observed. Uncouplers such as the
dinitrophenolate ion or arsenate ion,
acting as nucleophilic displacing
groups, could substitute for a group
such as X. Spontaneous breakdown
of labile intermediates would permit
oxidation to proceed unimpaired.
Since there are three different sites
of phosphorylation, we might expect
to have three different enzymes of
the type Y in the scheme of Fig. 18-12,
but it would be necessary to have
only one X.
In Lipmanns original scheme
group Y was visualized as adding
to a carboncarbon double bond
to initiate the sequence. Isotopic
exchange reactions ruled out the
possibility that either ADP or P
i
might serve as Y, but it was attrac-
tive to think that a bound phosphate
ion, e.g., in a phospholipid or coen-
zyme, could be involved. Y ~ B of
Fig. 18-12 would be similar in reac-
tivity to an acyl phosphate or thio-
ester. However, whatever the nature
of Y ~ B, part of group Y would be
left attached to B after the transfer of
Y to X. For example, if Y were YOH
compound X ~ OY would be formed, and the carrier
would be left in step d in the form of BOH. Elimination
of a hydroxyl group would be required to regenerate B.
Perhaps nature has shunned this mechanism because
there is no easy way to accomplish such an elimination.
Many variations on the scheme of Fig. 18-12 were
proposed,
166
and some were discussed in the first
edition of this textbook.
167
However, as attractive as
these ideas may have seemed, all attempts to identify
discrete intermediates that might represent X ~ Y failed.
Furthermore, most claims to have seen Y ~ B by any means
have been disproved.
Peter Mitchells chemiosmotic theory. To
account for the inability to identify high energy inter-
mediates as well as the apparent necessity for an intact
membrane, Peter Mitchell, in 1961, offered his chemi-
osmotic theory of oxidative phosphorylation.
168 175a
aThis theory also accounts for the existence of energy-
linked processes such as the accumulation of cations
by mitochondria. The principal features of the Mitchell
theory are illustrated in Fig. 18-13. Mitchell proposed
Figure 18-12 An early proposal for formation of ATP via
high-energy chemical intermediates.
a
b
c
d
e
f
A
AH
2
BH
2
B Y ~ B
Y BH
2
C
CH
2
Spontaneous hydro-
lysis of intermediate
X ~ Y would cause
the ATPase reaction
X ~ Y Uncouplers could
substitute for X in
this reaction
Reactions causing P
i
ATP
and other exchanges and
ATPase reactions
Hypothetical coupling
site for ATP synthesis
ADP
ATP
X
X ~ P
Y
P
i
Y
X
C. Oxidative Phosphorylation
Figure 18-13 Principal features of Mitchells chemiosmotic theory of oxida-
tive phosphorylation.
Membrane
BH
2
B
A
red
A
ox
C
red
C
ox
Oriented ATP
synthase couples
synthesis of ATP
to flow of protons
back through
the membrane
Membrane is otherwise
impermeable to protons
Proton pump operated by
electron flow is arranged to
pick up protons from the inside
and to deposit them on the
outside of the membrane
Removal of protons
lowers [H
+
] of matrix space
Expelled protons
raise [H
+
] in
intermembrane space
2 H
+
Accumulating
positive and negative
charges on membrane
surface induce
membrane potential
2 H
+
H
+
H
+
H
+
H
+
H
+
X

ADP + P
i
2 H
+
ATP
2 H
+
Counterion X

may flow out to


balance positive
charge of protons
pumped permitting
accumulation of
H
+
X

in bulk of
intermembrane
fluid
X

Or cations such as Ca
2+
may diffuse inward to
preserve electrical neutrality
when protons are expelled
Ca
2+
Intermembrane
space (outside)
Matrix
(inside)
ATP
synthase
Ch 18IND page - E 2/14/03, 11:08 AM 1037
1038 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
that the inner membrane of the mitochondrion is a
closed, proton-impermeable coupling membrane,
which contains proton pumps operated by electron
flow and which cause protons to be expelled through
the membrane from the matrix space. As indicated
in Fig. 18-13, an oxidized carrier B, upon reduction
to BH
2
, acquires two protons. These protons do not
necessarily come from reduced carrier AH
2
, and
Mitchell proposed that they are picked up from the
solvent on the matrix side of the membrane. Then, when
BH
2
is reoxidized by carrier C, protons are released on
the outside of the membrane. On the basis of existing
data, Mitchell assumed a stoichiometry of two protons
expelled for each ATP synthesized. It followed that there
should be three different proton pumps in the electron
transport chain corresponding to the three phosphory-
lation sites.
The postulated proton pumps would lead either to
bulk accumulation of protons in the intermembrane
space and cytoplasm, with a corresponding drop in
pH, or to an accumulation of protons along the membrane
itself. The latter would be expected if counterions X

do not pass through the membrane with the protons.


The result in such a case would be the development of
a membrane potential, a phenomenon already well
documented for nerve membranes (Chapter 8).
A fundamental postulate of the chemiosmotic
theory is the presence of an oriented ATP synthase
that utilizes the Gibbs energy difference of the proton
gradient to drive the synthesis of ATP (Fig. 18-9).
Since G (pH 7) for ATP synthesis is +34.5 kJ mol
1
and, if as was assumed by Mitchell, the passage of two
protons through the ATP synthase is required to form
one ATP, the necessary pH gradient (given by Eq. 6-25
or Eq. 18-9 with E
m
= 0) would be 34.5/(2 x 5.708) = 3.0
pH units at 25C. On the other hand, if the phospho-
rylation state ratio is ~ 10
4
M
1
, the pH difference
would have to be 5 units. Most investigators now
think that 4 H
+
per ATP are needed by the synthase.
If so, a pH difference of 2.5 units would be adequate.
Various experiments have shown that passage of
electrons does induce a pH difference, and that an
artifically induced pH difference across mitochondrial
membranes leads to ATP synthesis. However, pH
gradients of the required size have not been observed.
Nevertheless, if the membrane were charged as indi-
cated in Fig. 18-13, without accumulation of protons
in the bulk medium, a membrane potential would be
developed, and this could drive the ATP synthase, just
as would a proton gradient.
The mitochondrial membrane potential E
m
(or )
is the potential difference measured across a mem-
brane relative to a reference electrode present in the
surrounding solution.
176
For both mitochondria and
bacteria E
m
normally has a negative value. The Gibbs
energy change
H
+ for transfer of one mole of H
+
from
the inside of the mitochondrion to the outside, against
the concentration and potential gradients, is given by
Eq. 18-8. This equation follows directly from Eqs. 6-25
G
H
+ = 2.303 RT pH E
m
F
= 5.708 pH 96.5 E
m
kJ/ mol at 25C
where pH = pH (inside) pH (outside)
and 6-63 with n = 1. The same information is conveyed
in Eq. 18-9, which was proposed by Mitchell for what
he calls the total protonic potential difference p.
Mitchell was struck by the parallel between the force
and flow of electrons, which we call electricity, and
the force and flow of protons, which he named
proticity.
174
This led one headline writer in Nature
177
to describe Mitchell as a man driven by proticity,
but if Mitchell is right, as seems to be the case, we
are all driven by proticity! Mitchell also talked about
protonmotive processes and referred to p as the
protonmotive force. Although it is a potential rather
than a force, this latter name is a popular designation
for p.
The reader should be aware that considerable con-
fusion exists with respect to names and definitions.
176
For example, the G
H
+ of Eq. 18-8 can also be called
the proton electrochemical potential
H
+, which
is analogous to the chemical potential of an ion (Eq.
6-24) and has units of kJ/mol (Eq. 18-10).
G
H
+=
H
+ = F p
= 96.5 p kJ/ mol at 25C
However, many authors use
H
+ as identical to the
protonmotive force p.
From Eq. 18-9 or Eq. 18-10 it can be seen that a
membrane potential E
m
of 296 mV at 25C would be
equivalent to a 5.0 unit change in pH and would be
sufficient, if coupled to ATP synthesis via 2 H
+
, to raise
R
p
to 10
4
M
1
. Any combination of pH and E
m
pro-
viding p of 296 mV would also suffice. If the ratio
H
+
/ATP = 4, p of 148 mV would suffice.
The chemiosmotic hypothesis had the great virtue
of predicting the following consequences which could
be tested: (1) electron-transport driven proton pumps
with defined stoichiometries and (2) a separate ATP
synthase, which could be driven by a pH gradient
or membrane potential. Mitchells hypothesis was
initially greeted with skepticism but it encouraged
many people, including Mitchell and his associate
Jennifer Moyle, to test these predictions, which were
soon found to be correct.
178
(18-9)
(18-8)
(18-10)

p E
RT
F
p E
E
m
(volts) = (volts) 2.303 pH
(mV) = (mV) 59.2 pH at 25 C
=
m
m

Ch 18IND page - E 2/14/03, 11:08 AM 1038


1039
Observed values of E
m
and pH. One of the
problems
179
in testing Mitchells ideas has been the dif-
ficulty of reliably measuring p. To evaluate the pH
term in Eq. 18-10 measurements have been made with
microelectrodes and indicator dyes. However, the
most reliable approach has been to observe the distri-
bution of weak acids and bases across the mitochon-
drial membrane.
180
This is usually done with a
suspension of freshly isolated active mitochondria.
The method has been applied widely using, for example,
methylamine. A newer method employs an isotope
exchange procedure to measure the pH-sensitive
carbonic anhydrase activity naturally present in mito-
chondria.
181
The measurement of E
m
() is also difficult.
179
Three methods have been used: (1) measurement with
microelectrodes; (2) observation of fluorescent probes;
(3) distribution of permeant ions. Microelectrodes
inserted into mitochondria
182
have failed to detect a
significant value for E
m
. Fluorescent probes are not
very reliable,
179,183
leaving the distribution of permeant
ions the method of choice. In this method a mitochon-
drial suspension is exposed to an ion that can cross
the membrane but which is not pumped or subject
to other influences that would affect its distribution.
Under such conditions the ion will be distributed
according to Eq. 18-11. The most commonly used ions
are K
+
, the same ion that is thought to reflect the mem-
brane potential of nerve axons (Chapter 30), or Rb
+
. To
make the inner mitochondrial membrane permeable to
K
+
, valinomycin (Fig. 8-22) is added. The membrane
potential, with n = 1 in Eq. 9-1, becomes:
E
m
= 59.2 ([K
+
]
inside
/ [K
+
]
outside
) volts
In these experiments respiring mitochondria are
observed to take up the K
+
or Rb
+
to give a high ratio
of K
+
inside to that outside and consequently a nega-
tive E
m
. There are problems inherent in the method.
The introduction of a high concentration of ion perturbs
the membrane potential, and there are uncertainties
concerning the contribution of the Donnan equilibrium
(Eq. 8-5) to the observed ion distribution.
184
In most instances, either for mitochondrial suspen-
sions or whole bacteria, pH is less negative than 0.5
unit making a contribution of, at most, 30 mV to p.
The exception is found in the thylakoid membranes of
chloroplasts (Chapter 23) in which protons are pumped
into the thylakoid vesicles and in which the internal
pH falls dramatically upon illumination of the chloro-
plasts.
185
The pH reaches a value of 3.0 or more
units and p is ~180 mV, while E
m
remains ~0. Reported
values of E
m
for mitochondria and bacteria range from
100 to 168 mV and p from 140 to 230 mV.
172,179
Wilson concluded that E
m
for actively respiring mito-
chondria, using malate or glutamate as substrates,
attains maximum (negative) values of E
m
= 130 mV and
p = 160 mV.
179
However, Tedeschi and associates
183,184
argued that E
m
is nearly zero for liver mitochondria
and seldom becomes more negative than 60 mV for
any mitochondria.
A crucially important finding is that submitochon-
drial particles or vesicles from broken chloroplasts will
synthesize ATP from ADP and P
i
, when an artificial
pH gradient is imposed.
172,186
Isolated purified F
1
F
0
ATPase from a thermophilic Bacillus has been co-
reconstituted into liposomes with the light-driven
proton pump bacteriorhodopsin (Chapter 23). Illumi-
nation induced ATP synthesis.
187
These observations
support Mitchells proposal that the ATP synthase is
both spatially separate from the electron carriers in
the membrane and utilizes the protonmotive force
to make ATP. Thus, the passage of protons from the
outside of the mitochondria back in through the ATP
synthase induces the formation of ATP. What is the
stoichiometry of this process?
It is very difficult to measure the flux of protons
across the membrane either out of the mitochondria
into the cytoplasm or from the cytoplasm through the
ATP synthase into the mitochondria. Therefore, esti-
mates of the stoichiometry have often been indirect.
One argument is based on thermodynamics. If p
attains values no more negative than 160 mV and R
p
within mitochondria reaches at least 10
4
M
1
, we must
couple G
H
of 15.4 kJ/ mol to G of formation of ATP
of +57.3 kJ/ mol. To do this four H
+
must be translo-
cated per ATP formed. Recent experimental measure-
ments with chloroplast ATP synthase
188
also favor four
H
+
. It is often proposed that one of these protons is
used to pump ADP into the mitochondria via the
ATP-ADP exchange carrier (Section D). Furthermore,
if R
p
reaches 10
6
M
1
in the cytoplasm, it must exceed
10
4
M
1
in the mitochondrial matrix.
Proton pumps driven by electron transport.
What is the nature of the proton-translocating pumps
that link p with electron transport? In his earliest
proposals Mitchell suggested that electron carriers,
such as flavins and ubiquinones, each of which accepts
two protons as well as two electrons upon reduction,
could serve as the proton carriers. Each pump would
consist of a pair of oxidoreductases. One, on the inside
(matrix side) of the coupling membrane, would deliver
two electrons (but no protons) to the carrier (B in Fig.
18-13). The two protons needed for the reduction
would be taken from the solvent in the matrix. The
second oxidoreductase would be located on the outside
of the membrane and would accept two electrons from
the reduced carrier (BH
2
in Fig. 18-13) leaving the two
released protons on the outside of the membrane. To
complete a loop that would allow the next carrier to
be reduced, electrons would have to be transferred
through fixed electron carriers embedded in the
C. Oxidative Phosphorylation
(18-11)
Ch 18IND page - E 2/14/03, 11:08 AM 1039
1040 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
membrane from the reduced electron acceptor (C
red
in
Fig. 18-13) to the oxidized form of the oxidoreductase
to be used as reductant for the next loop. These loops,
located in complexes I and III of Fig. 18-5, would
pump three protons per electron or six H
+
/ O. With
a P/ O ratio of three this would provide two H
+
per
ATP formed. Mitchell regarded this stoichiometry as
appropriate.
The flavin of NAD dehydrogenase was an obvious
candidate for a carrier, as was ubiquinone. However,
the third loop presented a problem. Mitchells solution
was the previously discussed Q cycle, which is shown
in Fig. 18-9. This accomplishes the pumping in complex
III of 2 H
+
/e

, the equivalent of two loops.


111
However,
as we have seen, the magnitude of p suggests that 4
H
+
, rather than 2 H
+
, may be coupled to synthesis of
one ATP. If this is true, mitochondria must pump 12
H
+
/ O rather than six when dehydrogenating NADH,
or eight H
+
/ O when dehydrogenating succinate.
The stoichiometry of proton pumping was measured
by Lehninger and associates using a fast-responding
O
2
electrode and a glass pH electrode.
189,190
They
observed an export of eight H
+
/ O for oxidation of
succinate rat liver mitochondria in the presence of a
permeant cation that would prevent the buildup of
E
m
, and four H
+
/ O (2 H
+
/e
-
) for the cytochrome oxi-
dase system. These are equivalent to two H
+
/e

at
each of sites II and III as is indicated in Fig. 18-4.
Some others have found lower H
+
/e

ratios.
If two H
+
/e

are pumped out of mitochondria,


where do we find the pumping sites? One possibility
is that protons are pumped through the membrane by
a membrane Bohr effect, so named for its similarity to
the Bohr effect observed upon oxygenation of hemo-
globin. In the latter case (Chapter 7), the pK
a
values
of certain imidazole and terminal amino groups are
decreased when O
2
binds. This may result, in part,
from an electrostatic effect of O
2
in inducing a partial
positive charge in the heme. This partial charge may
then cause a decrease in the pK
a
values of nearby
groups. Similarly, complete loss of an electronic charge
from a heme group or an ironsulfur protein in the
electron transport chain would leave a positive charge,
an electron hole, which could induce a large change
in the pK
a
of a neighboring group. One manifestation
of this phenomenon may be a strong pH dependence
of the reduction potential (Eq. 16-19).
Protons that could logically be involved in a mem-
brane Bohr effect are those present on imidazole rings
coordinated to Fe or Cu in redox proteins. Removal of
an electron from the metal ion could be accompanied
by displacement of electrons within the imidazole,
within a peptide group that is hydrogen-bonded to an
imidazole, or within some other acidic group. A hypo-
thetical example is illustrated in Eq. 18-12 in which a
carboxyl group loses a proton when handed a second.
If the transiently enolized peptide linkage formed in
this process is tautomerized back to its original state
before the iron is reduced again, the proton originally
present on the carboxyl group will be released. It is
easy to imagine that a proton could then be ferried
in (as in Eq. 9-96) from the opposite side of the mem-
brane to reprotonate the imidazole group and complete
the pumping process.
In view of the large number of metal-containing
electron carriers in the mitochondrial chain, there are
many possible locations for proton pumps. However,
the presence of the three isopotential groups of Table
18-7 suggests that the pumps are clustered in complexes
I-III as pictured in Fig. 18-5. One site of pumping is
known to be in the cytochrome c oxidase complex.
When reconstituted into phospholipid, the purified
complex does pump protons in response to electron
transport, H
+
/e

ratios of ~1 being observed.


136,137,147,191
As mentioned in Section B,3 a large amount of experi-
mental effort has been devoted to identifying proton
transport pathways in cytochrome c oxidase and also
in the cytochrome bc
1
(complex II).
192
Proton pumping
appears to be coupled to chemical changes occurring
between intermediates P and F of Fig. 18-11, between
F and O,
136,193
and possibly between O and R.
137,138
Mechanisms involving direct coupling of chemical
changes at the A
3
Cu
B
center and at the Cu
A
dimetal
center have been proposed.
147,194
How do protons move from the pumping sites to
ATP synthase molecules? Since protons, as H
3
O
+
, are
sufficiently mobile, ordinary diffusion may suffice.
Because of the membrane potential they will tend to
stay close to the membrane surface, perhaps being
transported on phosphatidylethanolamine head
groups (see Chapter 8). According to the view of
R. J. P. Williams protons are not translocated across
the entire membrane by the proton pumps, but flow
through the proteins of the membrane to the ATP
synthase.
195
There the protons induce the necessary
conformational changes to cause ATP synthesis. A
related idea is that transient high-energy intermediates
N N Fe H O C
N H O
C
O
H
N N Fe H O C
N H O
C
O
e
Peptide
Enolized peptide
H
+
(released)
(18-12)
Ch 18IND page - E 2/14/03, 11:09 AM 1040
1041
generated by electron transport within membranes are
proton-carrying conformational isomers. When an
electron is removed from an electron-transporting
metalloprotein, the resulting positively charged hole
could be stable for some short time, while the protein
diffused within the membrane until it encountered an
F
0
protein of an ATP synthase. Then it might undergo
an induced conformational change at the same time
that it handed the Bohr effect proton of Eq. 18-14
to the F
0
protein and simultaneously induced a con-
formational change in that protein. The coupling of
proton transport to conformational changes seems
plausible, when we recall that the induction of confor-
mational changes within proteins almost certainly
involves rearrangement of hydrogen bonds.
A consequence of the chemiosmotic theory is that
there is no need for an integral stoichiometry between
protons pumped and ATP formed or for an integral
P/ O ratio. There are bound to be inefficiencies in
coupling, and p is also used in ways other than syn-
thesis of ATP.
4. ATP Synthase
In 1960, Racker and associates
196,197
discovered
that the knobs or little mushrooms visible in
negatively stained mitochondrial fragments or fragments
of bacterial membranes possess ATP-hydrolyzing
(ATPase) activity. Earlier the knob protein had been
recognized as one of several coupling factors required
for reconstitution of oxidative phosphorylation by
submitochondrial particles.
197
Electron micrographs
showed that the submitochondrial particles consist of
closed vesicles derived from the mitochondrial cristae,
and that the knobs (Fig. 18-14A) are on the outside of
the vesicles. They can be shaken loose by ultrasonic
oscillation with loss of phosphorylation and can be
added back with restoration of phosphorylation. The
knob protein became known as coupling factor F
1
.
Similar knobs present on the outside of the thylakoids
became CF
1
and those inside thermophilic bacteria
TF
1
. The ATPase activity of F
1
was a clue that the
knobs were really ATP synthase. It also became clear
that a portion of the ATP synthase is firmly embedded
in the membranes. This part became known as F
0
.
Both the names F
1
F
0
ATP synthase and F
1
F
0
ATPase
are applied to the complex, the two names describing
different catalytic activities. The ATPase activity is
usually not coupled to proton pumping but is a
readily measurable property of the F
1
portion. In a
well-coupled submitochondrial particle the ATPase
activity will be coupled to proton transport and will
represent a reversal of the ATP synthase activity.
The synthase structure. The F
1
complex has been
isolated from E. coli,
204,205
other bacteria,
206,207
yeast,
208a,b
animal tissues,
199,209211
and chloroplasts.
212214
In
every case it consists of five kinds of subunits with the
stoichiometry
3

3
.
214a,b
The F
0
complex of E. coli
contains three subunits designated a,b, and c. All of
these proteins are encoded in one gene cluster, the unc
operon (named for uncoupled mutants), with the
following order:
Here I is the regulatory gene (as in Fig. 28-1). The
E. coli F
0
appears to have approximately the unusual
stoichiometry ab
2
c
9-11
. This suggested the possibility
that 12 c subunits form a ring with D
6
or D
12
symmetry,
the latter being illustrated in the structural proposal
shown in Fig. 18-14E. However, crystallographic
evidence suggests that there may be 10, not 12 sub-
units.
214c
Mitochondrial ATP synthase of yeast contains at
least 13 different kinds of subunits
208
and that of ani-
mals
215
16, twice as many as in E. coli. Subunits , , ,
a, b, and c of the mitochondrial synthase correspond
to those of E. coli. However, the mitochondrial homo-
log of E. coli is called the oligomycin-sensitivity-
conferring protein (OSCP).
216 218
It makes the ATPase
activity sensitive to oligomycin. The mitochondrial
subunit corresponds to of E. coli or of chloroplasts.
217,219
Mitochondrial has no counterpart in bacteria.
209,220
In addition,
209,215
mitochondria contain subunits called
d, e, f, g, A6L, F6, and IF
1,
the last being an 84-residue
inhibitor, a regulatory subunit.
221
The subunits of
yeast ATP synthase correspond to those of the animal
mitochondrial synthase but include one additional
protein (h).
208a
Six of the relatively large (5057 kDa) and
subunits associate to an
3

3
complex that constitutes
the knobs.
202,210
Chemical crosslinking, directed
mutation, electron cryomicroscopy,
222,222a
and high-
resolution X-ray diffraction measurements
199,207,211,223,224
have established that the and subunits alternate in
a quasisymmetric cyclic head that contains active sites
for ATP formation in the three subunits (Fig. 18-14CE).
The subunits also contain ATP-binding sites, but
they are catalytically inactive, and their bound MgATP
does not exchange readily with external ATP and can
be replaced by the nonhydrolyzable AMP-PNP (Fig.
12-31) with retention of activity. The
3

3
complex is
associated with the F
0
part by a slender central stalk
C. Oxidative Phosphorylation
I B E F H A G D C Gene symbol
i a c b Subunit symbol
F
0
E. coli a c b
Mitochondria OSCP IF
1
F
1
a c b d f e g A6L F6
Ch 18IND page - E 2/14/03, 11:09 AM 1041
1042 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Figure 18-14 ATP synthase and vacuolar ATPase. (A)
Knobs of ATP synthase on mitochondrial membranes
negatively stained with phosphotungstate. (B) Vacuolar
proton-pumping ATPase from an intact vacuolar membrane
stained in the same way. Some images have been marked to
indicate well-resolved head groups (H), stalks (S), and basal
components (B). (A) and (B) are from Dschida and Bowman.
198
Courtesy of Barry J. Bowman. (C) Ribbon model of the atomic
structure of the F
1
part of bovine heart mitochondrial ATP
synthase. This section through the knob, which is drawn
schematially at the upper right, shows one subunit, con-
taining bound ADP,
DP
, to the left and one empty subunit,

E
, to the right. In the center are the N and C termini of
subunit . The arrow points to a disruption in the sheet of
the subunit structure in the
E
conformation. The asterisk
marks a loop that would collide with the C-terminal part of
subunit if the latter were rotated. (D) View of the F
1
ATP
synthase from the membrane side. The section shown
contains the nucleotide binding sites. Subunits with empty
sites are labeled
E
and
E
. Those with bound ADP are
labeled
DP
and
DP
, and those with bound ATP
TP
and
TP
.
Deviation from perfect threefold symmetry can be seen in
this view. (C) and (D) are from Abrahams et al.
199
Courtesy
of John E. Walker. (E) A recent model of the E. coli ATP
synthase. The
3

3
head contains the ATP-synthesizing sites
in the subunits. The
3

3
complex and also subunits a, b,
and remain stationary and form the stator for a protic engine,
whose rotor consists of 12 c subunits and attached and
subunits. Rotation is induced by the membrane potential or
difference in H
+
activity on the two sides of the membrane.
The carboxylate group of Asp 61 in each c subunit must be
protonated to COOH in order for it to move away from the
entrance channel in subunit a. The presence of the positively
charged Arg 210 near the exit channel in subunit a induces
release of the proton when the c subunit has rotated almost
360. According to this picture, 12 protons are required for
one rotation with synthesis of three molecules of ATP. After
Elston et al.
200
and Zhou et al.
201
See also Junge et al.
202
and
Engelbrecht and Junge.
203
10 nm
ATP
ADP + P
i
b
H
+
F
1
F
0
a
c
Membrane

R210
+
+ +
H
+
+ + +


Protonated on all
but one or two
subunits
C
O
OH
D61
A
B
C
D
E
50 nm
C
N
Ch 18IND page - E 2/14/03, 11:09 AM 1042
1043
or shaft. Much effort has gone into establishing the
subunit composition of the shaft and the F
0
parts of the
structure. As is indicated in Fig. 18-14E, subunits
and of the E. coli enzyme are both part of the central
shaft.
219,225,226
The same is true for the mitochondrial
complex, in which the subunit corresponds to bac-
terial .
227
The role of this subunit is uncertain. It is
part of the shaft but is able to undergo conformational
alterations that can permit its C-terminal portion to
interact either with F
0
or with the
3

3
head.
227,227a
The
unique subunit of mitochondrial ATPase appears also
to be part of the shaft.
220
The most prominent component of the central
shaft is the 270-residue subunit , which associates
loosely with the
3

3
head complex but more tightly
with F
0
. About 40 residues at the N terminus and 60
at the C terminus form an -helical coiled coil, which
is visible in Fig. 18-14E
199,211
and which protrudes into
the central cavity of the
3

3
complex. Because it is
asymmetric, the subunit apparently acts as a rotating
camshaft to physically alter the and subunits in a
cyclic manner. Asymmetries are visible in Fig. 18-14D.
211
The central part of subunit forms a more globular
structure, which bonds with the c subunits of F
0
.
205
Exact structures are not yet clear.
The subunit of E. coli ATP synthase (OSCP of
mitochondria) was long regarded as part of the central
stalk. However, more recent results indicate that it is
found in a second stalk, which joins the
3

3
complex
to F
0
. The central stalk rotates, relative to the second
stalk. The second stalk may be regarded as stationary
and part of a stator for a protic engine.
228,229
This
stalk has been identified
230
in electron micrographs
of chloroplast F
1
F
0
and by crosslinking studies. As is
depicted in Fig. 18-14E, a major portion of the second
stalk is formed by two molecules of subunit b. Recent
results indicate that bacterial subunit (mitochondrial
OSCP) extends further up than is shown in Fig. 18-14,
and together with subunit F6 may form a cap at the
top of the
3

3
head.
230a, 230b
The F
0
portion of bacterial ATP synthase, which is
embedded in the membrane, consists of one 271-residue
subunit a, an integral membrane protein probably
with five transmembrane helices,
231,232
two 156-residue
b subunits, and ~ twelve 79-residue c subunits. The c
subunit is a proteolipid, insoluble in water but soluble
in some organic solvents. The structure of monomeric
c in chloroform:methanol:H
2
O (4:4:1) solution has been
determined by NMR spectroscopy. It is a hairpin
consisting of two antiparallel helices.
233
Twelve of
the c subunits are thought to assemble into a ring
with both the N and C termini of the subunit chains
in the periplasmic (or intermembrane) face of the
membrane.
234,235
The ratio of c to a subunits has been
difficult to measure but has been estimated as 9 12.
The fact that both genetically fused c
2
dimers and c
3
trimers form function F
0
suggested that they assemble
to a c
12
ring as shown in Fig. 18-14E.
236
However, the
recent crystallographic results that revealed a C
10
ring
214c
raise questions about stoichiometry.
Since ATP synthesis takes place in F
1
, it has long
been thought that the F
0
part of the ATP synthase
contains a proton channel, which leads from the
inside of the mitochondria to the F
1
assemblage.
146
Such a channel would probably not be an open pore
but a chain of hydrogen-bonded groups, perhaps
leading through the interior of the protein and able to
transfer protons via icelike conduction. One residue
in the c subunit, Asp 61, which lies in the center of
the second of the predicted transmembrane helices,
is critical for proton transport.
236a
Natural or artificial
mutants at this position (e.g., D61G or D61N) do not
transport protons. This carboxyl group also has an
unusually high reactivity and specificity toward the
protein-modifying reagent dicyclohexylcarbodiimide
(DCCD; see Eq. 3-10).
146,237
Modification of a single c
subunit with DCCD blocks the proton conductance.
An interesting mutation is replacement of alanine
62 of the c subunit with serine. This mutant will sup-
port ATP synthase using Li
+
instead of H
+
.
237
Certain
alkylophilic bacteria, such as Propionigenium modestum,
have an ATP synthase that utilizes the membrane
potential and a flow of Na
+
ions rather than protons
through the c subunits.
238 240c
The sodium transport
requires glutamate 65, which fulfills the same role as
D61 in E. coli, and also Q32 and S66. Study of mutants
revealed that the polar side chains of all three of these
residues bind Na
+
, that E65 and S66 are needed to bind
Li
+
, and that only E65 is needed for function with H
+
.
The a subunit is also essential for proton translo-
cation.
231,232,241
Structural work on this extremely
hydrophobic protein has been difficult, but many
mutant forms have been studied. Arginine 210 is
essential as are E219 and H245. However, if Q252 is
mutated to glutamate, E219 is no longer essential.
241
One of the OXPHOS diseases (NARP; Box 18-B) is a
result of a leucine-to-arginine mutation in human
subunit a.
241a
The b subunit is an elongated dimer,
largely of -helical structure.
242,243
Its hydrophobic
N terminus is embedded in the membrane,
229,244
while the hydrophilic C-terminal region interacts with
subunit of F
1
, in the stator structure (Fig. 18-14E).
Some of the F
0
subunits (d, e, f, g, A6L) may form a
collar around the lower end of the central stalk.
210a,b
How is ATP made? No covalent intermediates
have been identified, and isotopic exchange studies
indicate a direct dehydration of ADP and P
i
to form
bound ATP.
245
For the nucleophilic terminal phospho
group of ADP to generate a high-energy linkage
directly by attack on the phosphorus atom of P
i
an
OH

ion must be eliminated (Eq. 18-13). This is not a


probable reaction at pH 7, but it would be reasonable
at low pH. Thus, one function of the oriented ATP
C. Oxidative Phosphorylation
Ch 18IND page - E 2/14/03, 11:09 AM 1043
1044 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
synthase might be to deliver one or more protons
flowing in from F
0
specifically to the oxygen that is to
be eliminated (Eq. 18-13). As we have seen (Section 2),
on thermodynamic grounds 3 4 protons would prob-
ably be needed. Perhaps they could be positioned
nearby to exert a large electrostatic effect, or they
could assist in releasing the ATP formed from the
synthase by inducing a conformational change. How-
ever, it isnt clear how protons could be directed to the
proper spots.
Paul Boyers binding change mechanism. Boyer
and associates suggested that ATP synthesis occurs
rapidly and reversibly in a closed active site of the
ATP synthase in an environment that is essentially
anhydrous. ATP would then be released by an energy-
dependent conformational change in the protein.
245 249
Oxygen isotope exchange studies verified that a rapid
interconversion of bound ADP, P
i
, and ATP does occur.
Studies of soluble ATP synthase, which is necessarily
uncoupled from electron transport or proton flow,
shows that ATP is exceedingly tightly bound to F
1
as
expected by Boyers mechanism.
248
According to his
conformational coupling idea, protons flowing
across the membrane into the ATP synthase would in
some way induce the conformational change necessary
for release of ATP.
The idea of conformational coupling of ATP syn-
thesis and electron transport is especially attractive
when we recall that ATP is used in muscle to carry out
mechanical work. Here we have the hydrolysis of
ATP coupled to motion in the protein components of
the muscle. It seems reasonable that ATP should be
formed as a result of motion induced in the protein
components of the ATPase. Support for this analogy
has come from close structural similarities of the F
1
ATPase subunits and of the active site of ATP cleav-
age in the muscle protein myosin (Chapter 19).
A simple version of Boyers binding change
mechanism is shown in Figure 18-15. The three F
1

subunits are depicted in three different conformations.
In O the active site is open, in T it is closed, and if ATP
is present in the active site it is tightly bound. In the
low affinity L conformation ligands are bound weakly.
In step a MgADP and P
i
enter the L site while MgATP
is still present in the T site. In step b a protonic-energy-
dependent step causes synchronous conformational
changes in all of the subunits. The tight site opens and
MgATP is free to leave. At the same time MgADP and
P
i
in the T site are converted spontaneously to tightly
bound ATP. The MgATP is in reversible equilibrium
with MgADP + P
i
, which must be bound less tightly
than is MgATP. That is, the high positive value of G
for formation of ATP must be balanced by a correspond-
ing negative G for a conformational or electronic
reorganization of the protein in the T conformation.
Opening of the active site in step b of Fig. 18-15 will
have a high positive G unless it is coupled to proton
flow throught F
0
. Of three sites in the subunits, one
binds MgATP very tightly (K
d
~ 0.1 M) while the
other sites bind less tightly (K
d
~ 20 M).
250,251
How-
ever, it has been very difficult to establish binding
constants or K
m
values for the ATPase reaction.
248
Each of the three sites probably, in turn, becomes
the high-affinity site, consistent with an ATP synthase
mechanism involving protein conformational changes.
Rotational catalysis. Boyer suggested that there
is a cyclic rotation in the conformations of the three
subunits of the ATP synthase, and that this might
involve rotation about the stalk. By 1984, it had been
shown that bacterial flagella are rotated by a protonic
motor (Chapter 19), and a protic rotor for ATP synthase
had been proposed by Cox et al.
252
and others.
245
However, the b subunits were thought to be in the
central stalk.
222
More recently chemical crosslinking
experiments,
201,253
as well as electron microscopy,
confirmed the conclusion that an intact stator struc-
ture must also be present as in Fig. 18-14E.
202
The
necessary second stalk is visible in CF
1
F
0
ATPase of
chloroplasts
230
and also in the related vacuolar ATPase,
a proton or Na
+
pump from a clostridium.
254
See also
Section 5. Another technique, polarized absorption
recovery after photobleaching, was applied after
labeling of Cys 322, the penultimate residue at the C
terminus of the subunit with the dye eosin. After
photobleaching with a laser beam the polarization
of the light absorption by the dye molecule relaxed
because of rotation. Relaxation was observed when
ATP was added but not with ADPPNP.
202,255,256
The most compelling experiments were performed
by Noji et al.
202,257 260
They prepared the
3

3
sub-
complex of ATPase from a thermophilic bacterium.
The complex was produced in E. coli cells from the
cloned genes allowing for some engineering of the
proteins. A ten-histidine tag was added at the N
termini of the subunits so that the complex could
be glued to a microscope coverslip coated with a
nickel complex with a high affinity for the His tags.
The subunit shafts protrude upward as shown in
Fig. 18-16. The subunit was mutated to replace its
(18-13)
O
P
O
Adenosine
P
O O
O
O
O P O
O
O
OH
H
Mg
2

H
O
C
O

MgATP

H
2
O
Nucleophilic phospho
group of ADP attacks
P
i
to form ATP
Proton delivered to this
point by COOH side
chain makes oxygen more
electrophilic and permits
displacement of H
2
O
Ch 18IND page - E 2/14/03, 11:09 AM 1044
1045
a
b
c
d
ADP + P
i
L
O T
A
T
P
A
T
P
ADP + P
i
L
O T
L O
T
ATP
L O
T
A
T
P
ADP + P
i
ATP
Energy-dependent
and cooperative
conformational
change
Figure 18-15 Boyers binding change mechanism for ATP
synthase in a simple form. After Boyer
245
but modified to
include a central camshaft which may drive a cyclic alter-
ation in conformations of the subunits. The small pointer
on this shaft is not to be imagined as real but is only an
indicator of rotation with induced conformational changes.
The rotation could occur in 120 steps rather than the smaller
steps suggested here.
C. Oxidative Phosphorylation
Figure 18-16 (A) The system used for observation of the
rotation of the subunit in the
3

3
subcomplex. The
observed direction of the rotation of the subunit is indicated
by the arrows. (B) Sequential
images of a rotating actin
filament attached as in (A).
(C) Similar images obtained
with the axis of rotation near
the middle of the filament.
The images correspond to
the view from the top in (A).
Total length of the filament,
2.4 m; rotary rate, 1.3 revolu-
tions per second; time inter-
val between images, 33 ms.
From Noji et al.
258

Coverslip coated with nickel nitriloacetate


His-tag
Streptavidin
Actin filament

3
complex
A
only cysteine by serine and to introduce a cysteine
in place of Ser 107 of the stalk region of . The new
cysteine was biotinylated and attached to streptavidin
(see Box 14-B) which was also attached to a fluorescently
labeled actin filament (Fig. 7-10) ~13 m in length as
shown in Fig. 18-16. The actin fiber rotated in a counter-
clockwise direction when ATP was added but did not
rotate with AMPPNP. At low ATP concentrations
the rotation could be seen to occur in discrete 120
steps.
258,261,262
Each 120 step seems to consist of ~ 90
and ~ 30 substeps, each requiring a fraction of a milli-
second.
262a
The ATPase appears to be acting as a
stepper motor, hydrolysis of a single ATP turning
the shaft 120. Rotation at a rate of ~14 revolutions
per second would require the hydrolysis of ~ 42 ATP
per second. If the motor were attached to the F
0
part
it would presumably pump four (or perhaps three) H
+
across a membrane for each ATP hydrolyzed. Acting
in reverse, it would make ATP. A modification of the
experiment of Fig. 18-16 was used to demonstrate
that the c subunits also rotate with respect to the
3

3
head.
262b
Other experiments support rotation of the
c ring relative to subunit a.
262c,d
Still to be answered are important questions.
How does ATP hydrolysis turn the shaft? Are four
H
+
pumped for each step, or are there smaller single
proton substeps? Is the simple picture in Fig. 18-15
correct or, as proposed by some investigators,
263 265
must all three subunits be occupied for maximum
catalytic activity?
266
How is the coupling of H
+
trans-
port to mechanical motion accomplished?
267,267ad
5. ATP-driven Proton Pumps
Not all proton pumps are driven by electron trans-
port. ATP synthase is reversible, and if p is low, hydro-
lysis of ATP can pump protons out of mitochondria
or across bacterial plasma membranes.
268
Cells of
Streptococcus faecalis, which have no respiratory chain
B
C
Ch 18IND page - E 2/14/03, 11:09 AM 1045
1046 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
and form ATP by glycolysis, use an F
1
F
0
ATP synthase
complex to pump protons out to help regulate cyto-
plasmic pH.
268
Similar vacuolar (V-type) H
+
-ATPases
or V
1
V
0
ATPases pump protons into vacuoles, Golgi
and secretory vesicles, coated vesicles, and lyso-
somes
198,269-270
in every known type of eukaryotic
cell.
271,272
These proton pumps are similar in appear-
ance (Fig. 18-14,B) and in structure to F
1
F
0
ATPases.
272ac
The 65- to 77-kDa A subunits and 55- to 60-kDa B
subunits are larger than the corresponding F
1
F
0
and
subunits. Accessory 40-, 39-, and 33-kDa subunits
are also present in V
1
. The V
0
portion appears to
contain a hexamer of a 16-kDa proteolipid together
with 110- and 21-kDa subunits.
271
V-type ATPases are
also found in archaebacteria
271,273
and also in some
clostridia
254
and other eubacteria.
273a
A type of proton
pump, the V-PPase, uses hydrolysis of inorganic
pyrophosphate as a source of energy.
274
It has been
found in plants, in some phototrophic bacteria, and
in acidic calcium storage vesicles (acidocalcisomes) of
trypanosomes.
274a
Other ATP-dependent proton pumps are present
in the plasma membranes of yeast and other fungi
274b
and also in the acid-secreting parietal cells of the
stomach (Fig. 18-17). The H
+
-ATPase of Neurospora
pumps H
+
from the cytoplasm without a counterion.
It is electrogenic.
275,275a
However, the gastric H
+
,K
+
-
ATPase exchanges H
3
O
+
for K
+
and cleaves ATP with
formation of a phosphoenzyme.
276
It belongs to the
family of P-type ion pumps that includes the mamma-
lian Na
+
,K
+
-ATPase (Fig. 8-25) and Ca
2+
-ATPase (Fig.
8-26). These are discussed in Chapter 8. The H
+
,K
+
-
ATPases, which are widely distributed within eukary-
otes, are also similar, both in sequence and in the fact
that a phospho group is transferred from ATP onto a
carbox-ylate group of an aspartic acid residue in the
protein. All of them, including a Mg-ATPase of Salmo-
nella, are two-subunit proteins. A large catalytic
subunit contains the site of phosphorylation as well
as the ATP- and ion-binding sites. It associates nonco-
valently with the smaller heavily glycosylated sub-
unit.
276 278
For example, the rabbit H
+
,K
+
-ATPase
consists of a 1035-residue chain which has ten trans-
membrane segments and a 290-residue chain with
a single transmembrane helix and seven N-linked
glycosylation sites.
278
6. Uncouplers and Energy-linked Processes in
Mitochondria
Many compounds that uncouple electron trans-
port from phosphorylation, like 2,4-dinitrophenol, are
weak acids. Their anions are nucleophiles. According
to the scheme of Fig. 18-12, they could degrade a high
energy intermediate, such as Y ~ B, by a nucleophilic
attack on Y to give an inactive but rapidly hydrolyzed
Surface mucous cells
Mucous neck cells
Somatostatin cell
Endocrine cell
Chief cells
Parietal cells
derivative of Y. On the other hand, according to Mitchells
hypothesis uncouplers facilitate the transport of protons
back into the mitochondria thereby destroying p.
The fact that the anions of the uncouplers are large,
often aromatic, and therefore soluble in the lipid bi-
layer supports this interpretation; the protonated
uncouplers can diffuse into the mitochondria and the
anion can diffuse back out. Mitochondria can also be
uncoupled by a combination of ionophores, e.g., a
mixture of valinomycin (Fig. 8-22), which carries K
+
into the mitochondria, plus nigericin, which catalyzes
an exchange of K
+
(out) for H
+
(in).
172
The uncoupler carbonyl cyanide p-(trifluoro-
methoxy)phenylhydrazone (FCCP) and related com-
pounds are widely used in biochemical studies. Their
action can be explained only partially by increased
proton conduction.
Figure 18-17 Schematic diagram of an acid-producing
oxyntic gland of the stomach. The normal human stomach
contains about 10
9
parietal (oxyntic) cells located in the walls
of these glands. From Wolfe and Soll.
279
Modified from Ito.
These glands also produce mucus, whose role in protecting
the stomach lining from the high acidity is uncertain.
280
O N
N C
CN
CN F
3
C
H
Carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone (FCCP)
Ch 18IND page - E 2/14/03, 11:09 AM 1046
1047
Uncoupling is sometimes important to an organism.
The generation of heat by uncoupling is discussed in
Box 18-C. The fungus Bipolaris maydis caused a crisis
in maize production when it induced pore formation
in mitochondrial membranes of a special strain used
in production of hybrid seeds.
281,282
Synthesis of ATP by mitochondria is inhibited by
oligomycin, which binds to the OSCP subunit of ATP
synthase. On the other hand, there are processes that
require energy from electron transport and that are
not inhibited by oligomycin. These energy-linked
processes include the transport of many ions across
the mitochondrial membrane (Section E) and reverse
electron flow from succinate to NAD
+
(Section C,2).
Dinitrophenol and many other uncouplers block the
reactions, but oligomycin has no effect. This fact can
be rationalized by the Mitchell hypothesis if we assume
that p can drive these processes.
Another energy-linked process is the transhydro-
genase reaction by which NADH reduces NADP
+
to
form NADPH. In the cytoplasm various other reac-
tions are used to generate NADPH (Chapter 17, Sec-
tion I), but within mitochondria a membrane-bound
transhydrogenase has this function.
283 286a
It couples
the transhydrogenation reaction to the transport of
one (or possibly more than one) proton back into the
mitochondria (Eq. 18-14). A value of p of 180 mV
could increase the ratio of [NADPH] / [NADP
+
] within
mitochondria by a factor of as much as 1000.
Transhydrogenases function in a similar way within
bacteria. Whether from E. coli, photosynthetic bacteria,
or bovine mitochondria, transhydrogenases have
similar structures.
285
Two 510-residue subunits
associate with two 462-residue subunits to form an

2
tetramer with 10 14 predicted transmembrane
helices. The subunits contain separate NAD(H) and
NADP(H) binding sites. A conformational change
appears to be associated with the binding or release
of the NADP
+
or NADPH.
287
D. Transport and Exchange across
Mitochondrial Membranes
Like the external plasma membrane of cells, the
inner mitochondrial membrane is selective. Some
D. Transport and Exchange across Mitochondrial Membranes
Transhydrogenase
H
+
+
+
+
+
+
+
H
+

NADH + NADP
+
NAD
+
+ NADPH
Membrane
(18-14)
nonionized materials pass through readily but the
transport of ionic substances, including the anions of
the dicarboxylic and tricarboxylic acids, is restricted.
In some cases energy-dependent active transport is
involved but in others one anion passes inward in
exchange for another anion passing outward. In either
case specific translocating carrier proteins are needed.
Solutes enter mitochondria through pores in thou-
sands of molecules of the voltage-gated anion-
selective channel VDAC, also known as mitochon-
drial porin.
15,16,288,289
In the absence of a membrane
potential these pores allow free diffusion to molecules
up to ~ 1.2 kDa in mass and may selectively permit
passage of anions of 3- to 5-kDa mass. However, a
membrane potential greater than ~ 20 mV causes the
pores to close. NADH also decreases permeability.
In the closed state the outer membrane becomes
almost impermeant to ATP.
289,290
An example of energy-dependent transport is the
uptake of Ca
2+
by mitochondria. As indicated in the
lower part of Fig. 18-13, there are two possibilities for
preservation of electrical neutrality according to the
chemiosmotic theory. Counterions X

may flow out to


balance the protons discharged on the outside. On the
other hand, if a cation such as Ca
2+
flows inward to
balance the two protons flowing outward, neutrality
will be preserved and the mitochondrion will accumu-
late calcium ions. Experimentally such accumulations
via a calcium uniporter
4
are observed to accompany
electron transport. In the presence of a suitable iono-
phore energy-dependent accumulation of potassium
ions also takes place.
291
In contrast, an electroneutral
exchange of one Ca
2+
for two Na
+
is mediated by a
Na
+
Ca
2+
exchanger.
292,293
It permits Ca
2+
to leave
mitochondria. A controversial role of mitochondria in
accumulating Ca
2+
postulates a special rapid uptake
mode of exchange (see p. 1049).
294
It is thought that glutamate enters mitochondria
as the monoanion Glu

in exchange for the dianion of


aspartate Asp
2
. Like the uptake of Ca
2+
this exchange
is driven by p. Since a membrane potential can be
created by this exchange in the absence of p, the
process is electrogenic.
4
In contrast, an electroneutral
exchange of malate
2
and 2-oxoglutarate
2
occurs by
means of carriers that are not energy-linked.
295,296
This
dicarboxylate transporter is only one of 35 structurally
related mitochondrial carriers identified in the complete
genome of yeast.
296,297
Another is the tricarboxylic
transporter (citrate transport protein) which exchanges
the dianionic form of citrate for malate, succinate, iso-
citrate, phosphoenolpyruvate, etc.
298,298a,b
The important adenine nucleotide carrier
takes ADP into the mitochondrial matrix for phos-
phorylation in a 1:1 ratio with ATP that is exported
into the cytoplasm.
299-300b
This is one of the major
rate-determining processes in respiration. It has been
widely accepted that the carrier is electrogenic,
300
Ch 18IND page - E 2/14/03, 11:09 AM 1047
1048 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
A secondary but important role of metabolism
in warm-blooded animals is to generate heat. The
heat evolved from ordinary metabolism is often
sufficient, and an animal can control its temperature
by regulating the heat exchange with the environ-
ment. Shivering also generates heat and is used
from birth by pigs.
a
However, this is insufficient for
many newborn animals, for most small mammals
of all ages, and for animals warming up after hiber-
nation. The need for additional heat appears to be
met by brown fat, a tissue which contrasts strikingly
with the more abundant white adipose tissue. Brown
fat contains an unusually high concentration of
blood vessels, many mitochondria with densely
packed cristae, and a high ratio of cytochrome c
oxidase to ATP synthase. Also present are a large
number of sympathetic nerve connections, which
are also related to efficient generation of heat. New-
born humans have a small amount of brown fat,
and in newborn rabbits it accounts for 5 6% of the
body weight.
b d
It is especially abundant in species
born without fur and in hibernating animals.
Swordfish also have a large mass of brown adipose
tissue that protects their brains from rapid cooling
when traveling into cold water.
a
The properties of brown fat pose an interesting
biochemical question. Is the energy available from
electron transport in the mitochondria dissipated
as heat because ATP synthesis is uncoupled from
electron transport? Or does ATP synthesis take
place but the resulting ATP is hydrolyzed wastefully
through the action of ATPases? Part of the answer
came from the discovery that mitochondria of
brown fat cells synthesize a 32-kDa uncoupling
protein (UCP1 or thermogenin). It is incorporated
into the inner mitochondrial membranes where it
may account for 10 15% of total protein.
d f
This
protein, which is a member of a family of mitochon-
drial membrane metabolic carriers (Table 18-8),
provides a short-circuit that allows the protonmo-
tive force to be dissipated rapidly, perhaps by a flow
of protons out through the uncoupling protein.
g i
Synthesis of the uncoupling protein is induced by
exposure to cold, but when an animal is warm the
uncoupling action is inhibited.
The uncoupling protein resembles the ATP/ADP
and phosphate anion carriers (Table 18-8),
g,i
which
all have similar sizes and function as homodimers.
Each monomeric subunit has a triply repeated ~100-
residue sequence, each repeat forming two trans-
membrane helices. Most mitochondrial transporters
carry anions, and UCP1 will transport Cl

.
h,i
How-
ever, the relationship of chloride transport to its real
function is unclear. Does the protein transport H
+
BOX 18-C USING METABOLISM TO GENERATE HEAT: THERMOGENIC TISSUES
into, or does it carry HO

out from, the mitochon-


drial matrix?
g,h
Another possibility is that a fatty
acid anion binds H
+
on the intermembrane surface
and carries it across into the matrix as an unionized
fatty acid. The fatty acid anion could then pass back
out using the anion transporter function and assisted
by the membrane potential.
h,i
The uncoupling protein is affected by several
control mechanisms. It is inhibited by nucleotides
such as GDP, GTP, ADP, and ATP which may bind at
a site corresponding to that occupied by ATP or ADP
in the ADP/ATP carrier.
i
Uncoupling is stimulated
by noradrenaline,
f
which causes a rapid increase in
heat production by brown fat tissues, apparently
via activation of adenylate cyclase. Uncoupling is
also stimulated by fatty acids.
j
Recently UCP1 and
related uncoupling proteins have been found to
require both fatty acids and ubiquinone for
activity.
jj,jk
It has been suggested that brown adipose tissue
may also function to convert excess dietary fat into
heat and thereby to resist obesity.
k m
Mice lacking
the gene for the mitochondrial uncoupling protein
are cold-sensitive but not obese. However, other
proteins, homologous to UCP1, have been discov-
ered. They may partially compensate for the loss.
m,h
The bombardier beetle generates a hot, quinone-
containing defensive discharge, which is sprayed
in a pulsed jet from a special reaction chamber at a
temperature of 100C.
n p
The reaction mixture of
25% hydrogen peroxide and 10% hydroquinone
plus methylhydroquinone is stored in a reservoir as
shown in the accompanying figure and reacts with
explosive force when it comes into contact with
catalase and peroxidases in the reaction chamber.
The synthesis and storage of 25% H
2
O
2
poses inter-
esting biochemical questions!
Some plant tissues are thermogenic. For exam-
ple, the spadix (or inflorescence, a sheathed floral
spike) of the skunk cabbage Symplocarpus foetidus
can maintain a 15 35C higher temperature than
that of the surrounding air.
q
The voodoo lily in a
single day heats the upper end of its long spadex
to a temperature 22C above ambient, volatilizing
a foul smelling mixture of indoles and amines.
r,s
This is accomplished using the alternative oxidase
system
s
(Box D in Fig. 18-6). The lotus flower main-
tains a temperature of 30 35C, while the ambient
temperature may vary from 10 30C.
t
While the
volatilization of insect attractants may be the primary
role for thermogenesis in plants, the warm flowers
may also offer an important reward to insect polli-
nators. Beetles and bees require thoracic tempera-
tures above 30C to initiate flight and, therefore,
Ch 18IND page - E 2/14/03, 11:09 AM 1048
1049
BOX 18-C (continued)
benefit from the warm flowers.
t
While in flight bees
vary their metabolic heat production by altering
their rate of flight, hovering, and other changes in
physical activity.
u
a
Tyler, D. D. (1992) The Mitochondrion in Health and Disease, VCH
Publ., New York
b
Dawkins, M. J. R., and Hull, D. (1965) Sci. Am. 213(Aug), 6267
c
Lindberg, O., ed. (1970) Brown Adipose Tissue, Am. Elsevier,
New York
d
Nicholls, D. G., and Rial, E. (1984) Trends Biochem. Sci. 9, 489491
D. Transport and Exchange across Mitochondrial Membranes
bringing in ADP
3
and exporting ATP
4
in an exchange
driven by p. However, an electroneutral exchange,
e.g., of ADP
3
for ATP
3
, may also be possible. The car-
rier is an ~ 300-residue 32-kDa protein, which is spe-
cifically inhibited by the plant glycoside atractyloside
or the fungal antibiotic bongkrekate. The carrier is
associated with bound cardiolipin.
301
This one trans-
porter accounts for ~ 10% of all of the mitochondrial
protein.
302,303
A separate dimeric carrier allows P
i
to enter, prob-
ably as H
2
PO
4

.
304-305a
This ion enters mitochondria in
an electroneutral fashion, either in exchange for OH

or by cotransport with H
+
. A less important carrier
306
exchanges HPO
4
2
for malate
2
. Several other trans-
porters help to exchange organic and inorganic ions.
One of them allows pyruvate to enter mitochondria
in exchange for OH

or by cotransport with H
+
. Some
of the identified carriers are listed in Table 18-8. As
discussed in Chapter 8, exchange carriers are also
important in plasma membranes of organisms from
bacteria to human beings. For example, many meta-
bolites enter cells by cotransport with Na
+
using the
energy of the Na
+
gradient set up across the membrane
by the Na
+
,K
+
pump.
Under some circumstances the inner membrane
develops one or more types of large-permeability pore.
An increase in Ca
2+
may induce opening of an unselective
pore which allows rapid uptake of Ca
2+
.
294,307,307a
A
general anion-specific channel may be involved in
volume homeostasis of mitochondria.
308
Mitochondria are not permeable to NADH. How-
ever, reactions of glycolysis and other dehydrogenations
in the cytoplasm quickly reduce available NAD
+
to
NADH. For aerobic metabolism to occur, the reducing
equivalents from the NADH must be transferred into
the mitochondria. Fungi and green plants have solved
R
OH +
OH
H
2
O
2
Reservoir
Vestibule
R = H, CH
3
R
O
H
2
O
2
+ O
2
+
O
m
E
Reservoir and reaction vessel of the bombardier beetle.
From D. J. Aneshansley, et al.
n
E, for enzyme
m, muscle that controls valve
e
Cooney, G. J., and Newsholme, E. A. (1984) Trends Biochem. Sci.
9, 303305
f
Ricquier, D., Casteilla, L., and Bouillaud, F. (1991) FASEB J. 5,
22372242
g
Klingenberg, M. (1990) Trends Biochem. Sci. 15, 108112
h
Jaburek, M., Varecha, M., Gimeno, R. E., Dembski, M., Jezek, P.,
Zhang, M., Burn, P., Tartaglia, L. A., and Garlid, K. D. (1999) J.
Biol. Chem. 274, 2600326007
i
Gonzlez-Barroso, M. M., Fleury, C., Levi-Meyrueis, C.,
Zaragoza, P., Bouillaud, F., and Rial, E. (1997) Biochemistry 36,
1093010935
j
Hermesh, O., Kalderon, B., and Bar-Tana, J. (1998) J. Biol. Chem.
273, 39373942
jj
Echtay, K. S., Winkler, E., and Klingenberg, M. (2000) Nature
(London) 408, 609613
jk
Echtay, K. S., Winkler, E., Frischmuth, K., and Klingenberg, M.
(2001) Proc. Natl. Acad. Sci. U.S.A. 98, 14161421
k
Rothwell, N. J., and Stock, M. J. (1979) Nature (London) 281, 31
35
l
Tai, T.-A. C., Jennermann, C., Brown, K. K., Oliver, B. B.,
MacGinnitie, M. A., Wilkison, W. O., Brown, H. R., Lehmann,
J. M., Kliewer, S. A., Morris, D. C., and Graves, R. A. (1996)
J. Biol. Chem. 271, 2990929914
m
Enerbck, S., Jacobsson, A., Simpson, E. M., Guerra, C.,
Yamashita, H., Harper, M.-E., and Kozak, L. P. (1997) Nature
(London) 387, 9094
n
Aneshansley, D. J., Eisner, T., Widom, J. M., and Widom, B.
(1969) Science 165, 6163
o
Eisner, T., and Aneshansley, D. J. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 97059709
p
Dean, J., Aneshansley, D. J., Edgerton, H. E., and Eisner, T.
(1990) Science 248, 12191221
q
Knutson, R. M. (1974) Science 186, 746747
r
Diamond, J. M. (1989) Nature (London) 339, 258259
s
Rhoads, D. M., and McIntosh, L. (1991) Proc. Natl. Acad. Sci.
U.S.A. 88, 21222126
t
Seymour, R. S., and Schultze-Motel, P. (1996) Nature (London)
383, 305
u
Harrison, J. F., Fewell, J. H., Roberts, S. P., and Hall, H. G. (1996)
Science 274, 88 90
Ch 18IND page - E 2/14/03, 11:09 AM 1049
1050 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
ADP
3
ATP
3
or ADP
3
ATP
4
Electrogenic symport
H
2
PO
4

+ H
+
Electroneutral symport
or H
2
PO
4

OH

HPO
4
2
Malate
2
Malate
2
2-Oxoglutarate
2
Glutamate
2
+ H
+
Aspartate
2
Glutamate

OH

Pyruvate

OH

or Pyruvate

+ H
+
Electroneutral symport
Citrate
3
+ H
+
Malate
2
Ornithine
+
H
+
Acylcarnitine Carnitine
2 Na
+
Ca
2+
H
+
K
+
H
+
Na
+
General transporters
VDAC (porin) Outer membrane
Large anion pores Inner membrane
TABLE 18-8
Some Mitochondrial Membrane Transporters
a
Ion Diffusing In Ion Diffusing Out Comment
b
a
From Nicholls and Ferguson
172
and Tyler
4
.
b
Unless indicated otherwise the transporters are antiporters that
catalyze an electroneutral ion exchange.
the problem by providing two NADH dehydrogenases
embedded in the inner mitochondrial membranes (Fig.
18-6). One faces the matrix space and oxidizes the
NADH produced in the matrix while the second faces
outward to the intermembrane space and is able to
oxidize the NADH formed in the cytoplasm. In ani-
mals the reducing equivalents from NADH enter the
mitochondria indirectly. There are several mecha-
nisms, and more than one may function simulta-
neously in a tissue.
In insect flight muscle, as well as in many mam-
malian tissues, NADH reduces dihydroxyacetone
phosphate. The resulting sn-3-glycerol P passes
through the permeable outer membrane of the mito-
chondria, where it is reoxidized to dihydroxyacetone
phosphate by the FAD-containing glycerol-phosphate
dehydrogenase embedded in the outer surface of the
inner membrane (Figs. 18-5, 18-6). The dihydroxyac-
etone can then be returned to the cytoplasm. The
overall effect of this glycerol-phosphate shuttle
(Fig. 18-18A) is to provide for mitochondrial oxidation
of NADH produced in the cytoplasm. In heart and
liver the same function is served by a more complex
malateaspartate shuttle (Fig. 18-18B).
309
Reduction
of oxaloacetate to malate by NADH, transfer of malate
into mitochondria, and reoxidation with NAD
+
accom-
plishes the transfer of reduction equivalents into the
mitochondria. Mitochondrial membranes are not very
permeable to oxaloacetate. It returns to the cytoplasm
mainly via transamination to aspartate, which leaves
the mitochondria together with 2-oxoglutarate. At
the same time glutamate enters the mitochondria in
exchange for aspartate. The 2-oxoglutarate presumably
exchanges with the entering malate as is indicated in
Fig. 18-18B. The export of aspartate may be energy-
linked as a result of the use of an electrogenic carrier
that exchanges glutamate

+ H
+
entering mitochondria
for aspartate

leaving the mitochondria. Thus, p may


help to expel aspartate from mitochondria and to drive
the shuttle.
The glycerol-phosphate shuttle, because it depends
upon a mitochondrial flavoprotein, provides ~ 2 ATP per
electron pair (P/ O = 2), whereas the malateaspartate
shuttle may provide a higher yield of ATP. The glycerol-
phosphate shuttle is essentially irreversible, but the
reactions of the malateaspartate shuttle can be reversed
and utilized in gluconeogenesis (Chapter 17).
E. Energy from Inorganic Reactions
Some bacteria obtain all of their energy from inor-
ganic reactions. These chemolithotrophs usually
have a metabolism that is similar to that of hetero-
trophic organisms, but they also have the capacity to
obtain all of their energy from an inorganic reaction.
In order to synthesize carbon compounds they must
be able to fix CO
2
either via the reductive pentose
phosphate cycle or in some other way. The chloro-
plasts of green plants, using energy from sunlight,
supply the organism with both ATP and the reducing
agent NADPH (Chapter 17). In a similar way the
lithotrophic bacteria obtain both energy and reducing
materials from inorganic reactions.
Chemolithotrophic organisms often grow slowly,
making study of their metabolism difficult.
310
Never-
theless, these bacteria usually use electron transport
chains similar to those of mitochondria. ATP is
formed by oxidative phosphorylation, the amount
formed per electron pair depending upon the number
of proton-pumping sites in the chain. This, in turn,
depends upon the electrode potentials of the reactions
involved. For example, H
2
, when oxidized by O
2
, leads
to passage of electrons through the entire electron
transport chain with synthesis of ~ 3 molecules of ATP
per electron pair. On the other hand, oxidation by O
2
of nitrite, for which E (pH 7) = +0.42 V, can make use
only of the site III part of the chain. Not only is the
yield of ATP less than in the oxidation of H
2
but also
there is another problem. Whereas reduced pyridine
Ch 18IND page - E 2/14/03, 11:09 AM 1050
1051
nucleotides needed for biosynthesis can be generated
readily from H
2
, nitrite is not a strong enough reducing
agent to reduce NAD
+
to NADH. The only way that
reducing agents can be formed in cells utilizing oxi-
dation of nitrite as an energy source is via reverse
electron flow driven by hydrolysis of ATP or by p.
Such reverse electron flow is a common process for
many chemolithotrophic organisms.
Let us consider the inorganic reactions in two
groups: (1) oxidation of reduced inorganic compounds
by O
2
and (2) oxidation reactions in which an inorganic
oxidant, such as nitrate or sulfate, substitutes for O
2
.
The latter reactions are often referred to as anaerobic
respiration.
1. Reduced Inorganic Compounds as
Substrates for Respiration
The hydrogen-oxidizing bacteria. Species from
several genera including Hydrogenomonas, Pseudomonas,
and Alcaligenes oxidize H
2
with oxygen:
H
2
+ 1/2 O
2
H
2
O
G = 237.2 kJ mol
1
Figure 18-18 (A) The glycerol-
phosphate shuttle and (B) the
malateaspartate shuttle for trans-
port from cytoplasmic NADH into
mitochondria. The heavy arrows
trace the pathway of the electrons
(as 2H) transported.
Inner membrane Mitochondrial matrix Cytoplasm
Dihydroxyacetone
phosphate
FADH
2
FAD + 2H
+
Malate
NAD
+
NADH + H
+
Oxaloacetate
aminotransferase
Aspartate
Glutamate
exchange
carriers
2-oxoglutarate 2-oxoglutarate
Glutamate
Aspartate
aminotransferase
Oxaloacetate
Malate
NAD
+
H
+
+ NADH
NAD
+
H
+
+ NADH
sn-Glycerol
3-phosphate
E. Energy from Inorganic Reactions
A
B Malate-aspartate shuttle
Glycerol phosphate shuttle
Some can also oxidize carbon monoxide:
CO + 1/2 O
2
CO
2
G = 257.1 kJ mol
1
The hydrogen bacteria can also oxidize organic
compounds using straightforward metabolic path-
ways. The key enzyme is a membrane-bound nickel-
containing hydrogenase (Fig. 16-26), which delivers
electrons from H
2
into the electron transport chain.
310a
A second soluble hydrogenase (sometimes called hy-
drogen dehydrogenase) transfers electrons to NADP
+
to form NADPH for use in the reductive pentose phos-
phate cycle and for other biosynthetic purposes.
Nitrifying bacteria. Two genera of soil bacteria
oxidize ammonium ion to nitrite and nitrate (Eqs. 18-17
and 18-18).
311
NH
4
+
+ 3/2 O
2
NO
2

+ 2 H
+
+ H
2
O (Nitrosomonas)
G (pH 7) = 272 kJ mol
1
NO
2

+ 1/2 O
2
NO
3

(Nitrobacter)
G (pH 7) = 76 kJ mol
1
The importance of these reactions to the energy
metabolism of the bacteria was recognized in 1895
by Winogradsky, who first proposed the concept of
chemiautotrophy. Because the nitrifying bacteria grow
(18-15)
(18-16)
(18-17)
(18-18)
Ch 18IND page - E 2/14/03, 11:09 AM 1051
1052 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
slowly (generation time ~ 10 12 h) it has been hard to
get enough cells for biochemical studies and progress
has been slow. The reaction catalyzed by Nitrosomonas
(Eq. 18-17) is the more complex; it occurs in two or
more stages and is catalyzed by two enzymes as illus-
trated in Fig. 18-19. The presence of hydrazine blocks
oxidation of hydroxylamine (NH
2
OH) in step b and
permits that intermediate to accumulate. The oxida-
tion of ammonium ion by O
2
to hydroxylamine (step a)
is endergonic with G (pH 7) = 16 kJ mol
1
and is
incapable of providing energy to the cell. It occurs by
a hydroxylation mechanism (see Section G). On the
other hand, the oxidation of hydroxylamine to nitrite
by O
2
in step b is highly exergonic with G (pH 7) =
228 kJ mol
1
. The hydroxylamine oxidoreductase
that catalyzes this reaction is a trimer of 63-kDa sub-
units, each containing seven c-type hemes and an
unusual heme P450, which is critical to the enzymes
function
312 314a
and which is covalently linked to a
tyrosine as well as to two cysteines.
The electrode potentials for the two- and four-
electron oxidation steps are indicated in Fig. 18-19. It
is apparent that step b can feed four electrons into the
electron transport system at about the potential of
ubiquinone. Two electrons are needed to provide a
cosubstrate (Section G) for the ammonia monooxy-
genase and two could be passed on to the terminal
cytochrome aa
3
oxidase. The stoichiometry of proton
pumping in complexes III and IV is uncertain, but if it
is assumed to be as shown in Fig. 18-19 and similar to
that in Fig. 18-5, there will be ~ 13 protons available to
be passed through ATP synthase to generate ~ 3 ATP
per NH
3
oxidized. However, to generate NADH for
reductive biosynthesis Nitrosomonas must send some
electrons to NADH dehydrogenase (complex I) using
reverse electron transport, a process that depends
upon p to drive the reaction via a flow of protons
through the NADH dehydrogenase from the peri-
plasm back into the bacterial cytoplasm (Fig. 18-19).
Nitrobacter depends upon a simpler energy-
yielding reaction (Eq. 18-18) with a relatively small
Gibbs energy decrease. The two-electron oxidation
delivers electrons to the electron transport chain at
E = +0.42 V. The third oxygen in NO
3

originates
from H
2
O, rather than from O
2
as might be suggested
by Eq. 18-18.
316,317
It is reasonable to anticipate that a
single molecule of ATP should be formed for each pair
of electrons reacting with O
2
. However, Nitrobacter
contains a confusing array of different cytochromes in
its membranes.
311
Some of the ATP generated by pas-
sage of electrons from nitrite to oxygen must be used to
drive a reverse flow of electrons through both a bc
1
-type
complex and NADH dehydrogenases. This generates
reduced pyridine nucleotides required for biosynthetic
reactions (Fig. 18-20).
An interesting feature of the structure of Nitrobacter
is the presence of several double-layered membranes
a
b
I
NADH
dehydrogenase
III
Cytochrome bc
1
complex
Membrane Periplasm Cytoplasm
H
+
H
+
NADH
NAD
+
+ H
+
Reversed
electron
transport
+ 0.06 V
Hydroxylamine
oxidoreductase
QH
2
8H
+
Q
8H
+
+ 0.90 V
Ammonia
monooxygenase
IV
Cytochrome aa
3
oxidase
4H
+
4H
+
H
2
O
2H
+
O
2
1
2
Cyt c
552
2e

2e

NH
2
OH
H
2
O
5H
+
NO
2

NH
3
2H
+
O
2
NH
2
OH
H
2
O
4e

Cyt c
554
Figure 18-19 The ammonia oxidation system of the bac-
teriumNitrosomonas. Oxidation of ammonium ion (as free
NH
3
) according to Eq. 18-17 is catalyzed by two enzymes.
The location of ammonia monooxygenase (step a) is uncer-
tain but hydroxylamine oxidoreductase (step b) is periplas-
mic. The membrane components resemble complexes I, III,
and IV of the mitochondrial respiratory chain (Fig. 18-5) and
are assumed to have similar proton pumps. Solid green lines
trace the flow of electrons in the energy-producing reactions.
This includes flow of electrons to the ammonia monoxy-
genase. Complexes III and IV pump protons out but complex
I catalyzes reverse electron transport for a fraction of the
electrons from hydroxylamine oxidoreductase to NAD
+
.
Modified from Blaut and Gottschalk.
315
which completely envelop the interior of the cell.
Nitrite entering the cell is oxidized on these mem-
branes and cannot penetrate to the interior, where it
might have toxic effects.
The sulfur-oxidizing bacteria. Anaerobic condi-
tions prevail in marine sediments, in poorly stirred
swamps, and around hydrothermal vents at the bottom
of the sea. Sulfate-reducing bacteria form high con-
centrations (up to mM) of H
2
S (in equilibrium with
HS

and S
2
)
318 320
This provides the substrate for bac-
teria of the genus Thiobacillus, which are able to oxidize
sulfide, elemental sulfur, thiosulfate, and sulfite to sulfate
and live where the aerobic and anaerobic regions
meet.
311,321 323
Most of these small gram-negative
Ch 18IND page - E 2/14/03, 11:09 AM 1052
1053
organisms, found in water and soil, are able to grow in
a simple salt medium containing an oxidizable sulfur
compound and CO
2
. One complexity in understanding
their energy-yielding reactions is the tendency of sulfur
to form chain molecules. Thus, when sulfide is oxi-
dized, it is not clear that it is necessarily converted to
monoatomic elemental sulfur as indicated in Eq. 18-19.
Elemental sulfur (S
8
0
) often precipitates. In Beggiotoa,
another sulfide-oxidizing bacterium, sulfur is often
seen as small globules within the cells. Fibrous sulfur
precipitates are often abundant in the sulfide-rich
layers of ponds, lakes, and oceans.
318
The reactions of Eq. 18-19 occur in the periplasmic
space of some species.
315,323a,324
Steps a and b of
Figure 18-20 Electron transport system for oxidation of the
nitrite ion to the nitrate ion by Nitrobacter. Only one site of
proton pumping for oxidative phosphorylation is available.
Generation of NADH for biosynthesis requires two stages of
reverse electron transport.
NADH
dehydrogenase
Membrane Periplasm Cytoplasm
H
+
H
+
NADH
NAD
+
+ H
+
Reversed
electron
transport
2e

H
+
H
+
Cytochrome bc
1
Q
Cytochrome aa
3
oxidase
4H
+
4H
+
H
2
O
2H
+
O
2
1
2
Cyt c
550
2e

Nitrite
oxidase
2H
+
NO
3

H
2
O
NO
2

2e

2e

E. Energy from Inorganic Reactions


Eq. 18-19 are catalyzed by a 67-kDa sulfide dehydro-
genase in the periplasm of a purple photosynthetic
bacterium.
324
The enzyme consists of a 21-kDa sub-
unit containing two cytochrome c-like hemes, presum-
ably the site of binding of S
2
, and a larger 46-kDa
FAD-containing flavoprotein resembling glutathione
reductase.
324
The molybdenum-containing sulfite
oxidase (Fig. 16-32), which is found in the intermem-
brane space of mitochondria, may be present in the
periplasmic space of these bacteria. However, there is
also an intracellular pathway for sulfite oxidation (see
Eq. 18-22).
The sulfide-rich layers inhabited by the sulfur
oxidizers also contain thiosulfate, S
2
O
3
2
. It may arise,
in part, from reaction of glutathione with elemental
sulfur:
GSH + S
8
0
G S S
8
H
The linear polysulfide obtained by this reaction may
be oxidized, the sulfur atoms being removed from the
chain either one at a time to form sulfite or two at a
time to form thiosulfate.
322,322a
Thiosulfate is oxidized
by all species, the major pathway beginning with
cleavage to S
0
and SO
3
2
(Eq. 18-21, step a). At high
thiosulfate concentrations some may be oxidized to
tetrathionate (step b), which is hydrolyzed to sulfate
(step c).
Oxidation of sulfite to sulfate within cells occurs
by a pathway through adenosine 5'-phosphosulfate
(APS, adenylyl sulfate). Oxidation via APS (Eq. 18-22)
provides a means of substrate-level phosphorylation,
H
2
S
2H
+
+ 2e

S
0
6H
+
+ 4e

3H
2
O
SO
3
2
2H
+
+ 2e

H
2
O
Sulfite
oxidase
SO
4
2
a
b
c
(18-19)
(18-20)
a
b
c
H
2
O
SO
4
2
SSO
3

Thiosulfate
S
0
+ SO
3
2

O
3
SSSSO
3

Tetrathionite
2H
+
SSO
3

+ S
0
2e

cyt c
(18-21)
S O
P
O
O
Adenine
OH OH
O O

O
O
O

Adenosine 5-phosphosulfate (APS)


ADP
SO
4
2
P
i
2
ATP
ADP
AMP
SO
3
2
2e

(18-22)
Ch 18IND page - E 2/14/03, 11:10 AM 1053
1054 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
(18-25)
the only one known among chemolithotrophic bacteria.
No matter which of the two pathways of sulfite oxi-
dation is used, thiobacilli also obtain energy via elec-
tron transport. With a value of E (pH 7) of 0.454 V
[E (pH 2) = 0.158 V] for the sulfatesulfite couple
an abundance of energy may be obtained. Oxidation
of sulfite to sulfate produces hydrogen ions. Indeed,
pH 2 is optimal for the growth of Thiobacillus thiooxidans,
and the bacterium withstands 5% sulfuric acid.
322
The iron bacterium Thiobacillus ferrooxidans
obtains energy from the oxidation of Fe
2+
to Fe
3+
with
subsequent precipitation of ferric hydroxide (Eq. 18-23).
However, it has been recognized recently that a pre-
viously unknown species of Archaea is much more
important than T. ferrooxydans in catalysis of this
reaction.
324a
Since the reduction potential for the Fe(II) / Fe(III)
couple is + 0.77 V at pH 7, the energy obtainable in
this reaction is small. These bacteria always oxidize
reduced sulfur compounds, too. Especially interesting
is their oxidation of pyrite, ferrous sulfide (Eq. 18-24).
The Gibbs energy change was calculated from pub-
lished data
325
using a value of G
f
for Fe (OH)
3
of
2 FeS + 41/2 O
2
+ 5 H
2
O 2 Fe (OH)
3
+ 2 SO
4
2
+ 4 H
+
G (pH 2) = 1340 kJ
688 kJ mol
1
estimated from its solubility product.
Because sulfuric acid is generated in this reaction, a
serious water pollution problem is created by the
bacteria living in abandoned mines. Water running
out of the mines often has a pH of 2.3 or less.
326
Various invertebrates live in S
2
-containing waters.
Among these is a clam that has symbiotic sulfur-
oxidizing bacteria living in its gills. The clam tissues
apparently carry out the first step in oxidation of the
sulfide.
327
Among the animals living near sulfide-rich
thermal vents in the ocean floor are giant 1-meter-long
tube worms. Both a protective outer tube and sym-
biotic sulfide-oxidizing bacteria protect them from
toxic sulfides.
319,320
2. Anaerobic Respiration
Nitrate as an electron acceptor. The use of nitrate
as an alternative oxidant to O
2
is widespread among
bacteria. For example, E. coli can subsist anaerobically
by reducing nitrate to nitrite (Eq. 18-25).
311,328
The
respiratory (dissimilatory) nitrate reductase that
NO
3

+ 2 H
+
+ 2e

NO
2

+ H
2
O
E (pH7) = +0.421 V
catalyzes the reaction is a large three-subunit molyb-
denum-containing protein. The enzyme is present in
the plasma membrane, and electrons flow from
ubiquinone through as many as six heme and FeS
centers to the molybdenum atom.
328-329
A second
molybdoenzyme, formate dehydrogenase (discussed
in Chapter 16), appears to be closely associated with
nitrate reductase. Formate is about as strong a reduc-
ing agent as NADH (Table 6-8) and is a preferred
electron donor for the reduction of NO
3

(Eq. 18-26).
329a,b
Since cytochrome c oxidase of the electron transport
chain is bypassed, one less ATP is formed than when
O
2
is the oxidant. Nitrate is the oxidant preferred by
bacteria grown under anaerobic conditions. The
HCOO

+ NO
3

+ H
+
NO
2

+ CO
2
+ H
2
O
G (pH 7) = 165 kJ mol
1
presence of NO
3

induces the synthesis of nitrate


reductase and represses the synthesis of alternative
enzymes such as fumarate reductase,
330,331
which
reduces fumarate to succinate (see also p. 1027). On
the other hand, if NO
3

is absent and fumarate, which


can be formed from pyruvate, is present, synthesis of
fumarate reductase is induced. Although it is a much
weaker oxidant than is nitrate (E
o
= 0.031 V), fumarate
is still able to oxidize H
2
or NADH with oxidative
phosphorylation. Like the related succinate dehydro-
genase, fumarate reductase of E. coli is a flavoprotein
with associated Fe / S centers. It contains covalently
linked FAD and Fe
2
S
2
, Fe
4
S
4
, and three-Fe ironsulfur
centers.
332
In some bacteria a soluble periplasmic
cytochrome c
3
carried out the fumarate reduction
step.
332a
Trimethylamine N-oxide
330,333
or dimethyl-
sulfoxide (DMSO; Eq. 16-62)
334,335
can also serve as
alternative oxidants for anaerobic respiration using
appropriate molybdemum-containing reductases
(Chapter 16).
Reduction of nitrite: denitrification. The nitrite
formed in Eq. 18-25 is usually reduced further to ammo-
nium ions (Eq. 18-27). The reaction may not be impor-
tant to the energy metabolism of the bacteria, but it
provides NH
4
+
for biosynthesis. This six-electron
reduction is catalyzed by a hexaheme protein containing
six c-type hemes bound to a single 63-kDa polypeptide
chain.
336,337
NO
2

+ 8 H
+
+ 6 e

2 H
2
O + NH
4
+
Several types of denitrifying bacteria
315,338 340
2 Fe
2+
H
2
O
2 H
+
2 Fe
3+
6 H
2
O
6 H
+
2 Fe (OH)
3
O
2
1
/
2
(18-23)
(18-24)
(18-26)
(18-27)
Ch 18IND page - E 2/14/03, 11:10 AM 1054
1055
use either nitrate or nitrite ions as oxidants and reduce
nitrite to N
2
. A typical reaction for Micrococcus denitri-
ficans is oxidation of H
2
by nitrate (Eq. 18-28). Thioba-
cillus denitrificans, like other thiobacilli, can oxidize
5 H
2
+ 2 NO
3

+ 2 H
+
N
2
+ 6 H
2
O
G (pH 7) = 561 kJ mol
1
of nitrate reduced or
224 kJ mol
1
of H
2
oxidized
sulfur as well as H
2
S or thiosulfate using nitrate as the
oxidant (Eq. 18-29):
5 S + 6 NO
3

+ 2 H
2
O 5 SO
4
2
+ 3 N
2
+ 4 H
+
G (pH 7) = 455 kJ mol
1
of nitrate reduced or
546 kJ mol
1
of S oxidized
The reactions begin with reduction of nitrate to nitrite
(Eq. 18-25) and continue with further reduction of
nitrite to nitric oxide, NO; nitrous oxide, N
2
O; and
dinitrogen, N
2
. A probable arrangement of the four
enzymes needed for the reactions of Eq. 18-30 in Para-
coccus denitrificans is shown in Fig. 18-21. See also pp.
884, 885.
Two types of dissimilatory nitrite reductases cata-
lyze step b of Eq. 18-30. Some bacteria use a copper-
containing enzyme, which contains a type 1 (blue)
copper bound to a barrel domain of one subunit
and a type 2 copper at the catalytic center. The type 1
copper is thought to receive electrons from the small
copper-containing carrier pseudoazurin (Chapter
16).
341342b
More prevalent is cytochrome cd
1
nitrite reduc-
tase.
340,343 346
The water-soluble periplasmic enzyme
is a homodimer of ~ 60-kDa subunits, each containing
a c-type heme in a small N-terminal domain and heme
d
1
, a ferric dioxoisobacteriochlorin (Fig. 16-6). The
E. Energy from Inorganic Reactions
(18-28)
(18-29)
Figure 18-21 Organization of the nitrate reduction system
in the outer membrane of the bacterium Paracoccus dinitrifi-
cans as outlined by Blaut and Gottschalk.
315
The equations
are not balanced as shown but will be balanced if two NO
3

ions are reduced to N


2
by five molecules of NADH (see also
Eq. 18-28). Although this will also require seven protons,
about 20 additional H
+
will be pumped to provide for ATP
synthesis.
Nitrate reductase
Membrane Periplasm Cytoplasm
e

Q
2e

2e

4H
+
2H
+
Cytochrome bc
2H
+
QH
2
3H
+
2H
+
NADH
Dehydrogenase
Q QH
2
Nitrite
reductase
NADH
NAD
+
NO
3

NO
2

2H
+
H
2
O
NO reductase
N
2
O
reductase
NO
2

2H
+
H
2
O
NO
2NO
2H
+
H
2
O
N
2
O
N
2
O
2H
+
H
2
O
N
2
NO
3

/NO
2

antiporter
NO
3

NO
2

Cyt c
2e

a
c
d
NO
3
2
NO
2

2e

2H
+
H
2
O
Nitrate reductase
(Eq. 18-25)
NO
2H
H
2
O
Nitrite reductase
e
+
2 NO
N
2
O
2e

Nitric oxide reductase


N
2
Nitrous oxide reductase
2e

b
latter is present in the central channel of an eight-blad-
ed -sheet propeller
345-346g
similar to that in Fig. 15-23A.
The heme d
1
is unusual in having its Fe atom ligated by
a tyrosine hydroxyl oxygen, which may
be displaced to allow binding of NO
3

. The electron
required for the reduction is presumably transferred
from the electron transfer chain in the membrane to the
heme d, via the heme c group.
347
Cytochrome cd
1
nitrite
reductases have an unexpected second enzymatic activ-
ity. They catalyze the four-electron reduction of O
2
to
H
2
O, as does cytochrome c oxidase. However, the rate
is much slower than that of nitrite reduction.
340,348
The enzyme catalyzing the third step of Eq.
18-30 (step c), nitric oxide reductase, is an unstable
membrane-bound protein cytochrome bc complex.
349,350
(18-30)
Ch 18IND page - E 2/14/03, 11:10 AM 1055
1056 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
It has been isolated as a two-subunit protein, but
genetic evidence suggests the presence of additional
subunits.
350
The small subunit is a cytochrome c, while
the larger subunit is predicted to bind two protohemes
as well as a nonheme iron center. This protein also
shows sequence homology with cytochrome c oxidase.
It contains no copper, but it has been suggested that a
heme bnonheme Fe center similar to the heme aCu
B
center of cytochrome c oxidase may be present. It may
be the site at which the nitrogen atoms of two molecules
of NO are joined to form N
2
O.
350,351
A different kind of
NO reductase is utilized by the denitrifying fungus
Fusarium oxysporum. It is a cytochrome P450 but with
an unusually low redox potential ( 0.307 V). This
cytochrome P450
nor
does not react with O
2
(as in Eq.
18-57) but binds NO to its heme Fe
3+
, reduces the com-
plex with two electrons from NADH, then reacts with
a second molecule of NO to give N
2
O and H
2
O.
352
Reduction of N
2
O to N
2
by bacteria (Eq. 18-30, step
d) is catalyzed by the copper-containing nitrous oxide
reductase. The purple enzyme is a dimer of 66-kDa
subunits, each containing four atoms of Cu.
353
It has
spectroscopic properties similar to those of cytochome
c oxidase and a dinuclear copperthiolate center simi-
lar to that of Cu
A
in cytochrome c oxidase (p. 1030).
The nature of the active site is uncertain.
354
Sulfate-reducing and sulfur-reducing bacteria.
A few obligate anaerobes obtain energy by using
sulfate ion as an oxidant.
355356a
For example, Desulfo-
vibrio desulfuricans catalyzes a rapid oxidation of H
2
with reduction of sulfate to H
2
S (Eq. 18-31).
4 H
2
+ SO
4
2
+ 2 H
+
H
2
S + 4 H
2
O
G (pH 7) = 154 kJ mol
1
of sulfate reduced
While this may seem like an esoteric biological process,
the reaction is quantitatively significant. For example,
it has been estimated that within the Great Salt Lake
basin bacteria release sulfur as H
2
S in an amount of
10
4
metric tons (10
7
kg) per year.
357
The reduction potential for sulfate is extremely low
(E, pH 7 = 0.454 V), and organisms are not known
to reduce it directly to sulfite. Rather, a molecule of
ATP is utilized to form adenosine 5'-phosphosulfate
(APS) through the action of ATP sulfurylase
(ATP:sulfate adenylyltransferase, Eq. 17-38).
358,359
APS is then reduced by cytochrome c
3
(Eq. 18-32,
step b). The 13-kDa low-potential (E, pH 7 = 0.21 V)
cytochrome c
3
contains four heme groups (Figure 16-8C)
and is found in high concentration in sulfate-reducing
bacteria.
360,361
Some of these bacteria have larger poly-
heme cytochromes c.
361a
For example, Desulfovibrio
vulgaris forms a 514-residue protein carrying 16 hemes
organized as four cytochrome c
3
-like domains.
362
Each
heme in cytochrome c
3
has a distinct redox potential
within the range 0.20 to 0.40 V.
361 363
APS is reduced (Eq. 18-32, step b) by APS reductase,
a 220-kDa ironsulfur protein containing FAD and
several Fe S clusters. An intermediate in the reaction
may be the adduct of sulfite with FAD, which may
be formed as in Eq. 18-32. The initial step in this
hypothetical mechanism is displacement on sulfur
by a strong nucleophile generated by transfer of elec-
trons from reduced ferredoxin to cytochrome c
3
to the
flavin.
364
Bisulfite produced according to Eq. 18-32 is reduced
further by a sulfite reductase, which is thought to
receive electrons from flavodoxin, cyt c
3
, and a hydro-
genase. ATP synthesis is coupled to the reduction.
Sulfite reductases generally contain both siroheme and
Fe
4
S
4
clusters (Fig. 16-19). They appear to be able to
carry out the 6-electron reduction to sulfide without
accumulation of intermediates.
365,366
However, in
contrast to the assimilatory sulfite reductases present
in many organisms, the dissimilatory nitrite reductases
of sulfur-reducing bacteria may also release some
thiosulfate S
2
O
3
2
.
367
A possible role of menaquinone
(vitamin K
2
), present in large amounts in Desulfovibrio,
has been suggested.
311
Although Desulfovibrio can
obtain their energy from Eq. 18-31, they are not true
autotrophs and must utilize compounds such as acetate
together with CO
2
as a carbon source.
(18-31)
N
N
NH
N
R
O
O
Adenosine O P
O
O
O S
O
O
O

H
+
N
N
NH
N
R
O
O
H
S
O
O O

a
b
FAD
2e

SO
4
2
Eq. 17-38
ATP
2 P
i
Adenosine 5-phosphosulfate (APS)
or adenylyl sulfate
APS
reductase
H
+
HSO
3

S
S
Cyt c
3
(18-32)
Ch 18IND page - E 2/14/03, 11:10 AM 1056
1057
Some thermophilic archaeobacteria are able to live
with CO
2
as their sole source of carbon and reduction
of elemental sulfur with H
2
(Eq. 18-33) as their sole
source of energy.
368,369
H
2
+ S
0
H
2
S
G = 27.4 kJ mol
1
This is remarkable in view of the small standard Gibbs
energy decrease. Some species of the archaeobacteri-
umSulfolobus are able either to live aerobically oxidiz-
ing sulfide to sulfate with O
2
(Eq. 18-22) or to live
anaerobically using reduction of sulfur by Eq. 18-33 as
their source of energy.
369
The sulfate-reducing bacterium Desulfovibrio sulfo-
dismutans carries out what can be described as inor-
ganic fermentations which combine the oxidation of
compounds such as sulfite or thiosulfate (as observed
for sulfur-oxidizing bacteria; Eq. 18-22), with reduction
of the same compounds (Eq. 18-34).
370,370a
Dismutation
of S
2
O
3
2
plus H
2
O to form SO
4
2
and H
2
S also occurs
but with a less negative Gibbs energy change.
4 SO
3
2
+ H
+
3 SO
4
2
+ HS

G (pH 7) = 185 kJ/mol


A strain of Pseudomonas obtains all of its energy by
reducing sulfate using phosphite, which is oxidized
to phosphate.
370b
Methane bacteria. The methane-producing
bacteria (Chapter 15) are also classified as chemi-
autotrophic organisms. While they can utilize sub-
stances such as methanol and acetic acid, they can
also reduce CO
2
to methane and water using H
2
(Fig.
15-22). The electron transport is from hydrogenase,
perhaps through ferredoxin to formate dehydrogen-
ase and via the deazaflavin F
420
and NADP
+
to the
methanopterin-dependent dehydrogenases that carry
out the stepwise reduction of formate to methyl
groups (Fig. 16-28). Generation of ATP probably
involves proton pumps, perhaps in internal coupling
membranes .
315,371
F. Oxygenases and Hydroxylases
For many years the idea of dehydrogenation
dominated thinking about biological oxidation.
Many scientists assumed that the oxygen found in
organic substances always came from water, e.g.,
by addition of water to a double bond followed by
dehydrogenation of the resulting alcohol. Neverthe-
less, it was observed that small amounts of O
2
were
essential, even to anaerobically growing cells.
372
In 1955, Hayaishi and Mason independently demon-
strated that
18
O was sometimes incorporated into
(18-33)
(18-34)
organic compounds directly from
18
O
2
as in Eq. 18-35.
Today a bewildering variety of oxygenases are
known to function in forming such essential metabo-
lites as sterols, prostaglandins, and active derivatives
of vitamin D. Oxygenases are also needed in the
catabolism of many substances, often acting on non-
polar groups that cannot be attacked readily by other
types of enzyme.
372
Oxygenases are classified either as dioxygenases
or as monooxygenases. The monooxygenases are
also called mixed function oxidases or hydroxylases.
Dioxygenases catalyze incorporation of two atoms of
oxygen as in Eq. 18-35, but monooxygenases incorpo-
rate only one atom. The other oxygen atom from the
O
2
is converted to water. A typical monooxygenase-
catalyzed reaction is the hydroxylation of an alkane
to an alcohol (Eq. 18-36).
A characteristic of the monooxygenases is that an
additional reduced substrate, a cosubstrate (BH
2
in
Eq. 18-36), is usually required to reduce the second
atom of the O
2
molecule to H
2
O.
Since O
2
exists in a triplet state with two un-
paired electrons, it reacts rapidly only with transition
metal ions or with organic radicals (Chapter 16). For
this reason, most oxygenases contain a transition metal
ion, usually of iron or copper, or contain a cofactor,
such as FAD, that can easily form a radical or act on a
cosubstrate or substrate to form a free radical.
1. Dioxygenases
Among the best known of the oxygenases that
incorporate both atoms of O
2
into the product are
those that participate in the biological degradation
of aromatic compounds by cleaving double bonds at
positions between two OH groups as in Eq. 18-35 or
adjacent to one OH group of an ortho or para hydroxyl
pair.
373
A much studied example is protocatechuate
3,4-dioxygenase,
373 375
which cleaves its substrate
F. Oxygenases and Hydroxylases
OH
OH
Catechol
COOH
COOH
+
18
O
2
Catechol
1,2- dioxygenase
Cis,cis-muconic acid
18
18
(18-35)
H
3
C (CH
2
)
n
CH
3
H
2
O O
2
BH
2
B
H
3
C (CH
2
)
n
CH
2
OH
(cosubstrate) (18-36)
Ch 18IND page - E 2/14/03, 11:10 AM 1057
1058 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
between the two OH groups (intradiol cleavage) as in
Eq. 18-35. A different enzyme, protocatechuate 4,5-
dioxygenase,
357
cleaves the same substrate next to just
one of the two OH groups (extradiol cleavage; Eq. 18-37)
to form the aldehyde -hydroxy--carboxymuconic
semialdehyde. Another extradiol cleaving enzyme,
protocatechuate 2,3-dioxygenase, acts on the same
substrate. Many other dioxygenases attack related
substrates.
376 380
Intradiol-cleaving enzymes are usually
iron-tyrosinate proteins (Chapter 16) in which the
iron is present in the Fe(III) oxidation state and remains
in this state throughout the catalytic cycle.
375
The
enzymes usually have two subunits and no organic
prosthetic groups. For example, a protocatechuate
3,4-dioxygenase from Pseudomonas has the composi-
tion (Fe)
12
with subunit masses of 23 () and 26.5
() kDa. The iron is held in the active site cleft between
the and subunits by Tyr 408, Tyr 447, His 460, and
His 462 of the subunit and a water molecule.
375
These enzymes and many other oxygenases probably
assist the substrate in forming radicals that can react
with O
2
to form organic peroxides. Some plausible
intermediate species are pictured in Fig. 18-22. The
reactions are depicted as occurring in two-electron
steps. However, O
2
is a diradical, and it is likely that
the Fe
3+
, which is initially coordinated to both pheno-
late groups of the ionized substrate, assists in forming
an organic free radical that reacts with O
2
.
Extradiol dioxygenases have single Fe
2+
ions in
their active sites. The O
2
probably binds to the Fe
2+
and
may be converted transiently to an Fe
3+
-superoxide
complex which adds to the substrate. Some extradiol
dioxygenases require an Fe
2
S
2
ferredoxin to reduce
any Fe
3+
-enzyme that is formed as a side reaction back
to the Fe
2+
state.
381
Possible intermediates are given in
Fig. 18-22 (left side) with two-electron steps used to
save space and to avoid giving uncertain details about
free radical intermediates. Formation of the organic
radical is facilitated by the iron atom, which may be
coordinated initially to both phenolate groups of the
ionized substrate. The peroxide intermediates, for both
types of dioxygenases, may react and be converted
to various final products by several mechanisms.
382
Tryptophan dioxygenase (indoleamine 2,3-
dioxygenase)
383
is a heme protein which catalyzes the
reaction of Eq. 18-38. The oxygen atoms designated by
COO
OH
OH
C
OOC
COO
OH

3
4
O
5
Protocatechuate
4,5- dioxygenase

O
2
2
(18-37)
Figure 18-22 Some possible intermediates in the action of
extradiol (left) and intradiol (right) aromatic dioxygenases.
Although the steps depict the flow of pairs of electrons during
the formation and reaction of peroxide intermediates, the
mechanisms probably involve free radicals whose formation
is initiated by O
2
. The asterisks show how two atoms of
labeled oxygen can be incorporated into final products.
After Ohlendorf et al.
374
C
O O
R
O
Fe
O
C
O O
R
O
Fe
O
O
O
C
O O
R
O
Fe
O
O
O
C
O O
O
Fe
O
O
R
O
C
O O
O
O
R
C
O
O
C
O O

R
2

O
Fe
O
1
3
4

O
O
2+
*

C
O O
R

O
Fe
O
2+
H

O
O

3+
O
C
O O
*
*
*
O
*
3+

R
*
O
*

H H
*

C
C
O O
*
O
C
R
*

O
*

O
O
*
*
*
*
Fe

Catechol
derivative
Extradiol
cleavage
Fe
2+
Intradiol
cleavage
Fe
3+
2+ 3+
Ch 18IND page - E 2/14/03, 11:10 AM 1058
1059
the asterisks are derived from O
2
. Again, the first step
is probably the formation of a complex between Fe(II)
and O
2
, but tryptophan must also be present before
this can occur. At 5C the enzyme, tryptophan, and O
2
combine to give an altered spectrum reminiscent of
that of compound III of peroxidase (Fig. 16-14). This
oxygenated complex may, perhaps, then be converted
to a complex of Fe(II) and superoxide ion.
There is much evidence, including inhibition by
superoxide dismutase and stimulation by added potas-
sium superoxide,
384
that the superoxide anion radical
is the species that attacks the substrate (Eq. 18-39). In
this reaction one electron is returned to the Fe(III) form
of the enzyme to regenerate the original Fe(II) form.
Subsequent reaction of the hydroperoxide anion would
give the observed products.
Some dioxygenases require a cosubstrate. For
example, phthalate dioxygenase
385
converts phthalate
to a cis-dihydroxy derivative with NADH as the co-
substrate (Eq. 18-40). Similar double hydroxylation
reactions catalyzed by soil bacteria are known for ben-
zene, benzoate,
386
toluene, naphthalene, and several
other aromatic compounds.
386a
The formation of the
cis-glycols is usually followed by dehydrogenation or
oxidative decarboxylation by NAD
+
to give a catechol,
whose ring is then opened by another dioxygenase
reaction (Chapter 25). An elimination of Cl

follows
dioxygenase action on p-chlorophenylacetate and
produces 3,4-dihydroxyphenylacetate as a product.
Pthalate dioxygenases consist of two subunits. The
50-kDa dioxygenase subunits receive electrons from
reductase subunits that contain a Rieske-type Fe S
centers and bound FMN.
387
The dioxygenase also
contains an Fe
2
S
2
center, and electrons flow from
NADH to FMN and through the two Fe S centers to
the Fe
2+
of the active site.
387388
Lipoxygenases catalyze oxidation of polyunsatu-
rated fatty acids in plant lipids. Within animal tissues
the lipoxygenase-catalyzed reaction of arachidonic acid
with O
2
is the first step in formation of leukotrienes
and other mediators of inflammation. These reactions
are discussed in Chapter 21.
2. Monooxygenases
Two classes of monooxygenases are known. Those
requiring a cosubstrate (BH
2
of Eq. 18-36) in addition
to the substrate to be hydroxylated are known as
external monooxygenases. In the other group, the
internal monooxygenases, some portion of the sub-
strate being hydroxylated also serves as the cosubstrate.
Many internal monooxygenases contain flavin cofactors
and are devoid of metal ions.
Flavin-containing monooxygenases. One group
of flavin-dependent monooxygenases form H
2
O
2
by
reaction of O
2
with the reduced flavin and use the H
2
O
2
to hydroxylate 2-oxoacids. An example is lactate
monooxygenase, which apparently dehydrogenates
lactate to pyruvate and then oxidatively decarboxylates
the pyruvate to acetate with H
2
O
2
(Eq. 15-36). One
atom of oxygen from O
2
is incorporated into the acetate
formed.
389,390
In a similar manner, the FAD-containing
bacterial lysine monooxygenase probably catalyzes
the sequence of reactions shown in Eq. 18-41.
391
When
native lysine monooxygenase was treated with sulf-
hydryl-blocking reagents the resulting modified enzyme
produced a 2-oxoacid, ammonia, and H
2
O
2
, just the
products predicted from the hydrolytic decomposition
of the bracketed intermediate of Eq. 18-41. Similar bacte-
rial enzymes act on tryptophan and phenylalanine.
392
NADPH can serve as a cosubstrate of flavoprotein
monooxygenase by first reducing the flavin, after
which the reduced flavin can react with O
2
to generate
the hydroxylating reagent.
393
An example is the bacte-
rial 4-hydroxybenzoate hydroxylase which forms
3,4-dihydroxybenzoate.
394
The 43-kDa protein consists
of three domains, the large FAD-binding domain being
folded in nearly the same way as that of glutathione
reductase (Fig. 15-10). The 4-hydroxybenzoate binds
F. Oxygenases and Hydroxylases
N
COO
H H
3
N
COO
O
N C
O
H
H
3
N
H
+

*O
2

H
H
*
*
+
Tryptophan Formylkynurenine
(18-38)
N
H
R
O
2
N
R

O
O
one electron
to Fe(III)

Hydroperoxide anion
(18-39)
COO
COO

COO
COO
O
2
NADH

HO
HO

NAD
+
+ H
+
(18-40)
Ch 18IND page - E 2/14/03, 11:10 AM 1059
1060 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
first into a deep cleft below the N-5 edge of the isoallox-
azine ring of the FAD; then the NADH binds. Spectro-
scopic studies have shown the existence of at least three
intermediates. The first of these has been identified as
the 4a-peroxide whose formation (Eq. 15-31) is discussed
in Chapter 15. The third intermediate is the correspond-
ing 4a-hydroxyl compound. The substrate hydroxyla-
tion must occur in a reaction with the flavin peroxide,
presumably with the phenolate anion form of the
substrate (Eq. 18-42).
395
The initial hydroxylation
product is tautomerized to form the product 3,4-
dihydroxybenzoate.
N
N
NH
N
R
O
O
O
OH
O C
O
O
H
4a
H
+

O C
O
O

H
OH
H
+ 3
4
Substrate
b
N
O
O
H
H
a
EflavinS
EflavinH
2
S
NADPH + H
+
NADP
+
O
2
Eq. 15-31
E-flavin
H
2
O
3,4-Dihydroxybenzoate
S = substrate
(18-42)
According to this mechanism, one of the two
oxygen atoms in the hydroperoxide reacts with the
aromatic substrate, perhaps as OH
+
or as a superoxide
radical. A variety of mechanisms for activating the
flavin peroxide to give a more potent hydroxylating
reagent have been proposed. These include opening of
the central ring of the flavin to give a carbonyl oxide
intermediate which could transfer an oxygen atom to
the substrate,
396
elimination of H
2
O to form an oxazir-
idine,
397
or rearrangment to a nitroxyl radical.
398
Any of these might be an active electrophilic hydroxy-
lating reagent. However, X-ray structural studies
suggest that conformational changes isolate the sub-
strateFAD-enzyme complex from the medium stabi-
lizing the 4a peroxide via hydrogen bonding
399400
in
close proximity to the substrate. Reaction could occur
by the simple mechanism of Eq. 18-42, a mechanism
also supported by
19
F NMR studies with fluorinated
substrate analogs
401
and other investigations.
401a,b
Related flavin hydroxylases act at nucleophilic
positions on a variety of molecules
393,402
including
phenol,
403
salicylate,
404
anthranilate,
405
p-cresol,
406
4-
hydroxyphenylacetate,
407,408
and 4-aminobenzoate.
409
Various microsomal flavin hydroxylases are also
known.
410
Flavin peroxide intermediates are also
able to hydroxylate some electrophiles.
411
For exam-
ple, the bacterial cyclohexanone oxygenase catalyzes
O
N
NH
N
R
O
O
O
N
N
NH
N
R
O
O
N
N
NH
N
R
O
OH O
Carbonyl oxide
+

An oxaziridine
A nitroxyl radical
O
NH
2
C
R COO
H H
3
N

+
R
C
NH
COO

R C
NH
2
O
*
F
FH
2
O
2
CO
2
, H
2
O
H
2
O
2
*
(18-41)
Ch 18IND page - E 2/14/03, 11:10 AM 1060
1061
the ketone to lactone conversion of Eq. 18-43.
411a
The
mechanism presumably involves the nucleophilic attack
of the flavin hydroperoxide on the carbonyl group of the
substrate followed by rearrangement. This parallels
the BaeyerVilliger rearrangement that results from
treatment of ketones with peracids.
393
Cyclohexanone
oxygenase also catalyzes a variety of other reactions,
412
including conversion of sulfides to sulfoxides.
Reduced pteridines as cosubstrates. A dihydro
form of biopterin (Fig. 15-17) serves as a cosubstrate,
that is reduced by NADPH (Eq. 18-44) in hydroxylases
that act on phenylalanine, tyrosine, and tryptophan.
F. Oxygenases and Hydroxylases
N
O O
O
O
N
O O
H
O
O

4a
N
O O
Flavin
hydroperoxide
+
H
H
4a
H
4a
O
O

H
Flavin + NADPH + H
+
O
2
(18-43)
N
N
NH
N NH
O
H
H
R
H
N
N
NH
N NH
O
H
H
R
H
5
8
7
H
3
C
HO
H HO
H
6
Quinonoid dihydrobiopterin
H
H
H
Tetrahydrobiopterin
R =
NADH
NAD
+
H
+
Tyrosine
H
2
O
Phenylalanine
O
2
The tetrahydrobiopterin formed in this reaction is
similar in structure to a reduced flavin. The mechanism
of its interaction with O
2
could reasonably be the same
as that of 4-hydroxybenzoate hydroxylase. However,
phenylalanine hydroxylase, which catalyzes the for-
mation of tyrosine (Eq. 18-45), a dimer of 451-residue
subunits, contains one Fe per subunit,
413 415a
whereas
flavin monooxygenases are devoid of iron. Tyrosine
hydroxylase
416 419a
and tryptophan hydroxylase
420
have very similar properties. All three enzymes con-
tain regulatory, catalytic, and tetramerization domains
as well as a common Fe-binding motif in their active
sites.
413,421,421a
The role of the iron atom in these enzymes must
be to accept an oxygen atom from the flavin peroxide,
perhaps forming a reactive ferryl ion and transferring
the oxygen atom to the substrate, e.g., as do cytochromes
P450 (see Eq. 18-57). The 4a-hydroxytetrahydrobiop-
terin, expected as an intermediate if the mechanism
parallels that of Eq. 18-42, has been identified by its
ultraviolet absorption spectrum.
422
A ring-opened
intermediate has also been ruled out for phenylalanine
hydroxylase.
423
However, the 4a OH adduct has been
observed by
13
C-NMR spectroscopy. Its absolute config-
uration is 4a(S) and the observation of an
18
O-induced
shift in the
13
C resonance of the 4a-carbon atom
424
confirms the origin of this oxygen from
18
O
2
(see Eq.
18-42). A stimulator protein needed for rapid reac-
tion of phenylalanine hydroxylase has been identified
as a 4a-carbinolamine dehydratase (Eq. 18-46).
425426
This protein also has an unexpected function as part of
a complex with transcription factor HNF1 which is
found in nuclei of liver cells.
425a,426
Dihydrobiopterin can exist as a number of isomers.
The quinonoid form shown in Eqs. 18-44 and 18-46 is (18-44)
H
NH
3
COO
H
NH
3
COO

3
H OH
3
H
+ +
O
2
2[H]
H
2
O
(18-45)
N
C
N
NH
N
H
R
O
NH
O
N
N
NH
N
H
R
O
NH
H
H
2
O
H
Dehydratase
4a
(18-46)
Ch 18IND page - E 2/14/03, 11:10 AM 1061
1062 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
a tautomer of 7,8-dihydrobiopterin, the form gener-
ated by dihydrofolate reductase (Chapter 15). A pyri-
dine nucleotide-dependent dihydropteridine reduc-
tase
427 429
catalyzes the left-hand reaction of Eq. 18-44.
The hereditary absence of phenylalanine hydroxyl-
ase, which is found principally in the liver, is the cause
of the biochemical defect phenylketonuria (Chapter
25, Section B).
430,430a
Especially important in the me-
tabolism of the brain are tyrosine hydroxylase, which
converts tyrosine to 3,4-dihydroxyphenylalanine, the
rate-limiting step in biosynthesis of the catechola-
mines (Chapter 25), and tryptophan hydroxylase,
which catalyzes formation of 5-hydroxytryptophan,
the first step in synthesis of the neurotransmitter 5-
hydroxytryptamine (Chapter 25). All three of the
pterin-dependent hydroxylases are under complex
regulatory control.
431,432
For example, tyrosine hydrox-
ylase is acted on by at least four kinases with phosphor-
ylation occurring at several sites.
431,433,433a
The kinases
are responsive to nerve growth factor and epidermal
growth factor,
434
cAMP,
435
Ca
2+
+ calmodulin, and Ca
2+
+ phospholipid (protein kinase C).
436
The hydroxylase
is inhibited by its endproducts, the catecholamines,
435
and its activity is also affected by the availability of
tetrahydrobiopterin.
436
Hydroxylation-induced migration. A general
result of enzymatic hydroxylation of aromatic com-
pounds is the intramolecular migration of a hydrogen
atom or of a substituent atom or group as is shown
for the
3
H atom in Eq. 18-45.
437
Dubbed the NIH shift
(because the workers discovering it were in a National
Institutes of Health laboratory), the migration tells us
something about possible mechanisms of hydroxyla-
tion. In Eq. 18-45 a tritium atom has shifted in response
to the entering of the hydroxyl group. The migration
can be visualized as resulting from electrophilic attack
on the aromatic system, e.g., by an oxygen atom from
Fe(N) = O or by OH
+
(Eq. 18-47).
Such an attack could lead in step a either to an
epoxide (arene oxide) or directly to a carbocation as
shown in Eq. 18-47. Arene oxides can be converted,
via the carbocation step b, to end products in which
the NIH shift has occurred.
438
The fact that phenylala-
nine hydroxylase also catalyzes the conversion of the
special substrate shown in Eq. 18-48 to a stable epoxide,
which cannot readily undergo ring opening, also
supports this mechanism.
Operation of the NIH shift can cause migration of
a large substituent as is illustrated by the hydroxylation
of 4-hydroxyphenylpyruvate (Eq. 18-49), a key step
in the catabolism of tyrosine (Chapter 25). Human
4-hydroxyphenylpyruvate dioxygenase is a dimer of
43-kDa subunits.
439
A similar enzyme from Pseudo-
monas is a 150-kDa tetrameric iron-tyrosinate protein,
which must be maintained in the reduced Fe(II) state
for catalytic activity.
440
Although this enzyme is a
dioxygenase, it is probably related in its mechanism
of action to the 2-oxoglutarate-dependent monooxy-
genases discussed in the next section (Eqs. 18-51, 18-52).
It probably uses the oxoacid side chain of the substrate
H
3
H
O
H
H
O
3
H
H
O
3
H
H
3
H
OH
Arene oxide
+
a
b
c
d
H
+
[HO ]
+
(18-47)
H
NH
3
COO
H
NH
3
COO
+

O
+
O
2
(18-48)
OH
O
COO

OH
HO
O
2
COO
CO
2
*
*
*
Attack here by a
dioxygenase opens ring
Homogentisate
p-Hydroxyphenylpyruvate
4
1
(18-49)
to generate a reactive oxygen intermediate such as
Fe(IV) = O by the decarboxylative mechanism of Eqs.
18-50 and 18-51. The iron-bound oxygen attacks C1 of
Ch 18IND page - E 2/14/03, 11:10 AM 1062
1063
the aromatic ring, the electron-donating p-hydroxyl
group assisting. This generates a hydroxylated carbo-
cation of the type shown in Eq. 18-47 in which the
whole two-carbon side chain undergoes the NIH shift.
2-Oxoglutarate as a decarboxylating cosub-
strate. Several oxygenases accept hydrogen atoms
from 2-oxoglutarate, which is decarboxylated in the
process to form succinate. Among these are enzymes
catalyzing hydroxylation of residues of proline in both
the 3- and 4-positions (Eq. 8-6)
441 444
and of lysine in
the 5-position (Eq. 8-7)
445,446
in the collagen precursor
procollagen. The hydroxylation of prolyl residues
also takes place within the cell walls of plants.
447
Similar enzymes hydroxylate the -carbon of aspartyl
or asparaginyl side chains in EGF domains (Table 7-3)
of proteins.
441
Thymine
448
and taurine
449,449a
are
acted on by related dioxygenases. A bacterial
oxygenase initiates the degradation of the herbicide
2,4-dichlorophenoxyacetic acid (2,4-D) using another
2-oxoglutarate-dependent hydroxylase.
450,451
In
the human body a similar enzyme hydroxylates
-butyrobetaine to form carnitine (Eq. 18-50).
452
All of
these enzymes contain iron and require ascorbate,
whose function is apparently to prevent the oxidation
of the iron to the Fe(III) state.
When
18
O
2
is used for the hydroxylation of -
butyrobetaine (Eq. 18-51), one atom of
18
O is found in
the carnitine and one in succinate. The reaction is
stereospecific and occurs with retention of configura-
tion at C-3, the pro-R hydrogen being replaced by OH
while the pro-S hydrogen stays.
453
Under some condi-
tions these enzymes decarboxylate 2-oxoglutarate in
the absence of a hydroxylatable substrate, the iron
being oxidized to Fe
3+
and ascorbate being consumed
stoichiometrically.
454
A plausible mechanism (Eq. 18-51)
involves formation of an Fe(II)O
2
complex, conversion
to Fe(III)
+ .
O
2

, and addition of the superoxide ion to


2-oxoglutarate to form an adduct.
451,455
Decarboxyla-
tion of this adduct could generate the oxidizing reagent,
perhaps Fe(IV) =O. In the absence of substrate S the
ferryl iron could be reconverted to Fe(II) by a suitable
reductant such as ascorbate. In the absence of ascorbate
the Fe(IV) might be reduced to a catalytically inert
Fe(III) form.
An unusual oxygenase with a single Fe
2+
ion in its
active site closes the four-membered ring in the bio-
synthesis of penicillins (Eq. 18-52). It transfers four
N(CH
3
)
3
COO
H
H
N(CH
3
)
3
COO
H
O

+ +
H

*
-Butyrobetaine l-Carnitine
2-Oxoglutarate
Succinate
*
O
2
CO
2
*
(18-50)
F. Oxygenases and Hydroxylases
O O
COO
O
Fe(II)
2+

b
c
CO
2
O O
COO
O
Fe(III)
2+

O
O

O O
COO
O
Fe(III)
3+

O
O

C
COO
O O

O
2
a
Fe (II)
Fe (IV) O
SO S
substrate
2-Oxoglutarate
2+
H
2
O

2e
+
+ 2 H
2+
3+

(18-51)
N
SH
N
H
O
O COO
H
3
N
H H
H
CH
3
OOC
CH
3
H
N
O COO
H
3
N
H

+
l--(-Aminoadipoyl)-l-cysteinyl-d-valine
H

H
H
Isopenicillin N synthase
N
O
S
CH
3
CH
3
COO
+
O
2
2 H
2
O

Isopenicillin N (18-52)
Ch 18IND page - E 2/14/03, 11:10 AM 1063
1064 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
hydrogen atoms from its dipeptide substrate to form
two molecules of water and the product isopenicillin
N.
456,457
Sequence comparison revealed several regions
including the Fe-binding sites that are homologous
with the oxoacid-dependent oxygenases. A postulated
mechanism for isopenicillin N synthase involves
formation of an Fe
3+
superoxide anion complex as in
Eq. 18-51. However, instead of attack on an oxoacid as
in Eq. 18-51, it removes a hydrogen from the substrate
to initiate the reaction sequence.
457
Other related oxy-
genases include aminocyclopropane-1-carboxylate
oxidase (Eq. 24-35); deacetoxycephalosporin C
synthase,
457a
an enzyme that converts penicillins to
cephalosporins (Box 20-G); and clavaminate syn-
thase,
458,459
an enzyme needed for synthesis of the
-lactamase inhibitor clavulanic acid, and clavaminate
synthase.
458,459
This 2-oxoglutarate-dependent oxy-
genase catalyzes three separate reactions in the syn-
thesis of the clinically important -lactamase inhibitor
clavulanic acid. The first step is similar to that in Eq.
18-50. The second is an oxidative cyclization and the
third a desaturation reaction.
Copper-containing hydroxylases. Many Fe(II)-
containing hydroxylases require a reducing agent to
maintain the iron in the reduced state, and ascorbate
is often especially effective. In addition, ascorbate is
apparently a true cosubstrate for the copper-containing
dopamine -hydroxylase, an enzyme required in
the synthesis of noradrenaline according to Eq. 18-53.
This reaction takes place in neurons of the brain and
in the adrenal gland, a tissue long known as especially
rich in ascorbic acid. The reaction requires two mole-
cules of ascorbate, which are converted in two one-
electron steps to semidehydroascorbate.
460
Both
the structure of this free radical and that of the fully
oxidized form of vitamin C, dehydroascorbic acid,
are shown in Box 18-D. Dopamine -hydroxylase is
a 290-kDa tetramer, consisting of a pair of identical
disulfide-crosslinked homodimers, which contains
two Cu ions per subunit.
461
A similar copper-dependent hydroxylase consti-
tutes the N-terminal domain of the peptidylglycine
-amidating enzyme (Eq. 10-11). This bifunctional
enzyme hydroxylates C-terminal glycines in a group
of neuropeptide hormones and other secreted pep-
tides. The second functional domain of the enzyme
cleaves the hydroxylated glycine to form a C-terminal
amide group and glyoxylate.
462 464b
The three-
dimensional structure of a 314-residue catalytic core
of the hydroxylase domain is known.
463
Because of
similar sequences and other properties, the structures
of this enzyme and of dopamine -hydroxylase are
thought to be similar. The hydroxylase domain of the
-amidating enzyme is folded into two eight-stranded
antiparallel jelly-roll motifs, each of which binds one
of the two copper ions. Both coppers can exist in a
Cu(II) state and be reduced by ascorbate to Cu(I).
One Cu (Cu
A
) is held by three imidazole groups and
is thought to be the site of interaction with ascorbate.
The other copper, Cu
B
, which is 1.1 nm away from Cu
A
,
is held by two imidazoles. The substrate binds adja-
cent to Cu
B
.
463
The reaction cycle of these enzymes begins with
reduction of both coppers from Cu(II) to Cu(I) (Eq.
18-54, step a). Both O
2
and substrate bind (steps b and
c, but not necessarily in this order). The O
2
bound to
Cu
B
is reduced to a peroxide anion that remains bound
to Cu
B
. Both Cu
A
and Cu
B
donate one electron, both
being oxidized to Cu(II). These changes are also in-
cluded in step c of Eq. 18-54. One proposal is that the
resulting peroxide is cleaved homolytically while
removing the pro-S hydrogen of the glycyl residue.
HO
HO
NH
2
HO
HO
NH
2
H
OH
Dopamine
Noradrenaline
Ascorbate
Dehydroascorbate
(Box 18-D)
O
2
H
2
O
(18-53)
Cu
B
2 +
O
OH
Cu
A
2+
a
b
c
d
e
O
2
H
+
Substrate
Cu
A
+
Cu
B
+
O
2
Cu
A
+
Cu
B
+
2 Semidehydro-
ascorbate
2 Ascorbate
Peptide N
C
COO
H H
H

Cu
A
2+
Cu
B
2 +
O
Peptide N
C
COO
H H
H

H
Cu
A
2+
Cu
B
2+
Peptide N
C
COO
H H
H

HO
Substrate
(18-54)
Ch 18IND page - E 2/14/03, 11:10 AM 1064
1065
The resulting glycyl radical couples with the oxygen
radical that is bound to Cu
B
(step e).
A variety of other copper hydroxylases are known.
For example, tyrosinase, which contains a binuclear
copper center, catalyzes both hydroxylation of phenols
and aromatic amines and dehydrogenation of the
resulting catechols or o-aminophenols (Eq. 16-57). As
in hemocyanin, the O
2
is thought to be reduced to a
peroxide which bridges between the two copper atoms.
Methane-oxidizing bacteria, such as Methylococcus
capsulatus, oxidize methane to methanol to initiate its
metabolism. They do this with a copper-containing
membrane-embedded monooxygenase whose active
site is thought to contain a trinuclear copper center.
Again a bridging peroxide may be formed and may
insert an oxygen atom into the substrate.
465,466
The
same bacteria produce a soluble methane monooxy-
genase containing a binuclear iron center.
Hydroxylation with cytochrome P450. An
important family of heme-containing hydroxylases,
found in most organisms from bacteria to human
beings, are the cytochromes P450. The name comes
from the fact that in their reduced forms these enzymes
form a complex with CO that absorbs at 450 nm. In
soil bacteria cytochromes P450 attack compounds of
almost any structure. In the adrenal gland they par-
ticipate in steroid metabolism,
467,468
and in the liver
microsomal cytochromes P450 attack drugs, carcinogens,
and other xenobiotics (foreign compounds).
469 471
They convert cholesterol to bile acids
472
and convert
vitamin D,
473
prostaglandins, and many other metabo-
lites to more soluble and often biologically more active
forms. In plants cytochromes P450 participate in
hydroxylation of fatty acids at many positions.
474
They play a major role in the biosynthetic phenyl-
propanoid pathway (Fig. 25-8) and in lignin synthe-
sis.
475
More than 700 distinct isoenzyme forms have
been described.
476,476a
Cytochromes P450 are monooxygenases whose
cosubstrates, often NADH or NADPH, deliver electrons
to the active center heme via a separate flavoprotein
and often via an iron-sulfur protein as well.
476a,b
A
typical reaction (Eq. 18-55) is the 11 -hydroxylation
of a steroid, an essential step in the biosynthesis of
steroid hormones (Fig. 22-11). The hydroxyl group is
introduced without inversion of configuration. The
same enzyme converts unsaturated derivatives to
epoxides (Eq. 18-56), while other cytochromes P450
H
H
CH
3
H
HO
CH
3
11
O
2
2e

+
2H +
H
2
O
(18-55)
F. Oxygenases and Hydroxylases
epoxidize olefins.
477
Epoxide hydrolases, which act by
a mechanism related to haloalkane dehalogenase (Fig.
12-1), convert the epoxides to diols.
478
Cytochromes
P450 are able to catalyze a bewildering array of other
reactions
479 481
as well. Most of these, such as conver-
sion of amines and thioesters to N- or S- oxides, also
involve transfer of an oxygen atom to the substrate.
Others, such as the reduction of epoxides, N- oxides,
or nitro compounds, are electron-transfer reactions.
Several different cytochromes P450 are present
in mammalian livers.
470
All are bound to membranes
of the endoplasmic reticulum and are difficult to
solubilize. Biosynthesis of additional forms is in-
duced by such agents as phenobarbital,
470
3-methyl-
cholanthrene,
469
dioxin,
482
and ethanol.
483
These
substances may cause as much as a 20-fold increase
in P450 activity. Another family of cytochrome P450
enzymes is present in mitochondria.
483a
A large num-
ber of cytochrome P450 genes have been cloned and
sequenced. Although they are closely related, each
cytochrome P450 has its own gene. There are at least
ten families of known P450 genes and the total number
of these enzymes in mammals may be as high as 200.
Microorganisms, from bacteria to yeast, produce many
other cytochromes P450.
Microsomal cytochromes P450 receive electrons
from an NADPH-cytochrome P450 reductase, a
large 77-kDa protein that contains one molecule each
of FAD and FMN.
484485a
It is probably the FAD which
accepts electrons from NADPH and the FMN which
passes them on to the heme of cytochrome P450. Cyto-
chrome b
5
is also reduced by this enzyme,
486
and some
cytochromes P450 may accept one electron directly from
the flavin of the reductase and the second electron via
cytochrome b
5
. However, most bacterial and mitochon-
drial cytochromes P450 accept electrons only from
small ironsulfur proteins. Those of the adrenal gland
receive electrons from the 12-kDa adrenodoxin.
487,488
This small protein of the ferredoxin class contains one
Fe
2
S
2
cluster and is, therefore, able to transfer elec-
trons one at a time from the FAD-containing NADPH-
adrenodoxin reductase
489
to the cytochrome P450. The
camphor 5-monoxygenase from Pseudomonas putida
consists of three components: an FAD-containing re-
ductase, the Fe
2
S
2
cluster-containin putidaredoxin,
489a
and cytochrome P450
cam
.
490
Some other bacterial
CH
3
CH
3
O
2
2e

+
2H +
H
2
O
O
(18-56)
Ch 18IND page - E 2/14/03, 11:10 AM 1065
1066 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
BOX 18-D VITAMIN C: ASCORBIC ACID
Hemorrhages of skin, gums, and joints were
warnings that death was near for ancient sea voyagers
stricken with scurvy. It was recognized by the year
1700 that the disease could be prevented by eating
citrus fruit, but it was 200 years before efforts to isolate
vitamin C were made. Ascorbic acid was obtained in
crystalline form in 1927,
a e
and by 1933 the structure
had been established. Only a few vertebrates, among
them human beings, monkeys, guinea pigs, and some
fishes, require ascorbic acid in the diet; most species
are able to make it themselves. Compared to that of
other vitamins, the nutritional requirement is large.
e
Ten milligrams per day prevents scurvy, but subclinical
deficiency, as judged by fragility of small capillaries in
the skin, is present at that level of intake. Official
recommendations for vitamin C intake have ranged
from 30 to 70 mg / day. A more recent study
f
suggests
200 mg / day, a recommendation that is controversial.
g
The biological functions of vitamin C appear to
be related principally to its well-established reducing
properties and easy one-electron oxidation to a free
radical or two-electron reduction to dehydroascorbic
acid. The latter is in equilibrium with the hydrated
hemiacetal shown at the beginning of this box as well
as with other chemical species.
h j
Vitamin C is a weak
acid which also has metal complexing properties.
Ascorbate, the anion of ascorbic acid, tends to be
concentrated in certain types of animal tissues and
may reach 3 mM or more in leukocytes, in tissues
of eyes and lungs, in pituitary, adrenal, and parotid
glands,
k,l
and in gametes.
m
Uptake into vesicles of
the endoplasmic reticulum may occur via glucose
transporters.
n
Ascorbate concentrations are even
higher in plants and may exceed 10 mM in chloroplasts.
o
In animals the blood plasma ascorbate level of 20 100
M is tightly controlled.
p,q
Cells take up ascorbate but
any excess is excreted rapidly in the urine.
q
Both in
plasma and within cells most vitamin C exists as the
reduced form, ascorbate. When it is formed, the oxi-
dized dehydroascorbate is reduced back to ascorbate
or is degraded. The lactone ring is readily hydrolyzed
to 2,3-dioxogulonic acid, which can undergo decar-
boxylation and oxidative degradation, one product
being oxalate (see Fig. 20-2).
r
Tissues may also contain
smaller amounts of L-ascorbic acid 2-sulfate, a com-
pound originally discovered in brine shrimp. It is
more stable than free ascorbate and may be hydrolyzed
to ascorbate in tissues.
s
In the chromaffin cells of the adrenal glands and in
the neurons that synthesize catecholamines as neuro-
transmitters, ascorbate functions as a cosubstrate for
dopamine -hydroxylase (Eq. 18-53).
t,u
In fibroblasts it
is required by the prolyl and lysyl hydroxylases and in
hepatocytes by homogentisate dioxygenase (Eq. 18-49).
Any effect of ascorbic acid in preventing colds may be
a result of increased hydroxylation of procollagen and
an associated stimulation of procollagen secretion.
v
High levels of ascorbate in guinea pigs lead to more
rapid healing of wounds.
w
An important function
of ascorbate in the pituitary and probably in other
endocrine glands is in the -amidation of peptides (Eq.
10-11).
x,y
Together with Fe(II) and O
2
ascorbate is a
powerful nonenzymatic hydroxylating reagent for
aromatic compounds. Like hydroxylases, the reagent
attacks nucleophilic sites, e.g., converting phenylala-
nine to tyrosine. Oxygen atoms from
18
O
2
are incorpo-
rated into the hydroxylated products. While H
2
O
2
is
formed in the reaction mixture, it cannot replace ascor-
bate. The relationship of this system to biochemical
functions of ascorbate is not clear. An unusual func-
tion for vitamin C has been proposed for certain
sponges that are able to etch crystalline quartz (SiO
2
)
particles from sand or rocks.
z
Ascorbate is a major antioxidant, protecting cells
and tissues from damage by free radicals, peroxides,
and other metabolites of O
2
.
p,r,aa,bb
It is chemically
suited to react with many biologically important radi-
cals and is present in high enough concentrations to
be effective. It probably functions in cooperation with
glutathione (Box 11-B),
cc
-tocopherol (Fig. 15-24),
dd
and lipoic acid.
ee
Ascorbate can react with radicals in
one-election transfer reactions to give the monodehy-
droascorbate radical
aa
:
O
O
HO OH
H
C HOH
2
C
HO
H
1
2
2e

+ 2H
+
E' (pH7) = +0.058V
Dehydroascorbic acid
This proton dissociates with pK
a
4.2
O
O
O O
H
C HOH
2
C
HO
H
l-Ascorbic acid
O
O
H
C
HO H
OH
OH HO
O
H
2
C
O
O
O O
C HOH
2
C
HO
H

O
O
O O
C HOH
2
C
HO
H
Ch 18IND page - E 2/14/03, 11:11 AM 1066
1067
BOX 18-D (continued)
Two ascorbate radicals can react with each other in a
disproportionation reaction to give ascorbate plus
dehydroascorbate. However, most cells can reduce
the radicals more directly. In many plants this is
accomplished by NADH + H
+
using a flavoprotein
monodehydroascorbate reductase.
o
Animal cells
may also utilize NADH or may reduce dehydroascor-
bate with reduced glutathione.
cc,ff
Plant cells also
contain a very active blue copper ascorbate oxidase
(Chapter 16, Section D,5), which catalyzes the opposite
reaction, formation of dehydroascorbate.
gg
A heme
ascorbate oxidase has been purified from a fungus.
hh
Action of these enzymes initiates an oxidative degra-
dation of ascorbate, perhaps through the pathway of
Fig. 20-2.
Ascorbate can also serve as a signal. In cultured
cells, which are usually deficient in vitamin C, addition
of ascorbate causes an enhanced response to added
iron, inducing synthesis of the iron storage protein
ferritin.
ii
Ascorbate indirectly stimulates transcription
of procollagen genes
v
and decreases secretion of insulin
by the pancreas.
jj
However, since its concentration in
blood is quite constant this effect is not likely to cause
a problem for a person taking an excess of vitamin C.
Should we take extra vitamin C to protect us from
oxygen radicals and slow down aging? Linus Pauling,
who recommended an intake of 0.25 10 g / day, main-
tained that ascorbic acid also has a specific beneficial
effect in preventing or ameliorating symptoms of
the common cold.
j,kk
However, critics point out that
unrecognized hazards may exist in high doses of this
seemingly innocuous compound. Ascorbic acid has
antioxidant properties, but it also promotes the genera-
tion of free radicals in the presence of Fe(III) ions, and
it is conceivable that too much may be a bad thing.
ll
Catabolism to oxalate may promote formation of cal-
cium oxalate kidney stones. Under some conditions
products of dehydroascorbic acid breakdown may
accumulate in the lens and contribute to cataract
formation.
i,mm,nn
However, dehydroascorbate, or its
decomposition products, apparently protects low-
density lipoproteins against oxidative damage.
bb
Pauling pointed out that nonhuman primates synthe-
size within their bodies many grams of ascorbic acid
daily, and that there is little evidence for toxicity.
Paulings claim that advanced cancer patients are
benefited by very high (10 g daily) doses of vitamin C
has been controversial, and some studies have failed
to substantiate the claim.
oo,pp
a
Hughes, R. E. (1983) Trends Biochem. Sci. 8, 146147
b
Staudinger, H. J. (1978) Trends Biochem. Sci. 3, 211212
c
Szent-Gyrgyi, A. (1963) Ann. Rev. Biochem. 32, 114
d
Bradford, H. F. (1987) Trends Biochem. Sci. 12, 344347
e
Packer, L., and Fuchs, J., eds. (1997) Vitamin C in Health and Disease,
Dekker, New York
f
Levine, M., Conry-Cantilena, C., Wang, Y., Welch, R. W., Washko.PW,
Dhariwal, K. R., Park, J. B., Lazarev, A., Graumlich, J. F., King, J., and
Cantilena, L. R. (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 37043709
F. Oxygenases and Hydroxylases
g
Young, V. R. (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 1434414348
h
Hroslif, J., and Pederson, B. (1979) Acta Chem. Scand. B33, 503511
i
Counsell, J. N., and Horing, D. H., eds. (1981) Vitamin C, Applied
Sciences Publ., London
j
Burns, J. J., Rivers, J. M., and Machlin, L. J., eds. (1987) Third Confer-
ence on Vitamin C, Vol. 498, New York Academy of Sciences, New
York
k
Washko, P. W., Wang, Y., and Levine, M. (1993) J. Biol. Chem. 268,
1553115535
l
Rosen, G. M., Pou, S., Ramos, C. L., Cohen, M. S., and Britigan, B. E.
(1995) FASEB J. 9, 200209
m
Moreau, R., and Dabrowski, K. (1998) Proc. Natl. Acad. Sci. U.S.A. 95,
1027910282
n
Bnhegyi, G., Marcolongo, P., Pusks, F., Fulceri, R., Mandl, J., and
Benedetti, A. (1998) J. Biol. Chem. 273, 27582762
o
Sano, S., Miyake, C., Mikami, B., and Asada, K. (1995) J. Biol. Chem.
270, 2135421361
p
May, J. M., Qu, Z.-c, and Whitesell, R. R. (1995) Biochemistry 34,
1272112728
q
Vera, J. C., Rivas, C. I., Velsquez, F. V., Zhang, R. H., Concha, I. I.,
and Golde, D. W. (1995) J. Biol. Chem. 270, 2370623712
r
Rose, R. C., and Bode, A. M. (1993) FASEB J. 7, 1135 1142
s
Benitez, L. V., and Halver, J. E. (1982) Proc. Natl. Acad. Sci. U.S.A. 79,
54455449
t
Dhariwal, K. R., Shirvan, M., and Levine, M. (1991) J. Biol. Chem. 266,
53845387
u
Tian, G., Berry, J. A., and Klinman, J. P. (1994) Biochemistry 33, 226
234
v
Chojkier, M., Houglum, K., Solis-Herruzo, J., and Brenner, D. A.
(1989) J. Biol. Chem. 264, 1695716962
w
Harwood, R., Grant, M. E., and Jackson, D. S. (1974) Biochem. J. 142,
641651
x
Bradbury, A. F., and Smyth, D. G. (1991) Trends Biochem. Sci. 16, 112
115
y
Eipper, B. A., Milgram, S. L., Husten, E. J., Yun, H.-Y., and Mains, R.
E. (1993) Protein Sci. 2, 489497
z
Bavestrello, G., Arillo, A., Benatti, U., Cerrano, C., Cattaneo-Vietti, R.,
Cortesogno, L., Gaggero, L., Giovine, M., Tonetti, M., and Sar, M.
(1995) Nature (London) 378, 374376
aa
Kobayashi, K., Harada, Y., and Hayashi, K. (1991) Biochemistry 30,
83108315
bb
Retsky, K. L., Freeman, M. W., and Frei, B. (1993) J. Biol. Chem. 268,
13041309
cc
Ishikawa, T., Casini, A. F., and Nishikimi, M. (1998) J. Biol. Chem. 273,
2870828712
dd
May, J. M., Qu, Z.-c, and Morrow, J. D. (1996) J. Biol. Chem. 271,
1057710582
ee
Lykkesfeldt, J., Hagen, T. M., Vinarsky, V., and Ames, B. N. (1998)
FASEB J. 12, 11831189
ff
May, J. M., Cobb, C. E., Mendiratta, S., Hill, K. E., and Burk, R. F.
(1998) J. Biol. Chem. 273, 2303923045
gg
Gaspard, S., Monzani, E., Casella, L., Gullotti, M., Maritano, S., and
Marchesini, A. (1997) Biochemistry 36, 48524859
hh
Kim, Y.-R., Yu, S.-W., Lee, S.-R., Hwang, Y.-Y., and Kang, S.-O. (1996)
J. Biol. Chem. 271, 31053111
ii
Toth, I., and Bridges, K. R. (1995) J. Biol. Chem. 270, 1954019544
jj
Bergsten, P., Sanchez Moura, A., Atwater, I., and Levine, M. (1994) J.
Biol. Chem. 269, 10411045
kk
Pauling, L. (1970) Vitamin C and the Common Cold, Freeman, San
Francisco, California
11
Halliwell, B. (1999) Trends Biochem. Sci. 24, 255259
mm
Russell, P., Garland, D., Zigler, J. S., Jr., Meakin, S. O., Tsui, L.-C., and
Breitman, M. L. (1987) FASEB J. 1, 3235
nn
Nagaraj, R. H., Sell, D. R., Prabhakaram, M., Ortwerth, B. J., and
Monnier, V. M. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 1025710261
oo
Moertel, C. G., Fleming, T. R., Creagan, E. T., Rubin, J., OConnell, M.
J., and Ames, M. M. (1985) N. Engl. J. Med. 312, 142146
pp
Lee, S. H., Oe, T., and Blair, I. A. (2001) Science 292, 20832086
Ch 18IND page - E 2/14/03, 11:11 AM 1067
1068 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
cytochrome P450s, such as a soluble fatty acid hydrox-
ylase from Bacillus megaterium, have reductase do-
mains with tightly bound FMN and FAD bound to the
same polypeptide chain as is the heme.
491
All cytochromes P450 appear to have at their active
sites a molecule of heme with a thiolate anion as an
axial ligand in the fifth position (Fig. 18-23). These
relatively large heme proteins of ~ 45- to 55-kDa mass
may consist of as many as 490 residues. Only a few
three-dimensional structures are known,
490,492 494
and
among these there are significant differences. How-
ever, on the basis of a large amount of experimental
effort
487,495,496
it appears that all cytochromes P450 act by
basically similar mechanisms.
474,496a,b,497
As indicated
in Eq. 18-57, the substrate AH binds to the protein
near the heme, which must be in the Fe(III) form.
An electron delivered from the reductase then reduces
the iron to the Fe(II) state (Eq. 18-57, step b). Then O
2
combines with the iron, the initial oxygenated com-
plex formed in step c being converted to an Fe(III)-
superoxide complex (Eq. 18-57, step d). Subsequent
events are less certain.
497a
Most often a second elec-
tron is transferred in from the reductase (Eq. 18-57,
step e) to give a peroxide complex of Fe(III), which is
then converted in step f to a ferryl iron form, as in the
action of peroxidases (Fig. 16-14). This requires trans-
fer of two H
+
into the active site. The ferryl Fe(IV) = O
donates its oxygen atom to the substrate regenerating
the Fe(III) form of the heme (step g) and releasing the
product (step h).
Microsomal cytochromes P450 often form hydrogen
peroxide as a side product. This may arise directly
from the Fe O O

intermediate shown in Eq. 18-57.


Some cytochromes P450 use this reaction in reverse to
carry out hydroxylation utilizing peroxides instead of
O
2
(Eq. 18-58).
AH + ROOH AOH + ROH
Cytochromes P450 often convert drugs or other
foreign compounds to forms that are more readily
excreted.
499
However, the result is not always bene-
ficial. For example, 3-methylcholanthrene, a strong
inducer of cytochrome P450, is converted to a power-
ful carcinogen by the hydroxylation reaction.
500
See
also Box 18-E.
Other iron-containing oxygenases.
Hydroxylases with properties similar to
those of cytochrome P450 but containing
nonheme iron catalyze -oxidation of
alkanes and fatty acids in certain bacteria,
e.g., Pseudomonas oleovarans. A flavoprotein
rubredoxin reductase, is also required.
501
The methylotrophs Methylococcus and
Methylosinus hydroxylate methane using
as cosubstrate NADH or NADPH (Eq. 18-
59). A soluble complex consists of 38-kDa
reductase containing FAD and an Fe
2
S
2
CH
4
+ O
2
+ NADH + H
+

CH
3
OH + H
2
O + NAD
+
center, a small 15-kDa component, and a
245-kDa hydroxylase with an ()
2
com-
position and a three-dimensional struc-
ture
502503
similar to that of ribonucleotide
reductase (Chapter 16, Section A,9). Each
large subunit contains a diiron center
similar to that shown in Fig. 16-20C. It is
likely that O
2
binds between the two iron
atoms in the Fe(II) oxidation state and,
oxidizing both irons to Fe(III), is converted
to a bridging peroxide group as shown in
Eq. 18-60. In this intermediate, in which
the two metals are held rigidly by the
surrounding ligands including a bridging
carboxylate side chain, the OO bond may
be broken as in Eq. 18-60, steps a and b, to
generate an Fe(IV)O

radical that may


a
b c
d
e
f g
Fe (II)
Fe (IV)
O
+
h
Reductase
e

O
2
Substrate,
AH
Product,
AOH
H
2
O
H
+
O
2

H
2
O
2
2H
+
Fe (III)
+
Fe (III)
+
Fe (III)
+
e

Reductase
Fe (II)
O
O
Fe (III)
O
O
Fe (III)
O
OH
AH
AH
AH
AH
AH
AOH
AH
H
+
(18-57)
(18-58)
(18-59)
Ch 18IND page - E 2/14/03, 11:11 AM 1068
1069 F. Oxygenases and Hydroxylases
remove a hydrogen atom from the substrate (step c)
and undergo subsequent reaction steps analogous to
those in the cytochrome P450 reaction cycle.
504 506
A group of related bacterial enzymes hydroxylate
alkanes,
507
toluene,
508
phenol,
509
and other substrates.
509a
Eukaryotic fatty acid desaturases (Fig. 16-20B) belong
to the same family.
508
Some bacteria use cytochrome
P450 or other oxygenase to add an oxygen atom to an
alkene to form an epoxide. For example, propylene
may be converted to either R or S epoxypropane
which may be hydrolyzed, rearranged by a coenzyme
M-dependent reaction, and converted to acetoacetate,
which can be used as an energy source.
509a,b
Nitric oxide and NO synthases. Nitric oxide
(NO) is a reactive free radical whose formula is often
written as

NO to recognize this characteristic. How-


ever, NO is not only a toxic and sometimes dangerous
metabolite but also an important hormone with func-
tions in the circulatory system, the immune system,
and the brain.
510 512
The hormonal effects of NO are
discussed in Chapter 30, but it is appropriate here to
mention a few reactions. Nitric oxide reacts rapidly
with O
2
to form nitrite (Eq. 18-61).
4 NO + O
2
+ 2 H
2
O 4 NO
2

+ 4 H
+
It also combines very rapidly with superoxide anion
radical to form peroxynitrite (Eq. 18-62).
513
This is
another reactive oxidant which, because of its relatively
high pK
a
of 6.8, is partially protonated and able to
diffuse through phospholipids within cells.
514,515
Figure 18-23 (A) Stereoscopic
-carbon backbone model of
cytochrome P450cam showing
the locations of the heme and
of the bound camphor molecule.
(B) View in the immediate vicinity
of the thiolate ligand from Cys 357.
From Poulos et al.
498
A
B
a
b
c
H
3
C OH
Fe(IV)
O
O
Fe(IV)
Fe(III)
O
Fe(IV)
O
H
C
H
H H
Fe(III)
O
O
Fe(III)
(18-60)
(18-61)
Ch 18IND page - E 2/14/03, 11:11 AM 1069
1070 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
BOX 18-E THE TOXICITY OF ACETAMINOPHEN
Most drugs, as well as toxins and other xeno-
biotic compounds, enter the body through mem-
branes of the gastrointestinal tract, lungs, or skin.
Drugs are frequently toxic if they accumulate in the
body. They are often rather hydrophobic and are
normally converted to more polar, water-soluble
substances before elimination from the body. Two
major types of reaction take place, usually in the liver.
These are illustrated in the accompanying scheme for
acetaminophen (N-acetyl-p-aminophenol), a widely
used analgesic and antipyretic (fever relieving) non-
prescription drug sold under a variety of trade names:
(1) A large water-soluble group such as sulfate
a
or
glucuronate is transferred onto the drug by a nucleo-
philic displacement reaction (steps a and b of scheme).
(2) Oxidation, demethylation, and other alterations
are catalyzed by one or more of the nearly 300 cyto-
chrome P450 monooxygenases present in the liver
(step c). Oxidation products may be detoxified by
glutathione S-transferases, step d (see also Box 11-B).
b,c,cc
These reactions protect the body from the accu-
mulation of many compounds but in some cases can
cause serious problems. The best known of these
involves acetaminophen. Its oxidation by cytochrome
P450 2E1 or by prostaglandin H synthase
d
yields a
highly reactive quinone imine which reacts with cell
proteins.
e
Since the cytochrome P450 oxidation can
occur in two steps, a reactive intermediate radical is
also created.
c,f
At least 20 drug-labeled proteins
arising in this way have been identified.
c
Both addi-
tion of thiol groups of proteins to the quinone imine
(step e of scheme) and oxidation of protein thiols
occur.
g
Mitochondria suffer severe damage,
h
some
of which is related to induction of Ca
2+
release.
i
Acetaminophen is ordinarily safe at the recom-
mended dosages, but large amounts exhaust the
reserve of glutathione and may cause fatal liver
damage. By 1989, more than 1000 cases of accidental
or intentional (suicide) overdoses had been reported
with many deaths. Prompt oral or intravenous
administration of N-acetylcysteine over a 72-hour
period promotes synthesis of glutathione and is an
effective antidote.
j
Similar problems exist for many other drugs.
Both acetaminophen and phenacetin, its ethyl ether
derivative, may cause kidney damage after many
years of use.
k,l
Metabolism of phenacetin and several
other drugs varies among individuals. Effective
detoxification may not occur in individuals lacking
certain isoenzyme forms of cytochrome P450.
m
Use
of the anticancer drugs daunomycin (daunorubicin;
Figs. 5-22 and 5-23) and adriamycin is limited by
severe cardiac toxicity arising from free radicals
generated during oxidation of the drugs.
n
These are
only a few examples of the problems with drugs,
pesticides, plasticizers, etc.
a
Klaassen, C. D., and Boles, J. W. (1997) FASEB J. 11, 404418
b
Lee, W. M. (1995) N. Engl. J. Med. 333, 11181127
c
Qiu, Y., Benet, L. Z., and Burlingame, A. L. (1998) J. Biol. Chem.
273, 1794017953
cc
Chen, W., Shockcor, J. P., Tonge, R., Hunter, A., Gartner, C., and
Nelson, S. D. (1999) Biochemistry 38, 8159 8166
d
Potter, D. W., and Hinson, J. A. (1987) J. Biol. Chem. 262, 974980
e
Lee, S. S. T., Buters, J. T. M., Pineau, T., Fernandez-Salguero, P.,
and Gonzalez, F. J. (1996) J. Biol. Chem. 271, 1206312067
f
Rao, D. N. R., Fischer, V., and Mason, R. P. (1990) J. Biol. Chem.
265, 844847
g
Tirmenstein, M. A., and Nelson, S. D. (1990) J. Biol. Chem. 265,
30593065
h
Burcham, P. C., and Harman, A. W. (1991) J. Biol. Chem. 266,
50495054
i
Weis, M., Kass, G. E. N., Orrenius, S., and Moldus, P. (1992)
J. Biol. Chem. 267, 804809
j
Smilkstein, M. J., Knapp, G. L., Kulig, K. W., and Rumack, B. H.
(1988) N. Engl. J. Med. 319, 15571562
k
Stolley, P. D. (1991) N. Engl. J. Med. 324, 191193
l
Rocha, G. M., Michea, L. F., Peters, E. M., Kirby, M., Xu, Y.,
Ferguson, D. R., and Burg, M. B. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 53175322
m
Distlerath, L. M., Reilly, P. E. B., Martin, M. V., Davis, G. G.,
Wilkinson, G. R., and Guengerich, F. P. (1985) J. Biol. Chem. 260,
90579067
n
Davies, K. J. A., and Doroshow, J. H. (1986) J. Biol. Chem. 261,
30603067
a
b
c
d
e
N
C
CH
3
O
O
Acetaminophen
(N-Acetyl-p-aminophenol)
Sulfotransferase
PAPS (Eq. 17-38)
Quinone imine
UDP-glucuronosyl-
transferase
Cytochrome
P450 2E1
Glutathione
Glutathione
transferase
Cell
proteins
Mercapturic acid
HN
C
CH
3
OH
O HN
C
CH
3
O
O
SO
3
HN
C
CH
3
O
O
O
OH
HO
OOC
HO

HN
C
CH
3
OH
O
S-glutathione
HN
C
CH
3
OH
O
S-protein
Ch 18IND page - E 2/14/03, 11:11 AM 1070
1071

NO +

O
2

O = N O O

NO binds to the iron atoms in accessible heme groups


such as those of hemoglobin
516
and of guanylate
cylases,
517,518
and in some Fe S proteins such as aconi-
tase. Some blood-sucking insects utilize salivary heme
proteins called nitrophorins to carry NO into host
tissues where it activates guanylate cyclase causing
vasodilation. Nitrophorins also bind histamine and
inhibit blood coagulation, which assists feeding.
518a,b
In the presence of a suitable oxidant such as O
2
, nitric
oxide reacts with thiol groups of proteins and small
molecules to give S-nitrosothiols (Eq. 18-63).
511,519-520
However, the physiological mechanisms of formation
of these S-nitroso compounds is not clear.
520521
One
mechanism may involve conversion by O
2
to nitrous
anhydride.
511,522
2 NO + O
2
ON O NO
Nucleophilic attack on this compound by SH, NH
2
,
and other nucleophiles would yield S - nitroso and
N- nitroso compounds with release of nitrous oxide N
2
O.
The relatively stable S-nitrosothiols derived from
glutathione, cysteine, and proteins such as hemo-
globin
516
may be important storage and transport
forms of NO. If so, mechanisms of release of NO are
important. A simple homolytic cleavage of R S NO to
NO plus a thiyl radical R S

has often been assumed.


However, rapid cleavage requires catalysis by a transi-
tion metal ion or reaction with reducing agents such
as ascorbate or other thiols.
523,524
S-Nitrosothiols may
also give rise to nitrosonium (NO
+
) or nitroxy (NO

)
ions.
524
NO synthases are oxygenases
that carry out a two-step oxidation of
L-arginine to L-citrulline with production
of NO. In the first step, a normal monooxy-
genase reaction, L-N

-hydroxyarginine is
formed (Eq. 18-65, step a). In the second
step (Eq. 18-65, step b) NO is formed in a
three-electron oxidation. In this equation
the symbols
*
and indicate positions of
incorporation of labeled O
2
atoms in the
intermediate and final products.
The human body contains three
types of nitric oxide synthase known as
neuronal (nNOS or NOS1), inducible
(iNOS or NOS2), and endothelial (eNOS or NOS3).
511af
These enzymes have a broad distribution within
various tissues, but NOS1 is especially active in neurons
and NOS3 in endothelial cells. The inducible NOS2
originally discovered in macrophages is transcription-
ally regulated. When these phagocytic cells are at rest
the activity of NOS2 is very low, but it becomes highly
active after induction by cytokines or by the lipopoly-
saccharides of bacterial cell membranes.
525,526
Both
NOS1 and NOS2 are constitutively expressed but
require calcium ions, which bind to a calmodulin
domain of the protein. The inducible NOS2 doesnt
require added Ca
2+
but does contain the calmodulin
domain. NOS3 carries an N-terminal myristoyl group
as well as cysteines that may be palmitoylated. It is
located in caveolae of plasma membranes and in Golgi
complexes.
527,528
Nucleotide sequences revealed a close similarity of
NO synthases to cytochrome P450 reductase.
525,529,530
Study of spectra suggested that NO synthases, in which
the heme is held by a thiolate sulfur of a cysteine side
chain, might be specialized cytochromes P450.
531
However, although the heme in NO synthases is
bound by a thiolate group the protein fold is unlike
that of cytochromes P450.
532,533
The NO synthases all
share a three-component structure.
534
In the NOS2
from macrophages residues 1489 form the catalytic
oxygenase domain, residues 499530 bind calmodulin,
and residues 5311144 form the reductase domain.
535
The last contains a binding site for NADPH as well as
bound FMN and FAD. The reductase domain structure
can be modeled after that of cytochrome P450 reduc-
tase.
533
Electrons from NADPH are apparently trans-
ferred to FAD, then to FMN, and then to the catalytic
site.
525
The least understood aspect of NO synthases is
the requirement for tetrahydrobiopterin, BH
4
, the
same coenzyme required by the other pterin-dependent
monooxygenases (Eq. 18-44). The presence of this co-
enzyme in the reduced BH
4
form is essential for step a
of Eq. 18-65 but not for step b. This suggests that in
step a an organic peroxide might be generated by BH
4
and used to form an oxo-iron hydroxylating reagent.
NO + R SH R S N OH
O
2
O
2

R S NO
S-Nitrosothiol

F. Oxygenases and Hydroxylases


(18-63)
(18-64)
H
N
H
2
N NH
2
COO H
3
N
H

+
l-Arginine
H
N
H
2
N N
COO H
3
N
H

+
H
N
O NH
2
COO H
3
N
H

+
OH
*
NADH
a
b
NADH
O
2
*
O
2

1
/2
NO
*
l-N

-hydroxyarginine l-Citrulline
(18-65)
(18-62)
Ch 18IND page - E 2/14/03, 11:11 AM 1071
1072 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
However, there is no evidence for the expected quino-
noid dihydropterin, and the three-dimensional struc-
ture suggested that BH
4
plays a structural role in
mediating essential conformational changes.
532,535a,b
Nevertheless, newer data indicate a role in electron
transfer.
535c
Step b of Eq. 18-65 is an unusual three-electron
oxidation, which requires only one electron to be
delivered from NADPH by the reductase domain.
Hydrogen peroxide can replace O
2
in this step.
536
A good possibility is that a peroxo or superoxide
complex of the heme in the Fe(III) state adds to the
hydroxyguanidine group. For example, the following
structure could arise from addition of Fe(III) O O

:
Breakup as indicated by the arrows on this structure
would give Fe(III) OH, citrulline, and O = N H,
nitroxyl. This is one electron (e

+ H
+
) more reduced
than NO. Perhaps the adduct forms from Fe(III) O O.
On the other hand, there is evidence that NO syn-
thases may produce nitroxyl or nitroxyl ion NO

as
the initial product.
537538
NO and other products such
as N
2
O and NO
2

may arise rapidly in subsequent


reactions. Nitrite is a major oxidation product of NO
in tissues.
538a
The chemistry of NO in biological systems
is complex and not yet fully understood. See also pp.
1754, 1755.
G. Biological Effects of Reduced Oxygen
Compounds
Although molecular oxygen is essential to the
aerobic mode of life, it is toxic at high pressures. Oxi-
dative damage from O
2
appears to be an important
cause of aging and also contributes to the development
of cancer. Reduced forms of oxygen such as super-
oxide, hydrogen peroxide, and hydroxyl radicals are
apparently involved in this toxicity.
539,540
The same
agents are deliberately used by phagocytic cells such
as the neutrophils (polymorphonuclear leukocytes)
to kill invading bacteria or fungi and to destroy malig-
nant cells.
541
The reactions shown with vertical arrows in Eq.
18-66 can give rise to the reduced oxygen compounds.
The corresponding standard redox potential at pH 7
for each is also given.
539,542 544
As indicated by the low
value of the redox potential for the O
2
/ O
2

couple,
O
C O
Fe(III)
H
N H
R
N
H
2
N
O
H
H
+
the formation of superoxide by reduction of O
2
is
spontaneous only for strongly reducing one-electron
donors. Superoxide ion is a strong reductant, but at
the same time a powerful one-electron oxidant, as is
indicated by the high electrode potential of the O
2

/
H
2
O
2
couple.
1. The Respiratory Burst of Neutrophils
Some 25 x 10
9
neutrophils circulate in an individuals
blood, and an equal number move along the surfaces
of red blood cells. Invading microorganisms are en-
gulfed after they are identified by the immune system
as foreign. Phagocytosis is accompanied by a rapid
many-fold rise in the rate of oxygen uptake as well as
an increased glucose metabolism. One purpose of this
respiratory burst
545 548
is the production of reduced
oxygen compounds that kill the ingested microorgan-
isms. In the very serious chronic granulomatous
disease the normal respiratory burst does not occur,
and bacteria are not killed.
549
The respiratory burst
seems to be triggered not by phagocytosis itself, but
by stimulation of the neutrophil by chemotactic for-
mylated peptides such as formyl-Met-Leu-Phe
550
and
less rapidly by other agonists such as phorbol esters.
The initial product of the respiratory burst appears
to be superoxide ion O
2

. It is formed by an NADPH
oxidase, which transports electrons from NADPH to
O
2
probably via a flavoprotein and cytochrome b
558
.
Either the flavin or the cytochrome b
558
must donate
one electron to O
2
to form the superoxide anion.
NADPH FAD cyt b
558
O
2
Flavocytochrome b
558
(also called b
245
) has the
unusually low redox potential of 0.245 V. It exists
in phagocytic cells as a heterodimer of membrane-
associated subunits p22-phox and gp91-phox where
phox indicates phagocytic oxidase. The larger 91-kDa
(18-66)
(18-67)
Standard electrode potential,
E
01
(pH 7), Volts
0.33
+0.94
+0.38
+2.33
O
2
H
2
O
2
2 H
2
O
O
2

H
2
O + OH (hydroxyl radical)
e

pK
a
= 4.8
HO
2

2H
+
+
H
+
+
e

H
+
+
HOO

Ch 18IND page - E 2/14/03, 11:11 AM 1072


1073
NO to form peroxynitrite (Eq. 18-62).
559b
Peroxynitrite,
in turn, can react with the ubitquitous CO
2
to give

CO
3

and

NO
2
radicals.
559c
Peroxynitrite anion also
reacts with metalloenzyme centers
559d
and causes
nitration and oxidation of aromatic residues in
proteins.
559d,e
However, neutrophils contain active
superoxide dismutases, and most of the superoxide
that is formed is converted quickly to O
2
and H
2
O
2
.
The latter may diffuse into the phagosomes as well as
into the extracellular space. The H
2
O
2
itself is toxic,
but longer lived, more toxic oxidants are also formed.
Reaction of H
2
O
2
with myeloperoxidase (Chapter 16)
produces hypochlorous acid, (HOCl; Eqs. 16-12, Fig.
18-24) and chloramines such as NH
2
Cl, RNHCl, and
RNCl
2
. An important intracellular chloramine may be
that of taurine.
Chloramine formed from taurine
Human neutrophils use HOCl formed by myelo-
peroxidase to oxidize -amino acids such as tyrosine
to reactive aldehydes that form adducts with SH,
NH
2
, imidazole, and other nucleophilic groups.
560
They also contain NO synthases, which form NO,
peroxynitrite (Fig. 18-24), and nitrite.
561,562
Hydroxyl radicals OH, which attack proteins,
nucleic acids, and a large variety of other cellular
constituents, may also be formed. Although too reac-
tive to diffuse far, they can be generated from H
2
O
2
by Eq. 18-68. This reaction involves catalysis by Fe
ions as shown.
562a
Figure 18-24 Some reactions by which
superoxide anions, hydrogen peroxide
and related compounds are generated
by neutrophils and to a lesser extent
by other cells: (a) NADPH oxidase, (b)
superoxide dismutase, (c) catalase, (d)
glucose-6-phosphate dehydrogenase,
(e) glutathione reductase, ( f ) glutathione
peroxidase. Abbrevations: GSH, gluta-
thione; GSSB, oxidized glutathione.
G. Biological Effects of Reduced Oxygen Compounds
O
2
H
2
O
2
O
2

a
b
H
2
O + O
2
b
c
HOCl
Cl

GSSG
H
2
O
GSH
e
NO
Cl
2

Cl
2
HO
+
O N O

O
NADH + H
+
NAD
+
d
6-P-gluconate
GLC 6-P
f
OH
Peroxynitrite
COO
C
CH
2
H
N Cl
SO
3

H
H
2
O
2
O
2
O
2

Fe
2+
Fe
3+
or other reductant
OH H
2
O + H
+
+
(18-68)
subunit contains two heme groups as well as one FAD
and the presumed NADPH binding site.
548,551552a
The
mechanism of interaction with O
2
is unclear. Unlike
hemoglobin but like other cytochromes b, cytochrome
b
558
does not form a complex with CO.
553
NADPH
oxidase also requires two cytosolic components p47-phox
and p67-phox. In resting phagocytes they reside in the
cytosol as a 240-kDa complex with a third component,
p40-phox, which may serve as an inhibitor.
548,554
Upon
activation of the phagocyte in response to chemotactic
signals the cytosolic components undergo phospho-
rylation at several sites,
554
and protein p47-phox and
p67-phox move to the membrane and bind to and with
the assistance of the small G protein Rac
552a,554a
activate
flavocytochrome b
558
. Phosphorylation of p47-phox
may be especially important.
555
In the X-chromosome-linked type of chronic gran-
ulomatous disease flavocytochrome b
558
is absent or
deficient, usually because of mutation in gp91-phox.
556,557
In an autosomal recessive form the superoxide-forming
oxidase system is not activated properly. In some
patients protein kinase C fails to phosphorylate p47-
phox.
556,558
Less severe symptoms arise from deficiences
in myeloperoxidase, chloroperoxidase (Chapter 16),
glucose 6-phosphate dehydrogenase, glutathione
synthetase, and glutathione reductase. The importance
of these enzymes can be appreciated by examination
of Fig. 18-24, which illustrates the relationship of
several enzymatic reactions to the formation of super-
oxide anion and related compounds. Not only neutro-
phils but monocytes, macrophages, natural killer
cells (NK cells), and other phagocytes apparently use
similar chemistry in attacking ingested cells (Chapter
31).
559
Superoxide-producing NADH oxidases have
also been found in nonphagocytic cells in various
tissues.
559a
What kills the ingested bacteria and other microor-
ganisms? Although superoxide anion is relatively
unreactive, its protonated form HO
2
, is very reactive.
Since its pK
a
is 4.8, there will be small amounts present
even at neutral pH. Some of the

O
2

may react with


Ch 18IND page - E 2/14/03, 11:11 AM 1073
1074 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Because Fe
3+
is present in such low concentrations,
there is uncertainty as to the biological significance of
this reaction.
563
However, other iron compounds may
function in place of Fe
3+
and Fe
2+
in Eq. 18-68.
564
A
mixture of ferrous salts and H
2
O
2
(Fenton reagent) has
long been recognized as a powerful oxidizing mixture,
which generates OH or compounds of similar reac-
tivity.
564 567
Ascorbate, and various other compounds,
can also serve as the reductant in Eq. 18-68.
568
Eosinophils, whose presence is stimulated by
parasitic infections, have a peroxidase which acts pref-
erentially on Br

to form HOBr.
569
This compound can
react with H
2
O
2
more efficiently than does HOCl (Eq.
16-16) to form the very reactive singlet oxygen.
570
Singlet oxygen can also be generated from H
2
O
2
and
O
2
by Eq. 18-69
571
and also photochemically.
572
O
2

+ H
2
O
2

1
O
2
(
1

g
) + OH + OH

Additional killing mechanisms used by phagocytes


include acidification of the phagocytic lysosomes with
the aid of a proton pump
573
and formation of toxic
peptides. For example, bovine neutrophils produce
the bacteriocidal peptide RLCRIVVIRVCR which has
a disulfide crosslinkage between the two cysteine
residues.
574
Microorganisms have their own defenses
against the oxidative attack by phagocytes. Some
bacteria have very active superoxide dismutases. The
protozoan Leishmania produces an acid phosphatase
that shuts down the production of superoxide of the
host cells in response to activating peptides.
575
A respiratory burst accompanies fertilization of
sea urchin eggs.
576,577
In this case, the burst appears
to produce H
2
O
2
as the major or sole product and is
accompanied by release of ovoperoxidase from cortical
granules. This enzyme uses H
2
O
2
to generate dityrosine
crosslinkages between tyrosine side chains during
formation of the fertilization membrane. Defensive
respiratory bursts are also employed by plant cells.
578,579
See also Box 18-B.
2. Oxidative Damage to Tissues
Superoxide anion radicals are formed not only in
phagocytes but also as an accidental by-product of the
action of many flavoproteins,
580,581
heme enzymes, and
other transition metal-containing proteins. An example
is xanthine oxidase. It is synthesized as xanthine
dehydrogenase which is able to use NAD
+
as an oxi-
dant, but upon aging, some is converted into the

O
2
-
utilizing xanthine oxidase (Chapter 16). This occurs
extensively during ischemia. When oxygen is read-
mitted to a tissue in which this conversion of xanthine
dehydrogenase to xanthine oxidase has occurred,
severe oxidative injury may occur.
582
In animals the
intravenous administration of superoxide dismutase
or pretreatment with the xanthine oxidase inhibitor
allopurinol (Chapter 25) prevents much of the damage,
suggesting that superoxide is the culprit.
Hydrogen peroxide is also generated within
cells
583
by flavoproteins and metalloenzymes and by
the action of superoxide dismutase on

O
2

. Since H
2
O
2
is a small uncharged molecule, it can diffuse out of
cells and into other cells readily. If it reacts with Fe(II),
it can be converted within cells to

OH radicals accord-
ing to Eq. 18-68. Such radicals and others have been
detected upon readmission of oxygen to ischemic
animal hearts.
584,585
It has also been suggested that
NADH may react with Fe(III) compounds in the same
way as does O
2

in Eq. 18-68 to provide a mechanism


for producing hydroxyl radicals from H
2
O
2
.
539
Nitric
oxide, formed by the various NO synthases in the
cytosol and in mitochondria
586
and by some cyto-
chromes P450,
587
is almost ubiquitous and can also
lead to formation of peroxynitrate (Eq. 18-62). Thus, the
whole range of reduced oxygen compounds depicted
in Eq. 18-24 are present in small amounts throughout
cells.
There is little doubt that these compounds cause
extensive damage to DNA, proteins, lipids, and other
cell constituents.
539,540,563,588
For example, one base in
150,000 in nuclear DNA is apparently converted from
guanine to 8-hydroxyguanine presumably as a result
of attack by oxygen radicals.
589
In mitochondrial DNA
one base in 8000 undergoes this alteration. This may
be a result of the high rate of oxygen metabolism in
mitochondria and may also reflect the lack of histones
and the relatively inefficient repair of DNA within
mitochondria. Proteins undergo chain cleavage,
crosslinking, and numerous side chain modification
reactions.
588
Dissolved O
2
can react directly with
exposed glycyl residues in protein backbones to create
glycyl radicals which may lead to chain cleavage as in
Eq. 15-39.
588a
Ironsulfur clusters, such as the Fe
4
S
4
center of aconitase (Fig. 13-4), are especially sensitive to
attack by superoxide anions.
588a c
Free iron released
from the FeS cluster may catalyze formation of addi-
tional damaging radicals.
Antioxidant systems. Cells have numerous
defenses against oxidative damage.
563,590,591
Both
within cells and in extracellular fluids superoxide
dismutase (Eq. 16-27) decomposes superoxide to O
2
and H
2
O
2
. The H
2
O
2
is then broken down by catalase
(Eq. 16-8) to O
2
and H
2
O. In higher animals the sele-
noenzyme glutathione peroxidase (Chapter 16) pro-
vides another route for decomposition of H
2
O
2
and
lipid peroxides of membranes. The oxidized gluta-
thione formed is reduced by NADPH. The system has
a critical role within erythrocytes (Box 15-H). In chloro-
plasts an analogous system utilizes ascorbate peroxi-
dase, ascorbate, and glutathione to break down
peroxides.
592
(18-69)
Ch 18IND page - E 2/14/03, 11:11 AM 1074
1075 G. Biological Effects of Reduced Oxygen Compounds
NADPH + H
+
glutathione ascorbate H
2
O
2
Ascorbate,
593594
glutathione, NADPH,
594a
and
tocopherols (Box 15-G)
595
all act as scavengers of free
radicals such as O
2

OH and ROO

CO
3

, and of
singlet oxygen. Antioxidant protection is needed in
extracellular fluids as well as within cells. In addition
to glutathione and ascorbate, bilirubin,
596
uric acid,
597
melatonin,
598,598a
circulating superoxide dismutase,
and the copper protein ceruloplasmin (Chapter 16) all
act as antioxidants. Methionine residues of proteins
may have a similar function.
599
Various proteins and
small chelating compounds such as citrate tie up Fe
3+
preventing it from promoting radical formation. Toco-
pherols, ubiquinols, and lipoic acid
600,600ac
protect
membranes. Beta carotene (Fig. 22-5), another lipid-
soluble antioxidant, is the most effective quencher of
singlet O
2
that is known. Even nitric oxide, usually
regarded as toxic, sometimes acts as an antioxidant.
601
Trehalose protects plants against oxidative damage.
601a
An increasing number of proteins are being recog-
nized as protectants against oxidative damage. The
exposed SH and SCH
3
groups of cysteine and methio-
nine residues in proteins may function as appropriately
located scavengers which may donate electrons to
destroy free radicals or react with superoxide ions to
become sulfonated. The thioredoxin (Box 15-C) and
glutathione (Eq. 18-70) systems, in turn, reduce the
protein radicals formed in this way.
601b d
Methionine
sulfoxide, both free and in polypeptides, is reduced
by methionine sulfoxide reductase in organisms
from bacteria to humans.
601e g
Biotin, together with
biotin sulfoxide reductase,
601h
may provide another
antioxidant system. Some bacteria utilize glutathione-
independent alkylperoxide reductases to scavenge
organic peroxides.
601i
while mammals accomplish
the same result with peroxiredoxins and with
thioredoxin.
601j
Many other proteins will doubtless
be found to participate in defense against oxidative
damage. Oxygen is always present and its reactions
in our bodies are essential. Generation of damaging
reduced oxygen compounds and radicals is inevitable.
Evolution will select in favor of many proteins that
have been modified to minimize the damage.
Antioxidant enzymes do not always protect us.
There was great excitement when it was found that
victims of a hereditary form of the terrible neurological
disease amyotrophic lateral sclerosis (ALS; see
also Chapter 30) carry a defective gene for Cu / Zn-
superoxide dismutase (SOD; Eq. 16-27).
602603b
This
discovery seemed to support the idea that superoxide
anions in the brain were killing neurons. However, it
now appears that in some cases of ALS the defective
SOD is too active, producing an excess of H
2
O
2
, which
damages neurons.
(18-70)
Transcriptional regulation of antioxidant
proteins. Certain proteins with easily accessible FeS
clusters, e.g., aconitase, are readily inactivated by oxi-
dants such as peroxynitrite.
540,604
At least two proteins
of this type function as transcription factors in E. coli.
These are known as SoxR and OxyR. The SoxR pro-
tein is sensitive to superoxide anion, which carries out
a one-electron oxidation on its Fe
2
S
2
centers.
540,605 607
In its oxidized form SoxR is a transcriptional regulator
that controls 30 40 genes, among them several that
are directly related to oxidative stress.
608
These
include genes for manganese SOD, glucose-6-phosphate
dehydrogenase, a DNA repair nuclease, and aconitase
(to replace the inactivated enzyme). The OxyR protein,
which responds to elevated [H
2
O
2
], is activated upon
oxidation of a pair of nearby SH groups to form a
disulfide bridge.
607,609
It controls genes for catalase,
glutathione reductase, an alkyl hydroperoxide reduc-
tase,
610,610a
and many others. Similar transcriptional
controls in yeast result in responses to low doses of
H
2
O
2
by at least 167 different proteins.
608
Animal
mitochondria also participate in sensing oxidant
levels.
611,612
(See also Chapter 28, Section C,6.)
Ch 18IND page - E 2/14/03, 11:11 AM 1075
1076 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
42. Oldenburg, D. J., and Bendich, A. J. (1998) J.
Mol. Biol. 276, 745758
42a. Oldenburg, D. J., and Bendich, A. J. (2001) J.
Mol. Biol. 310, 549562
43. Barrell, B. G., Bankier, A. T., and Drouin, J.
(1979) Nature (London) 282, 189194
44. Wallace, D. C. (1982) Microbiol. Rev. 46, 208240
45. Shoffner, J. M., and Wallace, D. C. (1995) in
The Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 1535
1609, McGraw-Hill, New York
46. Wallace, D. C. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 87398746
47. Butow, R. A., Perlman, P. S., and Grossman, L.
I. (1985) Science 228, 14961501
48. Douglas, M., and Takeda, M. (1985) Trends
Biochem. Sci. 10, 192194
49. Cavalier-Smith, T. (1987) Nature (London) 326,
332333
50. Schatz, G. (1997) Nature (London) 388, 121122
50a. Tokatlidis, K., and Schatz, G. (1999) J. Biol.
Chem. 274, 3528535288
50b. Gabriel, K., Buchanan, S. K., and Lithgow, T.
(2001) Trends Biochem. Sci. 26, 3640
51. Schatz, G., and Dobberstein, B. (1996) Science
271, 15191526
52. Stuart, R. A., and Neupert, W. (1996) Trends
Biochem. Sci. 21, 261267
53. Lithgow, T., Cuezva, J. M., and Silver, P. A.
(1997) Trends Biochem. Sci. 22, 110113
54. Rojo, E. E., Guiard, B., Neupert, W., and
Stuart, R. A. (1998) J. Biol. Chem. 273, 8040
8047
55. Hartmann, C., Christen, P., and Jaussi, R.
(1991) Nature (London) 352, 762763
56. Grtner, F., Voos, W., Querol, A., Miller, B. R.,
Craig, E. A., Cumsky, M. G., and Pfanner, N.
(1995) J. Biol. Chem. 270, 37883795
57. Pfanner, N., Douglas, M. G., Endo, T.,
Hoogenraad, N. J., Jensen, R. E., Meijer, M.,
Neupert, W., Schatz, G., Schmitz, U. K., and
Shore, G. C. (1996) Trends Biochem. Sci. 21, 51
52
58. Komiya, T., Rospert, S., Koehler, C., Looser,
R., Schatz, G., and Mihara, K. (1998) EMBO J.
17, 38863898
59. Dietmeier, K., Hnlinger, A., Bmer, U.,
Dekker, P. J. T., Eckerskorn, C., Lottspeich, F.,
Kbrich, M., and Pfanner, N. (1997) Nature
(London) 388, 195200
59a. Koehler, C. M., Leuenberger, D., Merchant, S.,
Renold, A., Junne, T., and Schatz, G. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 21412146
59b. Wallace, D. C., and Murdock, D. G. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 18171819
60. Chance, B., and Williams, G. R. (1955) J. Biol.
Chem. 217, 409427
61. Prebble, J. N. (1981) Mitochondria Chloroplasts
and Bacterial Membranes, Longman, London
and New York
62. King, T. E. (1967) Methods Enzymol. 10, 202
208
63. Ragen, C. I., and Racker, E. (1973) J. Biol.
Chem. 248, 25632569
64. Hafeti, Y. (1985) Ann. Rev. Biochem. 54, 1015
1019
65. Yu, L., and Yu, C.-A. (1982) J. Biol. Chem. 257,
20162021
66. Yu, C.-A., and Yu, L. (1982) J. Biol. Chem. 257,
61276131
67. Wakabayashi, S., Takao, T., Shimonishi, Y.,
Kuramitsu, S., Matsubara, H., Wang, T.,
Zhang, Z., and King, T. E. (1985) J. Biol. Chem.
260, 337343
68. Moore, A. L., and Rich, P. R. (1980) Trends
Biochem. Sci. 5, 284288
69. Palmer, J. M., and Moller, I. M. (1982) Trends
Biochem. Sci. 7, 258261
1. Tzagoloff, A. (1982) Mitochondria, Plenum,
New York
1a. Scheffler, I. E. (1999) Mitochondria, Wiley-Liss,
New York
2. Chappell, J. B. (1979) The Energetics of
Mitochondria, 2nd ed., Oxford Univ. Press,
London (Carolina Biology Reader No. 19)
3. Lee, C. P., Schatz, G., and Dallner, G., eds.
(1981) Mitochondria and Microsomes, Addison-
Wesley, Reading, Massachusetts
4. Tyler, D. D. (1992) The Mitochondrion in Health
and Disease, VCH Publ., New York
5. Noble, R. W., and Gibson, Q. H. (1970) J. Biol.
Chem. 245, 24092413
6. Ankel-Simons, F., and Cummins, J. M. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 1385913863
7. Champion, P. M., Mnck, E., Debrunner, P. G.,
Hollenberg, P. F., and Hager, L. P. (1973)
Biochemistry 12, 426435
8. Packer, L. (1973) in Mechanisms in Bioenergetics
(Azzone, G. F., Ernster, L., Papa, S.,
Quagliariello, E., and Siliprandi, N., eds), pp.
3352, Academic Press, New York
9. Smith, L. D., and Gholson, R. K. (1969) J. Biol.
Chem. 244, 6871
10. Harris, R. A., Williams, C. H., Caldwell, M.,
Green, D. E., and Valdivia, E. (1969) Science
165, 700703
11. Malhotra, S. K., and Sikewar, S. S. (1983)
Trends Biochem. Sci. 8, 358359
12. Frey, T. G., and Mannella, C. A. (2000) Trends
Biochem. Sci. 25, 319324
12a. Varmus, H. E. (1985) Nature (London) 314,
583584
12b. Rutter, G. A., and Rizzuto, R. (2000) Trends
Biochem. Sci. 25, 215221
13. Ernster, L., and Drahota, Z., eds. (1969)
Mitochondria: Structure and Function, Vol. 17,
FEBS Symposium, (pp. 531)
14. Pfaller, R., Freitag, H., Harmey, M. A., Benz,
R., and Neupert, W. (1985) J. Biol. Chem. 260,
81888193
15. Heins, L., Mentzel, H., Schmid, A., Benz, R.,
and Schmitz, U. K. (1994) J. Biol. Chem. 269,
2640226410
15a. Blter, B., and Soll, J. (2001) EMBO J. 20, 935
940
16. Mannella, C. A. (1992) Trends Biochem. Sci. 17,
315320
17. Ha, H., Hajek, P., Bedwell, D. M., and
Burrows, P. D. (1993) J. Biol. Chem. 268, 12143
12149
18. Sutfin, L. V., Holtrop, M. E., and Ogilvie, R. E.
(1971) Science 174, 947 949
19. Srere, P. A. (1982) Trends Biochem. Sci. 7,
375377
20. Srere, P. A. (1981) Trends Biochem. Sci. 6, 47
20a. Haggie, P. M., and Brindle, K. M. (1999) J. Biol.
Chem. 274, 39413945
21. McCabe, E. R. B. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 1631 1652, McGraw-
Hill, New York
22. Hackenbrock, C. R., and Hammon, K. M.
(1975) J. Biol. Chem. 250, 91859197
22a. Jiang, F., Ryan, M. T., Schlame, M., Zhao, M.,
Gu, Z., Klingenberg, M., Pfanner, N., and
Greenberg, M. L. (2000) J. Biol. Chem. 275,
2238722394
22b. Sedlk, E., and Robinson, N. C. (1999)
Biochemistry 38, 1496614972
22c. Gomez, B., Jr., and Robinson, N. C. (1999)
Biochemistry 38, 90319038
22d. McAuley, K. E., Fyfe, P. K., Ridge, J. P., Isaacs,
N. W., Cogdell, R. J., and Jones, M. R. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 1470614711
22e. Bazhenova, E. N., Deryabina, Y. I., Eriksson,
O., Zvyagilskaya, R. A., and Saris, N.-E. L.
(1998) J. Biol. Chem. 273, 43724377
22f. Horikawa, Y., Goel, A., Somlyo, A. P., and
Somlyo, A. V. (1998) Biophys. J. 74, 15791590
22g. Territo, P. R., French, S. A., Dunleavy, M. C.,
Evans, F. J., and Balaban, R. S. (2001) J. Biol.
Chem. 276, 25862599
22h. Smaili, S. S., Stellato, K. A., Burnett, P.,
Thomas, A. P., and Gaspers, L. D. (2001) J.
Biol. Chem. 276, 2332923340
22i. Beutner, G., Sharma, V. K., Giovannucci, D.
R., Yule, D. I., and Sheu, S.-S. (2001) J. Biol.
Chem. 276, 2148221488
22j. Arnaudeau, S., Kelley, W. L., Walsh, J. V., Jr.,
and Demaurex, N. (2001) J. Biol. Chem. 276,
2943029439
22k. Berridge, M. J., Bootman, M. D., and Lipp, P.
(1998) Nature (London) 395, 645648
23. Clayton, D. A. (1984) Ann. Rev. Biochem. 53,
573594
24. Slonirnski, P., Borst, P., and Attardi, G., eds.
(1982) Mitochondrial Genes, Cold Spring Harbor
Lab. Press, Cold Spring Harbor, New York
25. Attardi, G. (1981) Trends Biochem. Sci. 6, 86
89; 100103
26. Palmer, J. D. (1997) Nature (London) 387, 454
455
26a. Schwartz, M., and Vissing, J. (2002) N. Engl. J.
Med. 347, 576580
27. Wolstenholme, D. R. (1992) in Mitochondrial
GenomesInternational Review of Cytology, Vol.
141 (Wolstenholme, D. R., and Jeon, K. W.,
eds), pp. 173216, Academic Press, San
Diego, California
28. Wolstenholme, D. R., and Jeon, K. W., eds.
(1992) Mitochondrial Genomes, Vol. 141,
Academic Press, San Diego, California
28a. Wolfsberg, T. G., Schafer, S., Tatusov, R. L.,
and Tatusova, T. A. (2001) Trends Biochem. Sci.
26, 199203
29. Borst, P., and Grivell, L. A. (1981) Nature
(London) 290, 443444
30. Janke, A., Xu, X., and Arnason, U. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 12761281
31. Anderson, S., Bankier, A. T., Barrell, B. G.,
deBruijn, M. H. L., Coulson, A. R., Drouin, J.,
Eperon, I. C., Nierlich, D. P., Roe, B. A.,
Sanger, F., Schreier, P. H., Smith, A. J. H.,
Staden, R., and Young, I. G. (1981) Nature
(London) 290, 457470
32. Anderson, S., de Bruijn, M. H. L., Coulson, A.
R., Eperon, I. C., Sanger, F., and Young, I. G.
(1984) J. Mol. Biol. 156, 683717
33. Cantatore, P., Roberti, M., Rainaldi, G.,
Gadaleta, M. N., and Saccone, C. (1989) J. Biol.
Chem. 264, 1096510975
34. Gardner, M. J., and 26 other authors (1998)
Science 282, 11261132
35. Khler, S., Delwiche, C. F., Denny, P. W.,
Tilney, L. G., Webster, P., Wilson, R. J. M.,
Palmer, J. D., and Roos, D. S. (1997) Science
275, 14851489
36. Burger, G., Plante, I., Lonergan, K. M., and
Gray, M. W. (1995)
J. Mol. Biol. 245, 522537
37. Lang, B. F., Burger, G., OKelly, C. J.,
Cedergren, R., Golding, G. B., Lemieux, C.,
Sankoff, D., Turmel, M., and Gray, M. W.
(1997) Nature (London) 387, 493497
38. de Bruijn, M. H. L. (1983) Nature (London) 304,
234241
39. Bernardi, G. (1982) Trends Biochem. Sci. 7, 404
408
40. Palmer, J. D., and Shields, C. R. (1984) Nature
(London) 307, 437440
41. Leblanc, C., Boyen, C., Richard, O., Bonnard,
G., Grienenberger, J.-M., and Kloareg, B.
(1995) J. Mol. Biol. 250, 484495
References
Ch 18IND page - E 2/14/03, 11:11 AM 1076
1077
70. Douce, R., and Day, D. A., eds. (1985) Higher
Plant Cell Respiration, Vol. 18, Springer-Verlag,
New York
71. Rhoads, D. M., and McIntosh, L. (1991) Proc.
Natl. Acad. Sci. U.S.A. 88, 21222126
72. Clarkson, A. B., JR, Bienen, E. J., Pollakis, G.,
and Grady, R. W. (1989) J. Biol. Chem. 264,
1777017776
73. Hoefnagel, M. H. N., Atkin, O. K., and
Wiskich, J. T. (1998) Biochim. Biophys. Acta.
1366, 235255
74. Albury, M. S., Affourtit, C., and Moore, A. L.
(1998) J. Biol. Chem. 273, 30301 30305
75. Rhoads, D. M., Umbach, A. L., Sweet, C. R.,
Lennon, A. M., Rauch, G. S., and Siedow, J. N.
(1998) J. Biol. Chem. 273, 3075030756
76. Anraku, Y. (1988) Ann. Rev. Biochem. 57, 101
132
77. Trumpower, B. L., and Gennis, R. B. (1994)
Ann. Rev. Biochem. 63, 675716
78. Yamaguchi, M., Belogrudov, G. I., and Hatefi,
Y. (1998) J. Biol. Chem. 273, 80948098
78a. Grivennikova, V. G., Kapustin, A. N., and
Vinogradov, A. D. (2001) J. Biol. Chem. 276,
90389044
79. Gunebaut, V., Schlitt, A., Weiss, H., Leonard,
K., and Friedrich, T. (1998) J. Mol. Biol. 276,
105112
79a. Hellwig, P., Scheide, D., Bungert, S., Mntele,
W., and Friedrich, T. (2000) Biochemistry 39,
1088410891
80. Weidner, U., Geier, S., Ptock, A., Friedrich, T.,
Leif, H., and Weiss, H. (1993) J. Mol. Biol. 233,
109122
81. Schryvers, A., Lohmeier, E., and Weiner, J. H.
(1978) J. Biol. Chem. 253, 783788
82. Koland, J. G., Miller, M. J., and Gennis, R. B.
(1984) Biochemistry 23, 445453
83. Condon, C., Cammack, R., Patil, D. S., and
Owen, P. (1985) J. Biol. Chem. 260, 94279434
84. Berry, E. A., and Trumpower, B. L. (1985) J.
Biol. Chem. 260, 24582467
85. John, P. (1981) Trends Biochem. Sci. 6, 810
86. Kaysser, T. M., Ghaim, J. B., Georgiou, C., and
Gennis, R. B. (1995) Biochemistry 34, 13491
13501
87. Anraku, Y., and Gennis, R. B. (1987) Trends
Biochem. Sci. 12, 262266
88. Sun, J., Kahlow, M. A., Kaysser, T. M.,
Osborne, J. P., Hill, J. J., Rohlfs, R. J., Hille, R.,
Gennis, R. B., and Loehr, T. M. (1996)
Biochemistry 35, 24032412
88a. Zhang, J., Hellwig, P., Osborne, J. P., Huang,
H.-w, Monne-Loccoz, P., Konstantinov, A. A.,
and Gennis, R. B. (2001) Biochemistry 40,
85488556
88b. Gerscher, S., Dpner, S., Hildebrandt, P.,
Gleissner, M., and Schfer, G. (1996)
Biochemistry 35, 1279612803
88c. Das, T. K., Gomes, C. M., Teixeira, M., and
Rousseau, D. L. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 95919596
88d. Saraste, M. (1999) Science 283, 14881493
88e. Villani, G., Capitanio, N., Bizzoca, A., Palese,
L. L., Carlino, V., Tattoli, M., Glaser, P.,
Danchin, A., and Papa, S. (1999) Biochemistry
38, 22872294
89. Gunebaut, V., Vincentelli, R., Mills, D.,
Weiss, H., and Leonard, K. R. (1997) J. Mol.
Biol. 265, 409418
89a. Schuler, F., Yano, T., Di Bernardo, S., Yagi, T.,
Yankovskaya, V., Singer, T. P., and Casida, J. E.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 4149
4153
90. Grigorieff, N. (1998) J. Mol. Biol. 277, 1033
1046
91. Di Bernardo, S., Yano, T., and Yagi, T. (2000)
Biochemistry 39, 94119418
References
92. Schneider, R., Brors, B., Massow, M., and
Weiss, H. (1997) FEBS Lett. 407, 249252
93. Ohnishi, T., Ragan, C. I., and Hatefi, Y. (1985)
J. Biol. Chem. 260, 27822788
94. Yano, T., Sled, V. D., Ohnishi, T., and Yagi, Y.
(1996) J. Biol. Chem. 271, 59075913
95. Ragan, C. I., Galante, Y. M., Hatefi, Y., and
Ohnishi, T. (1984) Biochemistry 21, 590594
95a. Kashani-Poor, N., Zwicker, K., Kerscher, S.,
and Brandt, U. (2001) J. Biol. Chem. 276,
2408224087
96. Heinrich, H., Azevedo, J. E., and Werner, S.
(1992) Biochemistry 31, 1142011424
97. Ohshima, M., Miyoshi, H., Sakamoto, K.,
Takegami, K., Iwata, J., Kuwabara, K.,
Iwamura, H., and Yagi, T. (1998) Biochemistry
37, 64366445
98. Kowal, A. T., Werth, M. T., Manodori, A.,
Cecchini, G., Schrder, I., Gunsalus, R. P., and
Johnson, M. K. (1995) Biochemistry 34, 12284
12293
98a. Iverson, T. M., Luna-Chavez, C., Cecchini, G.,
and Rees, D. C. (1999) Science 284, 19611966
98b. Doherty, M. K., Pealing, S. L., Miles, C. S.,
Moysey, R., Taylor, P., Walkinshaw, M. D.,
Reid, G. A., and Chapman, S. K. (2000)
Biochemistry 39, 1069510701
98c. Lancaster, C. R. D., Krger, A., Auer, M., and
Michel, H. (1999) Nature (London) 402, 377
385
98d. Lancaster, C. R. D., Gross, R., Haas, A., Ritter,
M., Mntele, W., Simon, J., and Krger, A.
(2000) Proc. Natl. Acad. Sci. U.S.A. 97, 13051
13056
98e. Matsson, M., Tolstoy, D., Aasa, R., and
Hederstedt, L. (2000) Biochemistry 39, 8617
8624
99. Westenberg, D. J., Gunsalus, R. P., Ackrell, B.
A. C., Sices, H., and Cecchini, G. (1993) J. Biol.
Chem. 268, 815822
99a. Mowat, C. G., Pankhurst, K. L., Miles, C. S.,
Leys, D., Walkinshaw, M. D., Reid, G. A., and
Chapman, S. K. (2002) Biochemistry 41, 11990
11996
100. Van Hellemond, J. J., Klockiewicz, M.,
Gaasenbeek, C. P. H., Roos, M. H., and
Tielens, A. G. M. (1995) J. Biol. Chem. 270,
3106531070
101. Nakamura, K., Yamaki, M., Sarada, M.,
Nakayama, S., Vibat, C. R. T., Gennis, R. B.,
Nakayashiki, T., Inokuchi, H., Kojima, S., and
Kita, K. (1996) J. Biol. Chem. 271, 521527
102. Vibat, C. R. T., Cecchini, G., Nakamura, K.,
Kita, K., and Gennis, R. B. (1998) Biochemistry
37, 41484159
103. Yang, X., Yu, L., and Yu, C.-A. (1997) J. Biol.
Chem. 272, 96839689
104. Xia, D., Yu, C.-A., Kim, H., Xia, J.-Z.,
Kachurin, A. M., Zhang, L., Yu, L., and
Deisenhofer, J. (1997) Science 277, 6066
105. Zhang, Z., Huang, L., Shulmeister, V. M., Chi,
Y.-I., Kim, K. K., Hung, L.-W., Crofts, A. R.,
Berry, E. A., and Kim, S.-H. (1998) Nature
(London) 392, 677684
105a. Crofts, A. R., Hong, S., Zhang, Z., and Berry,
E. A. (1999) Biochemistry 38, 1582715839
106. Iwata, S., Lee, J. W., Okada, K., Lee, J. K.,
Iwata, M., Rasmussen, B., Link, T. A.,
Ramaswamy, S., and Jap, B. K. (1998) Science
281, 6471
107. Smith, J. L. (1998) Science 281, 5859
107a. Baymann, F., Robertson, D. E., Dutton, P. L.,
and Mntele, W. (1999) Biochemistry 38,
1318813199
107b. Lange, C., and Hunte, C. (2002) Proc. Natl.
Acad. Sci. U.S.A. 99, 28002805
108. Braun, H.-P., and Schmitz, U. K. (1995) Trends
Biochem. Sci. 20, 171175
108a. Deng, K., Shenoy, S. K., Tso, S.-C., Yu, L., and
Yu, C.-A. (2001) J. Biol. Chem. 276, 64996505
109. Saribas, A. S., Ding, H., Dutton, P. L., and
Daldal, F. (1995) Biochemistry 34, 1600416012
110. Denke, E., Merbitz-Zahradnik, T., Hatzfeld,
O. M., Snyder, C. H., Link, T. A., and
Trumpower, B. L. (1998) J. Biol. Chem. 273,
90859093
111. Mitchell, P. (1976) J. Theor. Biol. 62, 327367
112. Trumpower, B. L. (1990) J. Biol. Chem. 265,
1140911412
113. Orii, Y., and Miki, T. (1997) J. Biol. Chem. 272,
1759417604
113a. Bartoschek, S., Johansson, M., Geierstanger, B.
H., Okun, J. G., Lancaster, C. R. D., Humpfer,
E., Yu, L., Yu, C.-A., Griesinger, C., and Brandt,
U. (2001) J. Biol. Chem. 276, 3523135234
114. Jnemann, S., Heathcote, P., and Rich, P. R.
(1998) J. Biol. Chem. 273, 2160321607
114a. Darrouzet, E., Moser, C. C., Dutton, P. L., and
Daldal, F. (2001) Trends Biochem. Sci. 26, 445
451
114b. Darrouzet, E., Valkova-Valchanova, M., Moser,
C. C., Dutton, P. L., and Daldal, F. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 45674572
114c. Crofts, A. R., Barquera, B., Gennis, R. B.,
Kuras, R., Guergova-Kuras, M., and Berry, E.
A. (1999) Biochemistry 38, 1580715826
115. Tian, H., Yu, L., Mather, M. W., and Yu, C.-A.
(1998) J. Biol. Chem. 273, 2795327959
116. Meinharat, S. M., Yang, X., Trumpower, B. L.,
and Ohnishi, T. (1987) J. Biol. Chem. 262, 8702
8706
117. Saribas, A. M., Valkova-Valchanova, M.,
Tokito, M. K., Zhang, Z., Berry, E. A., and
Daldal, F. (1998) Biochemistry 37, 81058114
118. Yu, J., and Le Brun, N. E. (1998) J. Biol. Chem.
273, 88608866
119. Barber, J. (1984) Trends Biochem. Sci. 9, 209211
120. Joliot, P., and Joliot, A. (1998) Biochem. Soc.
Trans. 37, 1040410410
120a. Finazzi, G. (2002) Biochemistry 41, 74757482
121. Hochman, J., Ferguson-Miller, S., and
Scindler, M. (1985) Biochemistry 24, 25092516
122. Hackenbrock, C. R. (1981) Trends Biochem. Sci.
6, 151154
123. Bernardi, P., and Azzone, G. F. (1981) J. Biol.
Chem. 256, 71877192
124. Vincent, J. S., and Levin, I. W. (1986) J. Am.
Chem. Soc. 108, 35513554
125. Tsukihara, T., Aoyama, H., Yamashita, E.,
Tomizaki, T., Yamaguchi, H., Shinzawa-Itoh,
K., Nakashima, R., Yaono, R., and Yoshikawa,
S. (1996) Science 272, 11361144
126. Gennis, R., and Ferguson-Miller, S. (1995)
Science 269, 10631064
127. Wallin, E., Tsukihara, T., Yoshikawa, S., Von
Heijne, G., and Elofsson, A. (1997) Protein Sci.
6, 808815
128. Iwata, S., Ostermeier, C., Ludwig, B., and
Michel, H. (1995) Nature (London) 376, 660669
129. Tsukihara, T., Aoyama, H., Yamashita, E.,
Tomizaki, T., Yamaguchi, H., Shinzawa-Itoh,
K., Nakashima, R., Yaono, R., and Yoshikawa,
S. (1995) Science 269, 10691074
130. Luchinat, C., Soriano, A., Djinovic-Carugo, K.,
Saraste, M., Malmstrm, B. G., and Bertini, I.
(1997) J. Am. Chem. Soc. 119, 1102311027
131. Salgado, J., Warmerdam, G., Bubacco, L., and
Canters, G. W. (1998) Biochemistry 37, 7378
7389
132. Blackburn, N. J., de Vries, S., Barr, M. E., Houser,
R. P., Tolman, W. B., Sanders, D., and Fee, J. A.
(1997) J. Am. Chem. Soc. 119, 61356143
133. Yoshikawa, S., Shinzawa-Itoh, K., Nakashima,
R., Yaono, R., Yamashita, E., Inoue, N., Yao,
M., Fei, M. J., Libeu, C. P., Mizushima, T.,
Yamaguchi, H., Tomizaki, T., and Tsukihara,
T. (1998) Science 280, 17241729
Ch 18IND page - E 2/14/03, 11:11 AM 1077
1078 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
185. Jagendorf, A. T. (1975) in Bioenergetics of
Photosynthesis (Govindjee, ed), Academic
Press, New York (pp. 413492)
186. Jagendorf, A. T., and Uribe, E. (1966) Proc.
Natl. Acad. Sci. U.S.A. 55, 170177
187. Richard, P., Pitard, B., and Rigaud, J.-L. (1995)
J. Biol. Chem. 270, 2157121578
188. van Walraven, H. S., Strotmann, H., Schwarz,
O., and Rumberg, B. (1996) FEBS Lett. 379, 309
313
189. Costa, L. E., Reynafarje, B., and Lehninger, A.
L. (1984) J. Biol. Chem. 259, 48024811
190. Reynafarje, B., Costa, L. E., and Lehninger, A.
L. (1986) J. Biol. Chem. 261, 82548262
191. Capitanio, N., Capitanio, G., Demarinis, D.
A., De Nitto, E., Massari, S., and Papa, S.
(1996) Biochemistry 35, 1080010806
192. Wang, Y., Howton, M. M., and Beattie, D. S.
(1995) Biochemistry 34, 74767482
193. Wikstrm, M. (1989) Nature (London) 338,
776778
194. Wikstrm, M., Bogacher, A., Finel, M.,
Morgan, J. E., Puustinen, M., Raitio, M. L.,
Verkhovskaya, M. L., and Verkhovsky, M. I.
(1994) Biochim. Biophys. Acta. 1187, 106111
195. Williams, R. J. P. (1988) Ann. Rev. Biophys.
Biophys. Chem. 17, 7197
196. Penefsky, H. S., Pullman, M. E., Datta, A., and
Racker, E. (1960) J. Biol. Chem. 235, 33303336
197. Racker, E. (1976) A New Look at Mechanisms in
Bioenergetics, Academic Press, New York
198. Dschida, W. J., and Bowman, B. J. (1992) J.
Biol. Chem. 267, 1878318789
199. Abrahams, J. P., Leslie, A. G. W., Lutter, R.,
and Walker, J. E. (1994) Nature (London) 370,
621628
200. Elston, T., Wang, H., and Oster, G. (1998)
Nature (London) 391, 510513
201. Zhou, Y., Duncan, T. M., and Cross, R. L.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 10583
10587
202. Junge, W., Lill, H., and Engelbrecht, S. (1997)
Trends Biochem. Sci. 22, 420423
203. Engelbrecht, S., and Junge, W. (1997) FEBS
Letters 414, 485491
204. Amzel, L. M., and Pedersen, P. L. (1983) Ann.
Rev. Biochem. 52, 801824
205. Hausrath, A. C., Grber, G., Matthews, B. W.,
and Capaldi, R. A. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 1369713702
206. Matsui, T., Muneyuki, E., Honda, M., Allison,
W. S., Dou, C., and Yoshida, M. (1997) J. Biol.
Chem. 272, 82158221
207. Shirakihara, Y., Leslie, A. G. W., Abrahams, J.
P., Walker, J. E., Ueda, T., Sekimoto, Y.,
Kambara, M., Saika, K., Kagawa, Y., and
Yoshida, M. (1997) Structure 5, 825836
208. Spannagel, C., Vaillier, J., Chaignepain, S., and
Velours, J. (1998) Biochemistry 37, 615621
208a. Arnold, I., Pfeiffer, K., Neupert, W., Stuart, R.
A., and Schgger, H. (1999) J. Biol. Chem. 274,
3640
208b. Soubannier, V., Rusconi, F., Vaillier, J., Arselin,
G., Chaignepain, S., Graves, P.-V., Schmitter,
J.-M., Zhang, J. L., Mueller, D., and Velours, J.
(1999) Biochemistry 38, 1501715024
209. Collinson, I. R., Runswick, M. J., Buchanan, S.
K., Fearnley, I. M., Skehel, J. M., van Raaij, M.
J., Griffiths, D. E., and Walker, J. E. (1994)
Biochemistry 33, 79717978
210. Pedersen, P. L., and Amzel, L. M. (1993) J. Biol.
Chem. 268, 99379940
210a. Hee Ko, Y., Hullihen, J., Hong, S., and Pedersen,
P. L. (2000) J. Biol. Chem. 275, 3293132939
210b. Karrasch, S., and Walker, J. E. (1999) J. Mol.
Biol. 290, 379 384
211. Bianchet, M. A., Hullihen, J., Pedersen, P. L.,
and Amzel, L. M. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 1106511070
134. Ostermeier, C., Harrenga, A., Ermler, U., and
Michel, H. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 1054710553
135. Stubbe, J., and Riggs-Gelasco, P. (1998) Trends
Biochem. Sci. 23, 438443
135a. Buse, G., Soulimane, T., Dewor, M., Meyer, H.
E., and Blggel, M. (1999) Protein Sci. 8, 985
990
136. Babcock, G. T., and Wikstrm, M. (1992)
Nature (London) 356, 301309
136a. Babcock, G. T. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 1297112973
136b. Wikstrm, M. (2000) Biochemistry 39, 3515
3519
136c. Morgan, J. E., Verkhovsky, M. I., Palmer, G.,
and Wilstrm, M. (2001) Biochemistry 40,
68826892
137. Gennis, R. B. (1998) Proc. Natl. Acad. Sci.
U.S.A. 95, 1274712749
138. Michel, H. (1999) Biochemistry 38, 15129
15140
139. Sucheta, A., Georgiadis, K. E., and
Einarsdttir, O. (1997) Biochemistry 36, 554
565
140. Gibson, Q., and Greenwood, C. (1963)
Biochem. J. 86, 541554
141. Greenwood, C., and Gibson, Q. H. (1967) J.
Biol. Chem. 242, 17821787
142. Fabian, M., and Palmer, G. (2001) Biochemistry
40, 18671874
142a. Han, S., Takahashi, S., and Rousseau, D. L.
(2000) J. Biol. Chem. 275, 19101919
142b. Karpefors, M., delroth, P., Namslauer, A.,
Zhen, Y., and Brzezinski, P. (2000) Biochemistry
39, 1466414669
142c. Capitanio, N., Capitanio, G., Minuto, M., De
Nitto, E., Palese, L. L., Nicholls, P., and Papa,
S. (2000) Biochemistry 39, 63736379
143. Karpefors, M., delroth, P., Zhen, Y.,
Ferguson-Miller, S., and Brzezinski, P. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 1360613611
143a. Rigby, S. E. J., Jnemann, S., Rich, P. R., and
Heathcote, P. (2000) Biochemistry 39, 5921
5928
143b. Pecoraro, C., Gennis, R. B., Vygodina, T. V.,
and Konstantinov, A. A. (2001) Biochemistry
40, 96959708
144. Zaslavsky, D., Sadoski, R. C., Wang, K.,
Durham, B., Gennis, R. B., and Millett, F.
(1998) Biochemistry 37, 1491014916
145. Nicholls, P. (1983) Trends Biochem. Sci. 8, 353
146. Solioz, M. (1984) Trends Biochem. Sci. 9, 309
312
147. Musser, S. M., and Chan, S. I. (1995) Biophys. J.
68, 25432555
147a. Siletsky, S., Kaulen, A. D., and Konstantinov,
A. A. (1999) Biochemistry 38, 48534861
148. Aagaard, A., Gilderson, G., Mills, D. A.,
Ferguson-Miller, S., and Brzezinski, P. (2000)
Biochemistry 39, 1584715850
148a. Brndn, M., Sigurdson, H., Namslauer, A.,
Gennis, R. B., delroth, P., and Brzezinski, P.
(2001) Proc. Natl. Acad. Sci. U.S.A. 98, 5013
5018
149. Mitchell, D. M., Fetter, J. R., Mills, D. A.,
Adelroth, P., Pressler, M. A., Kim, Y., Aasa, R.,
Brzezinski, P., Malmstrm, B. G., Alben, J. O.,
Babcock, G. T., Ferguson-Miller, S., and
Gennis, R. B. (1996) Biochemistry 35, 13089
13093
150. Qian, J., Shi, W., Pressler, M., Hoganson, C.,
Mills, D., Babcock, G. T., and Ferguson-Miller,
S. (1997) Biochemistry 36, 25392543
151. delroth, P., Gennis, R. B., and Brzezinski, P.
(1998) Biochemistry 37, 24702476
152. Konstantinov, A. A., Siletsky, S., Mitchell, D.,
Kaulen, A., and Gennis, R. B. (1997) Proc. Natl.
Acad. Sci. U.S.A. 94, 90859090
153. Behr, J., Hellwig, P., Mntele, W., and Michel,
H. (1998) Biochemistry 37, 74007406
153a. Riistama, S., Laakkonen, L., Wikstrm, M.,
Verkhovsky, M. I., and Puustinen, A. (1999)
Biochemistry 38, 1067010677
153b. Jnemann, S., Meunier, B., Fisher, N., and
Rich, P. R. (1999) Biochemistry 38, 52485255
154. Marantz, Y., Nachliel, E., Aagaard, A.,
Brzezinski, P., and Gutman, M. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 85908595
155. Lehninger, L. A. (1972) in International
Symposium of Biochemistry and Biophysics of
Mitochondrial Membranes (Azzone, G. F., ed),
p. 1, Academic Press, New York
156. Kalckar, H. M. (1969) Biological Phosphoryla-
tions, Prentice-Hall, Englewood Cliffs, New
Jersey
157. Ernster, L. (1993) FASEB J. 7, 15201524
158. Lemasters, J. J. (1984) J. Biol. Chem. 259,
1312313130
159. Flatt, J. P., Pahud, P., Ravussin, E., and Jquier,
E. (1984) Trends Biochem. Sci. 9, 466468
160. Hinkle, P. C., Kumar, M. A., Resetar, A., and
Harris, D. L. (1991) Biochemistry 30, 3576
3582
161. Ferguson, S. J. (1986) Trends Biochem. Sci. 11,
351353
162. Berry, E. A., and Hinkle, P. C. (1983) J. Biol.
Chem. 258, 14741486
163. Chance, B., and Williams, G. R. (1956) Adv.
Enzymol. 17, 65134
164. Wilson, D. F., Erecinska, M., and Dutton, P. L.
(1974) Annu Rev Biophys Bioeng. 3, 203230
165. Veech, R. L., Lawson, J. W. R., Cornell, N. W.,
and Krebs, H. A. (1979) J. Biol. Chem. 254,
65386547
166. Lardy, H. A., and Ferguson, S. M. (1969) Ann.
Rev. Biochem. 38, 991 1034
167. Metzler, D. E. (1977) Biochemistry; The
Chemical Reactions of Living Cells, Academic
Press, New York (pp. 598-600)
168. Mitchell, P. (1961) Nature (London) 191, 144148
169. Mitchell, P. (1968) Chemiosmotic Coupling and
Energy Transduction, Glynn Res., Bodmin.,
Cornwall, England
170. Hinkle, P. C., and McCarty, R. E. (1978) Sci.
Am. 238(Mar), 104123
171. Skulachev, V. P., and Hinckle, P. C., eds. (1981)
Chemiosmotic Protein Circuits in Biological
Membranes, Addison-Wesley, Reading,
Massachusetts
172. Nicholls, D. G., and Ferguson, S. J. (1992)
Bioenergetics 2, Academic Press, London
173. Mitchell, P. (1966) Biol. Rev. Cambridge Phil.
Soc. 41, 445502
174. Mitchell, P. (1979) Science 206, 11481159
175. Mitchell, P. (1978) Trends Biochem. Sci. 3, N58
N61
175a. Williams, R. J. P., and Prebble, J. (2002) Trends
Biochem. Sci. 27, 393395
176. Lowe, A. G., and Jones, M. N. (1984) Trends
Biochem. Sci. 9, 1113
177. Garland, P. (1978) Nature (London) 276, 89
178. Mitchell, P., and Moyle, J. (1969) Eur. J.
Biochem. 7, 471484
179. Wilson, D. F. (1980) in Membrane Structure and
Function, Vol. 1 (Bittar, E. E., ed), Wiley, New
York (pp. 153195)
180. Waddell, W. J., and Butler, T. C. (1959) J. Clin.
Invest. 38, 770
181. Dodgson, S. J., Forster, R. E., II, and Storey, B.
T. (1982) J. Biol. Chem. 257, 17051711
182. Bowman, C., and Tedeschi, H. (1979) Nature
(London) 280, 597598
183. Kinnally, K. W., Tedeschi, H., and Maloff, B. L.
(1978) Biochemistry 17, 34193427
184. Tedeschi, H. (1980) Trends Biochem. Sci. 5,
VIII IX
References
Ch 18IND page - E 2/14/03, 11:11 AM 1078
1079
212. Possmayer, F. E., and Grber, P. (1994) J. Biol.
Chem. 269, 18961904
213. Sokolov, M., and Gromet-Elhanan, Z. (1996)
Biochemistry 35, 12421248
213a. Groth, G., and Pohl, E. (2001) J. Biol. Chem.
276, 13451352
214. Hammes, G. G. (1983) Trends Biochem. Sci. 8,
131134
214a. Noji, H., and Yoshida, M. (2001) J. Biol. Chem.
276, 16651668
214b. Schnitzer, M. J. (2001) Nature (London) 410,
878881
214c. Stock, D., Leslie, A. G. W., and Walker, J. E.
(1999) Science 286, 17001705
215. Belogrudov, G. I., Tomich, J. M., and Hatefi, Y.
(1996) J. Biol. Chem. 271, 2034020345
216. Collinson, I. R., van Raaij, M. J., Runswick, M.
J., Fearnley, I. M., Skehel, J. M., Orriss, G. L.,
Miroux, B., and Walker, J. E. (1994) J. Mol. Biol.
242, 408421
217. Golden, T. R., and Pedersen, P. L. (1998)
Biochemistry 37, 1387113881
218. Joshi, S., Cao, G. J., Nath, C., and Shah, J.
(1997) Biochemistry 36, 1093610943
219. Uhlin, U., Cox, G. B., and Guss, J. M. (1997)
Structure 5, 12191230
220. Gabellieri, E., Strambini, G. B., Baracca, A., and
Solaini, G. (1997) Biophys. J. 72, 18181827
221. Gordon-Smith, D. J., Carbajo, R. J., Yang, J.-C.,
Videler, H., Runswick, M. J., Walker, J. E., and
Neuhaus, D. (2001) J. Mol. Biol. 308, 325339
222. Capaldi, R. A., Aggeler, R., Turina, P., and
Wilkens, S. (1994) Trends Biochem. Sci. 19, 284
289
222a. Capaldi, R. A., and Aggeler, R. (2002) Trends
Biochem. Sci. 27, 154160
223. van Raaij, M. J., Abrahams, J. P., Leslie, A. G.
W., and Walker, J. E. (1996) Proc. Natl. Acad.
Sci. U.S.A. 93, 69136917
224. Abrahams, J. P., Buchanan, S. K., van Raaij, M.
J., Fearnley, I. M., Leslie, A. G. W., and Walker,
J. E. (1996) Proc. Natl. Acad. Sci. U.S.A. 93,
94209424
225. Wilkens, S., and Capaldi, R. A. (1998) J. Biol.
Chem. 273, 2664526651
226. Bulygin, V. V., Duncan, T. M., and Cross, R. L.
(1998) J. Biol. Chem. 273, 3176531769
227. Pan, W., Ko, Y. H., and Pedersen, P. L. (1998)
Biochemistry 37, 69116923
227a. Tsunoda, S. P., Rodgers, A. J. W., Aggeler, R.,
Wilce, M. C. J., Yoshida, M., and Capaldi, R.
A. (2001) Proc. Natl. Acad. Sci. U.S.A. 98, 6560
6564
228. Rodgers, A. J. W., Wilkens, S., Aggeler, R.,
Morris, M. B., Howitt, S. M., and Capaldi, R.
A. (1997) J. Biol. Chem. 272, 3105831064
229. Sawada, K., Kuroda, N., Watanabe, H.,
Moritani-Otsuka, C., and Kanazawa, H.
(1997) J. Biol. Chem. 272, 3004730053
230. Bttcher, B., Schwarz, L., and Grber, P. (1998)
J. Mol. Biol. 281, 757762
230a. McLachlin, D. T., Coveny, A. M., Clark, S. M.,
and Dunn, S. D. (2000) J. Biol. Chem. 275,
1757117577
230b. Wilkens, S., Zhou, J., Nakayama, R., Dunn, S.
D., and Capaldi, R. A. (2000) J. Mol. Biol. 295,
387391
231. Long, J. C., Wang, S., and Vik, S. B. (1998) J.
Biol. Chem. 273, 1623516240
232. Jiang, W., and Fillingame, R. H. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 66076612
233. Girvin, M. E., Rastogi, V. K., Abildgaard, F.,
Markley, J. L., and Fillingame, R. H. (1998)
Biochemistry 37, 88178824
234. Jones, P. C., Jiang, W., and Fillingame, R. H.
(1998) J. Biol. Chem. 273, 1717817185
235. Groth, G., Tilg, Y., and Schirwitz, K. (1998) J.
Mol. Biol. 281, 4959
References
236. Jones, P. C., and Fillingame, R. H. (1998) J.
Biol. Chem. 273, 2970129705
236a. Dmitriev, O. Y., Abildgaard, F., Markley, J. L.,
and Fillingame, R. H. (2002) Biochemistry 41,
55375547
237. Zhang, Y., and Fillingame, R. H. (1995) J. Biol.
Chem. 270, 8793
238. Kaim, G., Matthey, U., and Dimroth, P. (1998)
EMBO J. 17, 688695
239. Kaim, G., Wehrle, F., Gerike, U., and Dimroth,
P. (1997) Biochemistry 36, 91859194
240. Kaim, G., and Dimroth, P. (1998) EMBO J. 17,
58875895
240a. Kaim, G., and Dimroth, P. (1999) EMBO J. 18,
41184127
240b. Dimroth, P., Wang, H., Grabe, M., and Oster,
G. (1999) Proc. Natl. Acad. Sci. U.S.A. 96, 4924
4929
240c. Aufurth, S., Schgger, H., and Mller, V.
(2000) J. Biol. Chem. 275, 3329733301
241. Vik, S. B., and Antonio, B. J. (1994) J. Biol.
Chem. 269, 3036430369
241a. Nijtmans, L. G. J., Henderson, N. S., Attardi,
G., and Holt, I. J. (2001) J. Biol. Chem. 276,
67556762
242. Sorgen, P. L., Bubb, M. R., McCormick, K. A.,
Edison, A. S., and Cain, B. D. (1998)
Biochemistry 37, 923932
243. Dunn, S. D., and Chandler, J. (1998) J. Biol.
Chem. 273, 86468651
244. McLachlin, D. T., Bestard, J. A., and Dunn, S.
D. (1998) J. Biol. Chem. 273, 1516215168
245. Boyer, P. D. (1993) Biochim. Biophys. Acta. 1140,
215250
246. Zhou, J.-M., and Boyer, P. D. (1993) J. Biol.
Chem. 268, 15311538
247. Boyer, P. D. (1995) FASEB J. 9, 559561
248. Boyer, P. D. (1997) Ann. Rev. Biochem. 66, 717
749
249. Smith, L. T., Rosen, G., and Boyer, P. D. (1983)
J. Biol. Chem. 258, 1088710894
250. Souid, A.-K., and Penefsky, H. S. (1995) J. Biol.
Chem. 270, 90749082
251. Penefsky, H. S., and Cross, R. L. (1991) Adv.
Enzymol. 64, 173214
252. Cox, G. B., Jans, D. A., Fimmel, A. L., Gibson,
F., and Hatch, L. (1984) Biochim. Biophys. Acta.
768, 201208
253. Aggeler, R., Ogilvie, I., and Capaldi, R. A.
(1997) J. Biol. Chem. 272, 1962119624
254. Boekema, E. J., Ubbink-Kok, T., Lolkema, J. S.,
Brisson, A., and Konings, W. N. (1997) Proc.
Natl. Acad. Sci. U.S.A. 94, 1429114293
255. Sabbert, D., Engelbrecht, S., and Junge, W.
(1996) Nature (London) 381, 623625
256. Sabbert, D., Engelbrecht, S., and Junge, W.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 44014405
257. Noji, H. (1998) Science 282, 18441845
258. Noji, H., Yasuda, R., Yoshida, M., and
Kinosita, K., Jr. (1997) Nature (London) 386,
299302
259. Kato-Yamada, Y., Noji, H., Yasuda, R.,
Kinosita, K., Jr., and Yoshida, M. (1998) J. Biol.
Chem. 273, 1937519377
260. Berg, H. C. (1998) Nature (London) 394, 324325
261. Yasuda, R., Noji, H., Kinosita, K. J., Jr., and
Yoshida, M. (1998) Cell 93, 11171124
262. Kinosita, K. J., Jr., Yasuda, R., Noji, H., Ishiwata,
S., and Yoshida, S. (1998) Cell 93, 2124
262a. Yasuda, R., Noji, H., Yoshida, M., Kinosita, K.,
Jr., and Itoh, H. (2001) Nature (London) 410,
898904
262b. Sambongi, Y., Iko, Y., Tanabe, M., Omote, H.,
Iwamoto-Kihara, A., Ueda, I., Yanagida, T.,
Wada, Y., and Futai, M. (1999) Science 286,
17221724
262c. Hutcheon, M. L., Duncan, T. M., Ngai, H., and
Cross, R. L. (2001) Proc. Natl. Acad. Sci. U.S.A.
98, 85198524
262d. Nishio, K., Iwamoto-Kihara, A., Yamamoto, A.,
Wada, Y., and Futai, M. (2002) Proc. Natl. Acad.
Sci. U.S.A. 99, 1344813452
263. Ko, Y. H., Bianchet, M., Amzel, L. M., and
Pedersen, P. L. (1997) J. Biol. Chem. 272,
1887518881
264. Dou, C., Grodsky, N. B., Matsui, T., Yoshida,
M., and Allison, W. S. (1997) Biochemistry 36,
37193727
265. Grodsky, N. B., Dou, C., and Allison, W. S.
(1998) Biochemistry 37, 10071014
266. Dou, C., Fortes, P. A. G., and Allison, W. S.
(1998) Biochemistry 37, 1675716764
267. Wang, H., and Oster, G. (1998) Nature
(London) 396, 279282
267a. Rastogi, V. K., and Girvin, M. E. (1999) Nature
(London) 402, 263268
267b. Jones, P. C., Hermolin, J., Jiang, W., and
Fillingame, R. H. (2000) J. Biol. Chem. 275,
3134031346
267c. Le, N. P., Omote, H., Wada, Y., Al-Shawi, M.
K., Nakamoto, R. K., and Futai, M. (2000)
Biochemistry 39, 27782783
267d. Ko, Y. H., Hong, S., and Pedersen, P. L. (1999)
J. Biol. Chem. 274, 2885328856
268. Kobayashi, H., Suzuki, T., and Unemoto, T.
(1986) J. Biol. Chem. 261, 627630
269. Forgac, M. (1999) J. Biol. Chem. 274, 12951
12954
269a. Oka, T., Toyomura, T., Honjo, K., Wada, Y.,
and Futai, M. (2001) J. Biol. Chem. 276, 33079
33085
269b. Mller, M. L., Jensen, M., and Taiz, L. (1999)
J. Biol. Chem. 274, 1070610716
269c. Kawamura, Y., Arakawa, K., Maeshima, M.,
and Yoshida, S. (2000) J. Biol. Chem. 275, 6515
6522
270. Oka, T., Yamamoto, R., and Futai, M. (1998)
J. Biol. Chem. 273, 2257022576
271. van Hille, B., Richener, H., Evans, D. B.,
Green, J. R., and Bilbe, G. (1993) J. Biol. Chem.
268, 70757080
272. Nelson, H., Mandiyan, S., and Nelson, N.
(1994) J. Biol. Chem. 269, 2415024155
272a. Wilkens, S., Vasilyeva, E., and Forgac, M.
(1999) J. Biol. Chem. 274, 3180431810
272b. Boekema, E. J., van Breemen, J. F. L., Brisson,
A., Ubbink-Kok, T., Konings, W. N., and
Lolkema, J. S. (1999) Nature (London) 401, 3738
272c. Sagermann, M., Stevens, T. H., and Matthews,
B. W. (2001) Proc. Natl. Acad. Sci. U.S.A. 98,
71347139
273. Steinert, K., Wagner, V., Kroth-Pancic, P. G.,
and Bickel-Sandktter, S. (1997) J. Biol. Chem.
272, 62616269
273a. Yokoyama, K., Muneyuki, E., Amano, T.,
Mizutani, S., Yoshida, M., Ishida, M., and
Ohkuma, S. (1998) J. Biol. Chem. 273, 20504
20510
274. Zhen, R.-G., Kim, E. J., and Rea, P. A. (1997)
J. Biol. Chem. 272, 2234022348
274a. Scott, D. A., and Docampo, R. (2000) J. Biol.
Chem. 275, 2421524221
274b. Miranda, M., Allen, K. E., Pardo, J. P., and
Slayman, C. W. (2001) J. Biol. Chem. 276,
2248522490
275. Pederson, P. L., and Carafoli, E. (1987) Trends
Biochem. Sci. 12, 146150
275a. Khlbrandt, W., Zeelen, J., and Dietrich, J.
(2002) Science 297, 16921696
276. Lambrecht, N., Corbett, Z., Bayle, D., Karlish,
S. J. D., and Sachs, G. (1998) J. Biol. Chem. 273,
1371913728
277. Asano, S., Tega, Y., Konishi, K., Fujioka, M.,
and Takeguchi, N. (1996) J. Biol. Chem. 271,
27402745
278. Melle-Milovanovic, D., Milovanovic, M.,
Nagpal, S., Sachs, G., and Shin, J. M. (1998)
J. Biol. Chem. 273, 1107511081
Ch 18IND page - E 2/14/03, 11:11 AM 1079
1080 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
329a. Richardson, D., and Sawers, G. (2002) Science
295, 18421843
329b. Jormakka, M., Trnroth, S., Byrne, B., and
Iwata, S. (2002) Science 295, 18631868
330. Iuchi, S., and Lin, E. C. C. (1987) Proc. Natl.
Acad. Sci. U.S.A. 84, 39013905
331. Spiro, S., and Guest, J. R. (1991) Trends
Biochem. Sci. 16, 310314
332. Cecchini, G., Thompson, C. R., Ackrell, B. A.
C., Westenberg, D. J., Dean, N., and Gunsalus,
R. P. (1986) Proc. Natl. Acad. Sci. U.S.A. 83,
88988902
332a. Turner, K. L., Doherty, M. K., Heering, H. A.,
Armstrong, F. A., Reid, G. A., and Chapman,
S. K. (1999) Biochemistry 38, 33023309
333. Czjzek, M., Dos Santos, J.-P., Pommier, J.,
Giordano, G., Mjean, V., and Haser, R. (1998)
J. Mol. Biol. 284, 435447
334. Trieber, C. A., Rothery, R. A., and Weiner, J. H.
(1994) J. Biol. Chem. 269, 71037109
335. Rothery, R. A., and Weiner, J. H. (1996)
Biochemistry 35, 32473257
336. Costa, C., Moura, J. J. G., Moura, I., Liu, M. Y.,
Peck, H. D., Jr., LeGall, J., Wang, Y., and
Huynh, B. H. (1990) J. Biol. Chem. 265, 14382
14387
337. Blackmore, R. S., Gibson, Q. H., and Greenwood,
C. (1992) J. Biol. Chem. 267, 1095010955
338. Ferguson, S. J. (1987) Trends Biochem. Sci. 12,
353357
339. Wang, Y., and Averill, B. A. (1996) J. Am.
Chem. Soc. 118, 39723973
339a. Suharti, Strampraad, M. J. F., Schrder, I., and
de Vries, S. (2001) Biochemistry 40, 26322639
340. Nurizzo, D., Cutruzzol, F., Arese, M., Bourgeois,
D., Brunori, M., Cambillau, C., and Tegoni, M.
(1998) Biochemistry 37, 1398713996
341. Kukimoto, M., Nishiyama, M., Tanokura, M.,
Adman, E. T., and Horinouchi, S. (1996) J.
Biol. Chem. 271, 1368013683
342. Murphy, M. E. P., Turley, S., and Adman, E. T.
(1997) J. Biol. Chem. 272, 2845528460
342a. Peters-Libeu, C. A., Kukimoto, M.,
Nishiyama, M., Horinouchi, S., Adman, E. T.
(1997) Biochemistry 36, 13160 13179
342b. Inoue, T., Nishio, N., Suzuki, S., Kataoka, K.,
Kohzuma, T., and Kai, Y. (1999) J. Biol. Chem.
274, 17845 17852
343. Weeg-Aerssens, E., Wu, W., Ye, R. W., Tiedje,
J. M., and Chang, C. K. (1991) J. Biol. Chem.
266, 74967502
344. Williams, P. A., Flp, V., Garman, E. F.,
Saunders, N. F. W., Ferguson, S. J., and Hajdu,
J. (1997) Nature (London) 389, 406412
345. Baker, S. C., Saunders, N. F. W., Willis, A. C.,
Ferguson, S. J., Hajdu, J., and Flp, V. (1997)
J. Mol. Biol. 269, 440455
346. Nurizzo, D., Silvestrini, M.-C., Mathieu, M.,
Cutruzzol, F., Bourgeois, D., Flp, V.,
Hajdu, J., Brunori, M., Tegoni, M., and
Cambillau, C. (1997) Structure 5, 11571171
346a. Cutruzzol, F., Brown, K., Wilson, E. K.,
Bellelli, A., Arese, M., Tegoni, M., Cambillau,
C., and Brunori, M. (2001) Proc. Natl. Acad.
Sci. U.S.A. 98, 22322237
346b. Sjgren, T., Svensson-EK, M., Hajdu, J., and
Brzezinki, P. (2000) Biochemistry 39, 1096710974
346c. Einsle, O., Messerschmidt, A., Stach, P.,
Bourenkov, G. P., Bartunik, H. D., Huber, R.,
and Kroneck, P. M. H. (1999) Nature (London)
400, 476480
346d. Einsle, O., Stach, P., Messerschmidt, A.,
Simon, J., Krger, A., Huber, R., and Kroneck,
P. M. H. (2000) J. Biol. Chem. 275, 3960839616
346e. Sjgren, T., and Hajdu, J. (2001) J. Biol. Chem.
276, 2945029455
346f. Ranghino, G., Scorza, E., Sjgren, T., Williams,
P. A., Ricci, M., and Hajdu, J. (2000)
Biochemistry 39, 1095810966
279. Wolfe, M. M., and Soll, A. H. (1988) N. Engl.
J. Med. 319, 17071715
280. Priver, N. A., Rabon, E. C., and Zeidel, M. L.
(1993) Biochemistry 32, 24592468
281. Nicholls, D. G., and Ferguson, S. J. (1992)
Bioenergetics 2, Academic Press, London (p. 139)
282. Levings, C. S., III. (1990) Science 250, 942947
283. Hatefi, Y., and Yamaguchi, M. (1996) FASEB J.
10, 444452
284. Glavas, N. A., Hou, C., and Bragg, P. D. (1995)
Biochemistry 34, 76947702
285. Meuller, J., and Rydstrm, J. (1999) J. Biol.
Chem. 274, 1907219080
286. Grimley, R. L., Quirk, P. G., Bizouarn, T.,
Thomas, C. M., and Jackson, J. B. (1997)
Biochemistry 36, 1476214770
286a. Venning, J. D., Rodrigues, D. J., Weston, C. J.,
Cotton, N. P. J., Quirk, P. G., Errington, N.,
Finet, S., White, S. A., and Jackson, J. B. (2001)
J. Biol. Chem. 276, 3067830685
287. Fjellstrm, O., Axelsson, M., Bizouarn, T., Hu,
X., Johansson, C., Meuller, J., and Rydstrm, J.
(1999) J. Biol. Chem. 274, 63506359
288. Lee, A.-C., Zizi, M., and Colombini, M. (1994)
J. Biol. Chem. 269, 3097430980
289. Rostovtseva, T., and Colombini, M. (1996) J.
Biol. Chem. 271, 2800628008
290. Rostovtseva, T., and Colombini, M. (1997)
Biophys. J. 72, 19541962
291. Beavis, A. D., Lu, Y., and Garlid, K. D. (1993)
J. Biol. Chem. 268, 9971004
292. Li, W., Shariat-Madar, Z., Powers, M., Sun, X.,
Lane, R. D., and Garlid, K. D. (1992) J. Biol.
Chem. 267, 1798317989
293. Cox, D. A., and Matlib, M. A. (1993) J. Biol.
Chem. 268, 938947
294. Sparagna, G. C., Gunter, K. K., Sheu, S.-S.,
and Gunter, T. E. (1995) J. Biol. Chem. 270,
2751027515
295. Bisaccia, F., Capobianco, L., Brandolin, G., and
Palmieri, F. (1994) Biochemistry 33, 37053713
296. Fiermonte, G., Palmieri, L., Dolce, V., Lasorsa,
F. M., Palmieri, F., Runswick, M. J., and Walker,
J. E. (1998) J. Biol. Chem. 273, 2475424759
297. Nelson, D. R., Felix, C. M., and Swanson, J. M.
(1998) J. Mol. Biol. 277, 285308
298. Kaplan, R. S., Mayor, J. A., and Wood, D. O.
(1993) J. Biol. Chem. 268, 1368213690
298a. Xu, Y., Kakhniashvili, D. A., Gremse, D. A.,
Wood, D. O., Mayor, J. A., Walters, D. E., and
Kaplan, R. S. (2000) J. Biol. Chem. 275, 7117
7124
298b. Bandell, M., and Lolkema, J. S. (2000) J. Biol.
Chem. 275, 3913039136
299. Mller, V., Basset, G., Nelson, D. R., and
Klingenberg, M. (1996) Biochemistry 35,
1613216143
300. Broustovetsky, N., Bamberg, E., Gropp, T.,
and Klingenberg, M. (1997) Biochemistry 36,
1386513872
300a. Wiedemann, N., Pfanner, N., and Ryan, M. T.
(2001) EMBO J. 20, 951960
300b. Heimpel, S., Basset, G., Odoy, S., and
Klingenberg, M. (2001) J. Biol. Chem. 276,
1149911506
301. Beyer, K., and Klingenberg, M. (1985)
Biochemistry 24, 38213826
302. Muller, M., Krebs, J. J. R., Cherry, R. J., and
Kawato, S. (1984) J. Biol. Chem. 259, 30373043
303. Ziegler, M., and Penefsky, H. S. (1993) J. Biol.
Chem. 268, 25320 25328
304. Schroers, A., Burkovski, A., Wohlrab, H., and
Krmer, R. (1998) J. Biol. Chem. 273, 14269
14276
305. Briggs, C., Mincone, L., and Wohlrab, H.
(1999) Biochemistry 38, 50965102
305a. Majima, E., Ishida, M., Miki, S., Shinohara, Y.,
and Terada, H. (2001) J. Biol. Chem. 276, 9792
9799
306. Kaplan, R. S., Pratt, R. D., and Pederson, P. L.
(1986) J. Biol. Chem. 261, 1276712773
307. Nicolli, A., Petronilli, V., and Bernardi, P.
(1993) Biochemistry 32, 44614465
307a. Scorrano, L., Petronilli, V., Colonna, R., Di
Lisa, F., and Bernardi, P. (1999) J. Biol. Chem.
274, 2465724663
308. Liu, G., Hinch, B., Davatol-Hag, H., Lu, Y.,
Powers, M., and Beavis, A. D. (1996) J. Biol.
Chem. 271, 1971719723
309. Sugano, T., Handler, J. A., Yoshihara, H.,
Kizaki, Z., and Thurman, R. G. (1990) J. Biol.
Chem. 265, 2154921553
310. Schlegel, H. G., and Eberhardt, U. (1972) Adv.
Microbiol. Physiol. 7, 205242
310a. Pierik, A. J., Roseboom, W., Happe, R. P.,
Bagley, K. A., and Albracht, S. P. J. (1999) J.
Biol. Chem. 274, 33313337
311. Bothe, H., and Trebst, A., eds. (1981) Biology of
Inorganic Nitrogen and Sulfur, Springer, Berlin
312. Arciero, D. M., Golombek, A., Hendrich, M.
P., and Hooper, A. B. (1998) Biochemistry 37,
523529
313. Igarashi, N., Moriyama, H., Fujiwara, T.,
Fukumori, Y., and Tanaka, N. (1997) Nature
Struct. Biol. 4, 276284
313a. Schalk, J., de Vries, S., Kuenen, J. G., and Jetten,
M. S. M. (2000) Biochemistry 39, 54055412
314. Hendrich, M. P., Petasis, D., Arciero, D. M.,
and Hooper, A. B. (2001) J. Am. Chem. Soc. 123,
29973005
314a. Hendrich, M. P., Upadhyay, A. K., Riga, J.,
Arciero, D. M., and Hooper, A. B. (2002)
Biochemistry 41, 46034611
315. Blaut, M., and Gottschalk, G. (1997) in
Bioenergetics (Grber, P., and Milazzo, G., eds),
pp. 139 211, Birkhuser Verlag, Basel
316. DiSpirito, A. A., and Hooper, A. B. (1986)
J. Biol. Chem. 261, 1053410537
317. Logan, M. S. P., and Hooper, A. B. (1995)
Biochemistry 34, 92579264
318. Taylor, C. D., and Wirsen, C. O. (1997) Science
277, 14831485
319. Felbeck, H., and Somero, G. N. (1982) Trends
Biochem. Sci. 7, 201204
320. Gaill, F. (1993) FASEB J. 7, 558565
321. Trudinger, P. A. (1969) Ad. Microbiol. Physiol. 3,
111158
322. Roy, A. B., and Trudinger, P. A. (1970) The
Biochemistry of Inorganic Compounds of Sulfur,
Cambridge Univ. Press, London and New York
322a. Cheesman, M. R., Little, P. J., and Berks, B. C.
(2001) Biochemistry 40, 1056210569
323. Postgate, J. R., and Kelly, D. P., eds. (1982)
Sulphur Bacteria, The Royal Society,
323a. Griesbeck, C., Schtz, M., Schdl, T., Bathe, S.,
Nausch, L., Mederer, N., Vielreicher, M., and
Hauska, G. (2002) Biochemistry 41, 1155211565
324. Chen, Z.-w, Koh, M., Van Driessche, G., Van
Beeumen, J. J., Bartsch, R. G., Meyer, T. E.,
Cusanovich, M. A., and Mathews,
F. S. (1994) Science 266, 430432
324a. Edwards, K. J., Bond, P. L., Gihring, T. M., and
Banfield, J. F. (2000) Science 287, 17961799
325. Hodgman, C. D., ed. (19671968) CRC
Handbook of Chemistry and Physics, 48th ed.,
Chem. Rubber Publ. Co., Cleveland, Ohio
326. Dugan, P. R. (1972) Biochemical Ecology of
Water Pollution, Plenum, New York
327. Powell, M. A., and Somero, G. N. (1986)
Science 233, 563566
328. Rothery, R. A., Blasco, F., and Weiner, J. H.
(2001) Biochemistry 40, 52605268
328a. Anderson, L. J., Richardson, D. J., and Butt, J.
N. (2001) Biochemistry 40, 1129411307
329. Magalon, A., Asso, M., Guigliarelli, B.,
Rothery, R. A., Bertrand, P., Giordano, G., and
Blasco, F. (1998) Biochemistry 37, 73637370
References
Ch 18IND page - E 2/14/03, 11:11 AM 1080
1081
346g. Kobayashi, K., Koppenhfer, A., Ferguson, S.
J., Watmough, N. J., and Tagawa, S. (2001)
Biochemistry 40, 85428547
347. Kobayashi, K., Koppenhfer, A., Ferguson, S.
J., and Tagawa, S. (1997) Biochemistry 36,
1361113616
348. Cheesman, M. R., Ferguson, S. J., Moir, J. W. B.,
Richardson, D. J., Zumft, W. G., and Thomson,
A. J. (1997) Biochemistry 36, 1626716276
349. Sakurai, N., and Sakurai, T. (1997) Biochemis-
try 36, 1380913815
350. Hendriks, J., Warne, A., Gohlke, U., Haltia, T.,
Ludovici, C., Lbben, M., and Saraste, M.
(1998) Biochemistry 37, 1310213109
351. Monne-Loccoz, P., and de Vries, S. (1998) J.
Am. Chem. Soc. 120, 51475152
352. Shiro, Y., Fujii, M., Isogai, Y., Adachi, S.-i,
Iizuka, T., Obayashi, E., Makino, R.,
Nakahara, K., and Shoun, H. (1995)
Biochemistry 34, 90529058
353. Neese, F., Zumft, W. G., Antholine, W. E., and
Kroneck, P. M. H. (1996) J. Am. Chem. Soc. 118,
86928699
354. Farrar, J. A., Zumft, W. G., and Thomson, A. J.
(1998) Proc. Natl. Acad. Sci. U.S.A. 95, 9891
9896
355. Postgate, J. R., FRS. (1984) in The Sulfate-
Reducing Bacteria, 2nd ed., pp. 5663,
Cambridge University Press, Cambridge
356. Postgate, J. R. (1984) The Sulfate-reducing Bacteria,
2nd ed., Cambridge Univ. Press, London
356a. Michaelis, W., and 16 other authors. (2002)
Science 297, 10131015
357. Grey, D. C., and Jensen, M. L. (1972) Science
177, 10991100
358. Gavel, O. Y., Bursakov, S. A., Calvete, J. J.,
George, G. N., Moura, J. J. G., and Moura, I.
(1998) Biochemistry 37, 1622516232
359. Klenk, H.-P., and 50 other authors. (1997)
Nature (London) 390, 364370
360. Morais, J., Palma, P. N., Frazao, C., Caldeira,
J., LeGall, J., Moura, I., Moura, J. J. G., and
Carrondo, M. A. (1995) Biochemistry 34,
1283012841
361. Louro, R. O., Catarino, T., Turner, D. L.,
Picarra-Pereira, M. A., Pacheco, I., LeGall, J.,
and Xavier, A. V. (1998) Biochemistry 37,
1580815815
361a. Fritz, G., Griesshaber, D., Seth, O., and Kroneck,
P. M. H. (2001) Biochemistry 40, 13171324
362. Florens, L., Ivanova, M., Dolla, A., Czjzek, M.,
Haser, R., Verger, R., and Bruschi, M. (1995)
Biochemistry 34, 1132711334
363. Banci, L., Bertini, I., Bruschi, M., Sompornpisut,
P., and Turano, P. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1439614400
364. Feng, Y., and Swenson, R. P. (1997) Biochemis-
try 36, 1361713628
365. Lui, S. M., and Cowan, J. A. (1994) J. Am.
Chem. Soc. 116, 1153811549
366. Crane, B. R., Siegel, L. M., and Getzoff, E. D.
(1997) Biochemistry 36, 1210112119
367. Moura, I., LeGall, J., Lino, A. R., Peck, H. D.,
Jr., Fauque, G., Xavier, A. V., Der Vertanian, D.
V., Moura, J. J. G., and Huynh, B. H. (1988) J.
Am. Chem. Soc. 110, 10751082
368. Fischer, F., Zillig, W., Stetter, K. O., and Schreiber,
G. (1983) Nature (London) 301, 511515
369. Kelly, D. P. (1985) Nature (London) 313, 734
370. Bak, F., and Cypionka, H. (1987) Nature
(London) 326, 891892
370a. Morelli, X., Czjzek, M., Hatchikian, C. E.,
Bornet, O., Fontecilla-Camps, J. C., Palma, N.
P., Moura, J. J. G., and Guerlesquin, F. (2000) J.
Biol. Chem. 275, 2320423210
370b. Costas, A. M. C., White, A. K., and Metcalf, W.
W. (2001) J. Biol. Chem. 276, 1742917436
References
371. Mller, V., Blaut, M., and Gottschalk, G.
(1993) in Methanogenesis: Ecology, Physiology,
Biochemistry and Genetics (Ferry, J. G., ed), pp.
360406, Chapman and Hall, New York
372. Hayaishi, O., and Nozaki, M. (1969) Science
164, 389405
373. Orville, A. M., and Lipscomb, J. D. (1993) J.
Biol. Chem. 268, 85968607
374. Ohlendorf, D. H., Orville, A. M., and Lipscomb,
J. D. (1994) J. Mol. Biol. 244, 586608
375. Orville, A. M., Lipscomb, J. D., and Ohlendorf,
D. H. (1997) Biochemistry 36, 1005210066
376. Sanvoisin, J., Langley, G. J., and Bugg, T. D. H.
(1995) J. Am. Chem. Soc. 117, 78367837
377. Miller, M. A., and Lipscomb, J. D. (1996) J.
Biol. Chem. 271, 55245535
378. Han, S., Eltis, L. D., Timmis, K. N., Muchmore,
S. W., and Bolin, J. T. (1995) Science 270, 976
980
379. Shu, L., Chiou, Y.-M., Orville, A. M., Miller,
M. A., Lipscomb, J. D., and Que, L., Jr. (1995)
Biochemistry 34, 66496659
380. Senda, T., Sugiyama, K., Narita, H.,
Yamamoto, T., Kimbara, K., Fukuda, M., Sato,
M., Yano, K., and Mitsui, Y. (1996) J. Mol. Biol.
255, 735752
381. Hugo, N., Armengaud, J., Gaillard, J., Timmis,
K. N., and Jouanneau, Y. (1998) J. Biol. Chem.
273, 96229629
382. Fraser, M. S., and Hamilton, G. A. (1982) J.
Am. Chem. Soc. 104, 42034211
383. Leeds, J. M., Brown, P. J., McGeehan, G. M.,
Brown, F. K., and Wiseman, J. S. (1993) J. Biol.
Chem. 268, 1778117786
384. Ohnishi, T., Hirata, F., and Hayaishi, O. (1977)
J. Biol. Chem. 252, 46434647
385. Gassner, G. T., Ballou, D. P., Landrum, G. A.,
and Whittaker, J. W. (1993) Biochemistry 32,
48204825
386. Yamaguchi, M., and Fujisawa, H. (1982) J.
Biol. Chem. 257, 1249712502
386a. Senda, T., Yamada, T., Sakurai, N., Kubota,
M., Nishizaki, T., Masai, E., Fukuda, M., and
Mitsui, Y. (2000) J. Mol. Biol. 304, 397410
387. Gassner, G. T., Ludwig, M. L., Gatti, D. L.,
Correll, C. C., and Ballou, D. P. (1995) FASEB
J. 9, 14111418
387a. Coulter, E. D., Moon, N., Batie, C. J., Dunham,
W. R., and Ballou, D. P. (1999) Biochemistry 38,
1106211072
388. Bertrand, P., More, C., and Camensuli, P.
(1995) J. Am. Chem. Soc. 117, 18071809
389. Lockridge, O., Massey, V., and Sullivan, P. A.
(1972) J. Biol. Chem. 247, 80978106
390. Sanders, S. A., Williams, C. H., Jr., and Massey,
V. (1999) J. Biol. Chem. 274, 2228922295
391. Flashner, M. I. S., and Massey, V. (1974) J. Biol.
Chem. 249, 25792586
392. Emanuele, J. J., and Fitzpatrick, P. F. (1995)
Biochemistry 34, 37103715
393. Massey, V. (1994) J. Biol. Chem. 269, 22459
22462
394. Entsch, B., and van Berkel, W. J. H. (1995)
FASEB J. 9, 476483
395. Gatti, D. L., Entsch, B., Ballou, D. P., and
Ludwig, M. L. (1996) Biochemistry 35, 567578
396. Hamilton, G. A. (1971) Prog. Bioorg. Chem. 1,
83
397. Orf, H. W., and Dolphin, D. (1974) Proc. Natl.
Acad. Sci. U.S.A. 71, 26462650
398. Wagner, W. R., Spero, D. M., and Rastetter, W.
H. (1984) J. Am. Chem. Soc. 106, 14761480
399. Moran, G. R., Entsch, B., Palfey, B. A., and
Ballou, D. P. (1997) Biochemistry 36, 75487556
399a. Frederick, K. K., Ballou, D. P., and Palfey, B.
A. (2001) Biochemistry 40, 38913899
400. Eppink, M. H. M., Schreuder, H. A., and
van Berkel, W. J. H. (1998) J. Biol. Chem. 273,
2103121039
401. van der Bolt, F. J. T., van den Heuvel, R. H. H.,
Vervoort, J., and van Berkel, W. J. H. (1997)
Biochemistry 36, 1419214201
401a. Eppink, M. H. M., Overkamp, K. M.,
Schreuder, H. A., and Van Berkel, W. J. H.
(1999) J. Mol. Biol. 292, 8796
401b. Ortiz-Maldonado, M., Ballou, D. P., and
Massey, V. (2001) Biochemistry 40, 10911101
402. Eppink, M. H. M., Schreuder, H. A., and van
Berkel, W. J. H. (1997) Protein Sci. 6, 2454
2458
403. Maeda-Yorita, K., and Massey, V. (1993) J. Biol.
Chem. 268, 41344144
404. Einarsdottir, G. H., Stankovich, M. T., and Tu,
S.-C. (1988) Biochemistry 27, 32773285
405. Powlowski, J., Ballou, D. P., and Massey, V.
(1990) J. Biol. Chem. 265, 49694975
406. Mathews, F. S., Chen, Z.-w, and Bellamy, H.
D. (1991) Biochemistry 30, 238247
407. Arunachalam, U., Massey, V., and Miller, S.
M. (1994) J. Biol. Chem. 269, 150155
408. Prieto, M. A., and Garcia, J. L. (1994) J. Biol.
Chem. 269, 2282322829
409. Ysuji, H., Ogawa, T., Bando, N., Kimoto, M.,
and Sasaoka, K. (1990) J. Biol. Chem. 265,
1606416067
410. Itagaki, K., Carver, G. T., and Philpot, R. M.
(1996) J. Biol. Chem. 271, 2010220107
411. Jones, K. C., and Ballou, D. P. (1986) J. Biol.
Chem. 261, 25532559
411a. Sheng, D., Ballou, D. P., and Massey, V. (2001)
Biochemistry 40, 1115611167
412. Branchaud, B. P., and Walsh, C. T. (1985) J.
Am. Chem. Soc. 107, 21532161
413. Erlandsen, H., Flatmark, T., Stevens, R. C.,
and Hough, E. (1998) Biochemistry 37, 15638
15646
414. Loeb, K. E., Westre, T. E., Kappock, T. J., Mitic,
N., Glasfeld, E., Caradonna, J. P., Hedman, B.,
Hodgson, K. O., and Solomon, E. I. (1997) J.
Am. Chem. Soc. 119, 19011915
415. Chen, D., and Frey, P. A. (1998) J. Biol. Chem.
273, 2559425601
415a. Teigen, K., Froystein, N. ., and Martnez, A.
(1999) J. Mol. Biol. 294, 807823
416. Goodwill, K. E., Sabatier, C., and Stevens, R.
C. (1998) Biochemistry 37, 1343713445
417. Francisco, W. A., Tian, G., Fitzpatrick, P. F.,
and Klinman, J. P. (1998) J. Am. Chem. Soc. 120,
40574062
418. Michaud-Soret, I., Andersson, K. K., and Que,
L., Jr. (1995) Biochemistry 34, 55045510
419. Hillas, P. J., and Fitzpatrick, P. F. (1996)
Biochemistry 35, 69696975
419a. Alms, B., Toska, K., Teigen, K., Groehn, V.,
Pfleiderer, W., Martnez, A., Flatmark, T., and
Haavik, J. (2000) Biochemistry 39, 1367613686
420. Moran, G. R., Daubner, S. C., and Fitzpatrick,
P. F. (1998) J. Biol. Chem. 273, 1225912266
421. Ramsey, A. J., Hillas, P. J., and Fitzpatrick, P. F.
(1996) J. Biol. Chem. 271, 2439524400
421a. Fitzpatrick, P. F. (1999) Ann. Rev. Biochem. 68,
355381
422. Lazarus, R. A., DeBrosse, C. W., and
Benkovic, S. J. (1982) J. Am. Chem. Soc. 104,
68696871
423. Pike, D. C., Hora, M. T., Bailey, S. W., and
Ayling, J. E. (1986) Biochemistry 25, 47624771
424. Dix, T. A., Bollag, G. E., Domanico, P. L., and
Benkovic, S. J. (1985) Biochemistry 24, 2955
2958
425. Rebrin, I., Thny, B., Bailey, S. W., and Ayling,
J. E. (1998) Biochemistry 37, 1124611254
425a. Endrizzi, J. A., Cronk, J. D., Wang, W.,
Crabtree, G. R., and Alber, T. (1995) Science
268, 556559
426. Ficner, R., Sauer, U. H., Stier, G., and Suck, D.
(1995) EMBO J. 14, 20342042
Ch 18IND page - E 2/14/03, 11:11 AM 1081
1082 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
427. Su, Y., Varughese, K. I., Xuong, N. H., Bray, T.
L., Roche, D. J., and Whiteley, J. M. (1993) J.
Biol. Chem. 268, 2683626841
428. Varughese, K. I., Xuong, N. H., Kiefer, P. M.,
Matthews, D. A., and Whiteley, J. M. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 55825586
429. Kiefer, P. M., Grimshaw, C. E., and Whiteley, J.
M. (1997) Biochemistry 36, 94389445
430. Scriver, C. R., Kaufman, S., Eisensmith, R. C.,
and Woo, S. L. C. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 10151075, McGraw-
Hill, New York
430a. Kobe, B., Jennings, I. G., House, C. M.,
Michell, B. J., Goodwill, K. E., Santarsiero, B.
D., Stevens, R. C., Cotton, R. G. H., and
Kemp, B. E. (1999) Nature Struct. Biol. 6, 442
448
431. Ginns, E. L., Rehavi, M., Martin, B. M., Weller,
M., OMalley, K. L., LeMarca, M. E., McAllister,
C. G., and Paul, S. M. (1988) J. Biol. Chem. 263,
74067410
432. Grenett, H. E., Ledley, F. D., Reed, L. L., and
Woo, S. L. C. (1987) Proc. Natl. Acad. Sci.
U.S.A. 84, 55305534
433. Pigeon, D., Ferrara, P., Gros, F., and Thibault,
J. (1987) J. Biol. Chem. 262, 61556158
433a. Itagaki, C., Isobe, T., Taoka, M., Natsume, T.,
Nomura, N., Horigome, T., Omata, S.,
Ichinose, H., Nagatsu, T., Greene, L. A., and
Ichimura, T. (1999) Biochemistry 38, 15673
15680
434. McTigue, M., Cremins, J., and Halegoua, S.
(1985) J. Biol. Chem. 260, 90479056
435. Okuno, S., and Fujisawa, H. (1985) J. Biol.
Chem. 260, 26332635
436. Albert, K. A., Helmer-Matyjek, E., Nairn, A.
C., Muller, T. H., Waycock, J. W., Greene, L.
A., Goldstein, M., and Greengard, P. (1984)
Proc. Natl. Acad. Sci. U.S.A. 81, 77137717
437. Guroff, G., Daly, J. W., Jerina, D. M., Renson,
J., Witkop, B., and Udenfriend, S. (1967)
Science 157, 15241530
438. Kasperek, G. J., Bruice, T. C., Yagi, H.,
Kaubisch, N., and Jerina, D. M. (1972) J. Am.
Chem. Soc. 94, 78767882
439. Endo, F., Awata, H., Tanoue, A., Ishiguro, M.,
Eda, Y., Titani, K., and Matsuda, I. (1992) J.
Biol. Chem. 267, 2423524240
440. Bradley, F. C., Lindstedt, S., Lipscomb, J. D.,
Que, L., Jr., Roe, A. L., and Rundgren, M.
(1986) J. Biol. Chem. 261, 1169311696
441. McGinnis, K., Ku, G. M., VanDusen, W. J., Fu,
J., Garsky, V., Stern, A. M., and Friedman, P.
A. (1996) Biochemistry 35, 39573962
442. Lamberg, A., Pihlajaniemi, T., and Kivirikko,
K. I. (1995) J. Biol. Chem. 270, 99269931
443. Myllyharju, J., and Kivirikko, K. I. (1997)
EMBO J. 16, 11731180
444. Annunen, P., Autio-Harmainen, H., and
Kivirikko, K. I. (1998) J. Biol. Chem. 273, 5989
5992
445. Myllyl, R., Pihlajaniemi, T., Pajunen, L.,
Turpeenniemi-Hujanen, T., and Kivirikko, K.
I. (1991) J. Biol. Chem. 266, 28052810
446. Valtavaara, M., Szpirer, C., Szpirer, J., and
Myllyl, R. (1998) J. Biol. Chem. 273, 12881
12886
447. Bolwell, G. P., Robbins, M. P., and Dixon, R.
A. (1985) Biochem. J. 229, 693699
448. Thornburg, L. D., and Stubbe, J. (1993)
Biochemistry 32, 1403414042
449. Eichhorn, E., van der Ploeg, J. R., Kertesz, M.
A., and Leisinger, T. (1997) J. Biol. Chem. 272,
2303123036
449a. Elkins, J. M., Ryle, M. J., Clifton, I. J., Hotopp,
J. C. D., Lloyd, J. S., Burzlaff, N. I., Baldwin, J.
E., Hausinger, R. P., and Roach, P. L. (2002)
Biochemistry 41, 51855192
450. Whiting, A. K., Que, L., Jr., Saari, R. E.,
Hausinger, R. P., Frederick, M. A., and
McCracken, J. (1997) J. Am. Chem. Soc. 119,
34133414
451. Hotopp, J. C. D., and Hausinger, R. P. (2002)
Biochemistry 41, 97879794
452. Henderson, L. M., Nelson, P. J., and Henderson,
L. (1982) Fed. Proc. 41, 28432847
453. Englard, S., Blanchard, J. S., and Midelfort, C.
F. (1985) Biochemistry 24, 11101116
454. Myllyla, R., Majamaa, K., Gunzler, V.,
Hanauske-Abel, H. M., and Kivirikko, K. I.
(1984) J. Biol. Chem. 259, 54035405
455. Ho, R. Y. N., Mehn, M. P., Hegg, E. L., Liu, A.,
Ryle, M. J., Hausinger, R. P., and Que, L., Jr.
(2001) J. Am. Chem. Soc. 123, 50225029
456. Roach, P. L., Clifton, I. J., Flp, V., Harlos, K.,
Barton, G. J., Hajdu, J., Andersson, I.,
Schofield, C. J., and Baldwin, J. E. (1995)
Nature (London) 375, 700704
457. Roach, P. L., Clifton, I. J., Hensgens, C. M. H.,
Shibata, N., Schofield, C. J., Hajdu, J., and
Baldwin, J. E. (1997) Nature (London) 387, 827
830
457a. Lloyd, M. D., Lee, H.-J., Harlos, K., Zhang,
Z.-H., Baldwin, J. E., Schofield, C. J.,
Charnock, J. M., Garner, C. D., Hara, T.,
Terwisscha van Scheltinga, A. C., Valegrd,
K., Viklund, J. A. C., Hajdu, J., Andersson, I.,
Danielsson, ., and Bhikhabhai, R. (1999) J.
Mol. Biol. 287, 943960
458. Busby, R. W., Chang, M. D.-T., Busby, R. C.,
Wimp, J., and Townsend, C. A. (1995) J. Biol.
Chem. 270, 42624269
459. Zhou, J., Kelly, W. L., Bachmann, B. O.,
Gunsior, M., Townsend, C. A., and Solomon,
E. I. (2001) J. Am. Chem. Soc. 123, 73887398
460. Tian, G., Berry, J. A., and Klinman, J. P. (1994)
Biochemistry 33, 226234
461. Klinman, J. P., Krueger, M., Brenner, M., and
Edmondson, D. E. (1984) J. Biol. Chem. 259,
33993402
462. Merkler, D. J., Kulathila, R., Consalvo, A. P.,
Young, S. D., and Ash, D. E. (1992) Biochemis-
try 31, 72827288
463. Prigge, S. T., Kolhekar, A. S., Eipper, B. A.,
Mains, R. E., and Amzel, L. M. (1997) Science
278, 13001305
463a. Jaron, S., and Blackburn, N. J. (2001)
Biochemistry 40, 68676875
464. Francisco, W. A., Merkler, D. J., Blackburn, N.
J., and Klinman, J. P. (1998) Biochemistry 37,
82448252
464a. Jaron, S., and Blackburn, N. J. (1999)
Biochemistry 38, 1508615096
464b. Kolhekar, A. S., Bell, J., Shiozaki, E. N., Jin, L.,
Keutmann, H. T., Hand, T. A., Mains, R. E., and
Eipper, B. A. (2002) Biochemistry 41, 12384
12394
465. Wilkinson, B., Zhu, M., Priestley, N. D.,
Nguyen, H.-H. T., Morimoto, H., Williams, P.
G., Chan, S. I., and Floss, H. G. (1996) J. Am.
Chem. Soc. 118, 921922
466. Elliott, S. J., Zhu, M., Tso, L., Nguyen, H.-H.
T., Yip, J. H.-K., and Chan, S. I. (1997) J. Am.
Chem. Soc. 119, 99499955
467. Swinney, D. C., and Mak, A. Y. (1994)
Biochemistry 33, 21852190
468. Imai, T., Yamazaki, T., and Kominami, S.
(1998) Biochemistry 37, 80978104
469. Denison, M. S., and Whitlock, J. P., Jr. (1995) J.
Biol. Chem. 270, 1817518178
470. Coon, M. J., Ding, X., Pernecky, S. J., and Vaz,
A. D. N. (1992) FASEB J. 6, 669673
References
471. Gonzalez, F. J., and Lee, Y.-H. (1996) FASEB J.
10, 11121117
472. Nishimoto, M., Gotoh, O., Okuda, K., and
Noshiro, M. (1991) J. Biol. Chem. 266, 6467
6471
473. Dilworth, F. J., Scott, I., Green, A., Strugnell,
S., Guo, Y.-D., Roberts, E. A., Kremer, R.,
Calverley, M. J., Makin, H. L. J., and Jones, G.
(1995) J. Biol. Chem. 270, 1676616774
474. Cabello-Hurtado, F., Batard, Y., Salan, J.-P.,
Durst, F., Pinot, F., and Werck-Reichhart, D.
(1998) J. Biol. Chem. 273, 72607267
475. Meyer, K., Shirley, A. M., Cusumano, J. C.,
Bell-Lelong, D. A., and Chapple, C. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 66196623
476. Harris, D. L., and Loew, G. H. (1998) J. Am.
Chem. Soc. 120, 89418948
476a. French, K. J., Strickler, M. D., Rock, D. A.,
Rock, D. A., Bennett, G. A., Wahlstrom, J. L.,
Goldstein, B. M., and Jones, J. P. (2001)
Biochemistry 40, 95329538
476b. Schlichting, I., Berendzen, J., Chu, K., Stock,
A. M., Maves, S. A., Benson, D. E., Sweet, R.
M., Ringe, D., Petsko, G. A., and Sligar, S. G.
(2000) Science 287, 16151622
477. Vaz, A. D. N., McGinnity, D. F., and Coon, M.
J. (1998) Proc. Natl. Acad. Sci. U.S.A. 95, 3555
3560
478. Tzeng, H.-F., Laughlin, L. T., and Armstrong,
R. N. (1998) Biochemistry 37, 29052911
479. De Montellano, P. R. O., ed. (1995) Cytochrome
P450. Structure, Mechanism, and Biochemistry,
2nd ed., Plenum, New York
480. Coon, M. J., Vaz, A. D. N., and Bestervelt, L.
L. (1996) FASEB J. 10, 428434
481. Negishi, M., Uno, T., Darden, T. A., Sueyoshi,
T., and Pedersen, L. G. (1996) FASEB J. 10,
683689
482. Gilday, D., Gannon, M., Yutzey, K., Bader, D.,
and Rifkind, A. B. (1996) J. Biol. Chem. 271,
3305433059
483. Khani, S. C., Zophiropoulos, P. G., Fujita, V. S.,
Porter, T. D., Koop, D. R., and Coon, M. J.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 638642
483a. Pikuleva, I. A., Puchkaev, A., and Bjrkhem, I.
(2001) Biochemistry 40, 76217629
484. Porter, T. D. (1991) Trends Biochem. Sci. 16,
154158
485. Hubbard, P. A., Shen, A. L., Paschke, R.,
Kasper, C. B., and Kim, J.-J. P. (2001) J. Biol.
Chem. 276, 2916329170
485a. Gutierrez, A., Lian, L.-Y., Wolf, C. R., Scrutton,
N. S., and Roberts, G. C. K. (2001) Biochemistry
40, 19641975
486. Perret, A., and Pompon, D. (1998) Biochemistry
37, 1141211424
487. Vidakovic, M., Sligar, S. G., Li, H., and Poulos,
T. L. (1998) Biochemistry 37, 92119219
488. Takemori, S., and Kominami, S. (1984) Trends
Biochem. Sci. 9, 393396
489. Kobayashi, K., Miura, S., Miki, M., Ichikawa,
Y., and Tagawa, S. (1995) Biochemistry 34,
1293212936
489a. Pochapsky, T. C., Jain, N. U., Kuti, M., Lyons,
T. A., and Heymont, J. (1999) Biochemistry 38,
46814690
490. Poulos, T. L., and Raag, R. (1992) FASEB J. 6,
674679
491. Murataliev, M. B., Klein, M., Fulco, A., and
Feyereisen, R. (1997) Biochemistry 36, 8401
8412
492. Ravichandran, K. G., Boddupalli, S. S.,
Hasemann, C. A., Peterson, J. A., and
Deisenhofer, J. (1993) Science 261, 731736
493. Hasemann, C. A., Ravichandran, K. G.,
Peterson, J. A., and Deisenhofer, J. (1994) J.
Mol. Biol. 236, 11691185
494. Harris, D. L., and Loew, G. H. (1996) J. Am.
Chem. Soc. 118, 63776387
Ch 18IND page - E 2/14/03, 11:11 AM 1082
1083
495. Higgins, L., Bennett, G. A., Shimoji, M., and
Jones, J. P. (1998) Biochemistry 37, 70397046
496. Toy, P. H., Newcomb, M., and Hollenberg, P.
F. (1998) J. Am. Chem. Soc. 120, 77197729
496a. French, K. J., Strickler, M. D., Rock, D. A.,
Rock, D. A., Bennett, G. A., Wahlstrom, J. L.,
Goldstein, B. M., and Jones, J. P. (2001)
Biochemistry 40, 95329538
496b. Schlichting, I., Berendzen, J., Chu, K., Stock,
A. M., Maves, S. A., Benson, D. E., Sweet, R.
M., Ringe, D., Petsko, G. A., and Sligar, S. G.
(2000) Science 287, 16151622
497. Harris, D., Loew, G., and Waskell, L. (1998) J.
Am. Chem. Soc. 120, 43084318
497a. Kupfer, R., Liu, S. Y., Allentoff, A. J., and
Thompson, J. A. (2001) Biochemistry 40, 11490
11501
498. Poulos, T. L., Finzel, B. C., Gunsalus, I. C.,
Wagner, G. C., and Kraut, J. (1985) J. Biol.
Chem. 260, 1612216130
499. Wang, H., Dick, R., Yin, H., Licad-Coles, E.,
Kroetz, D. L., Szklarz, G., Harlow, G., Halpert,
J. R., and Correia, M. A. (1998) Biochemistry 37,
1253612545
500. Custer, L., Zajc, B., Sayer, J. M., Cullinane, C.,
Phillips, D. R., Cheh, A. M., Jerina, D. M.,
Bohr, V. A., and Mazur, S. J. (1999) Biochemis-
try 38, 569581
501. Shanklin, J., Achim, C., Schmidt, H., Fox, B.
G., and Mnck, E. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 29812986
502. Rosenzweig, A. C., Frederick, C. A., Lippard,
S. J., and Nordlund, P. (1993) Nature (London)
366, 537543
502a. Whittington, D. A., Sazinsky, M. H., and
Lippard, S. J. (2001) J. Am. Chem. Soc. 123,
17941795
503. Elango, N., Radhakrishnan, R., Froland, W.
A., Wallar, B. J., Earhart, C. A., Lipscomb, J.
D., and Ohlendorf, D. H. (1997) Protein Sci. 6,
556568
504. Shu, L., Nesheim, J. C., Kauffmann, K.,
Mnck, E., Lipscomb, J. D., and Que, L., Jr.
(1997) Science 275, 515518
505. Siegbahn, P. E. M., and Crabtree, R. H. (1997)
J. Am. Chem. Soc. 119, 31033113
505a. Valentine, A. M., LeTadic-Biadatti, M.-H., Toy,
P. H., Newcomb, M., and Lippard, S. J. (1999)
J. Biol. Chem. 274, 1077110776
505b. Chang, S.-L., Wallar, B. J., Lipscomb, J. D., and
Mayo, K. H. (2001) Biochemistry 40, 95399551
506. Liu, Y., Nesheim, J. C., Paulsen, K. E.,
Stankovich, M. T., and Lipscomb, J. D. (1997)
Biochemistry 36, 52235233
507. Katopodis, A. G., Wimalasena, K., Lee, J., and
May, S. W. (1984) J. Am. Chem. Soc. 106, 7928
7935
508. Pikus, J. D., Studts, J. M., Achim, C.,
Kauffmann, K. E., Mnck, E., Steffan, R. J.,
McClay, K., and Fox, B. G. (1996) Biochemistry
35, 91069119
509. Qian, H., Edlund, U., Powlowski, J., Shingler,
V., and Sethson, I. (1997) Biochemistry 36, 495
504
509a. Eichhorn, E., van der Ploeg, J. R., and
Leisinger, T. (1999) J. Biol. Chem. 274, 26639
26646
509b. Allen, J. R., Clark, D. D., Krum, J. G., and
Ensign, S. A. (1999) Biochemistry 96, 8432
8437
510. Mayer, B., and Hemmens, B. (1997) Trends
Biochem. Sci. 22, 477481
511. Stamler, J. S., Singel, D. J., and Loscalzo, J.
(1992) Science 258, 18981902
511a. Wolthers, K. R., and Schimerlik, M. I. (2001)
Biochemistry 40, 47224737
511b. Ledbetter, A. P., McMillan, K., Roman, L. J.,
Masters, BsS., Dawson, J. H., and Sono, M.
(1999) Biochemistry 38, 80148021
References
511c. Li, H., Raman, C. S., Martsek, P., Masters, B.
S. S., and Poulos, T. L. (2001) Biochemistry 40,
53995406
511d. Marletta, M. A. (2001) Trends Biochem. Sci. 26,
519521
511e. Rusche, K. M., and Marletta, M. A. (2001) J.
Biol. Chem. 276, 421427
511f. Crane, B. R., Arvai, A. S., Ghosh, S., Getzoff,
E. D., Stuehr, D. J., and Tainer, J. A. (2000)
Biochemistry 39, 46084621
512. Culotta, E., and Koshland, D. E., Jr. (1992)
Science 258, 18621863
513. Richeson, C. E., Mulder, P., Bowry, V. W., and
Ingold, K. U. (1998) J. Am. Chem. Soc. 120,
72117219
514. Pfeiffer, S., Gorren, A. C. F., Schmidt, K.,
Werner, E. R., Hansert, B., Bohle, D. S., and
Mayer, B. (1997) J. Biol. Chem. 272, 34653470
515. Marla, S. S., Lee, J., and Groves, J. T. (1997)
Proc. Natl. Acad. Sci. U.S.A. 94, 1424314248
516. Chan, N.-L., Rogers, P. H., and Arnone, A.
(1998) Biochemistry 37, 1645916464
517. Friebe, A., Schultz, G., and Koesling, D. (1996)
EMBO J. 15, 68636868
518. Schelvis, J. P. M., Zhao, Y., Marletta, M. A.,
and Babcock, G. T. (1998) Biochemistry 37,
1628916297
518a. Andersen, J. F., and Montfort, W. R. (2000) J.
Biol. Chem. 275, 3049630503
518b. Roberts, S. A., Weichsel, A., Qiu, Y., Shelnutt,
J. A., Walker, F. A., and Montfort, W. R. (2001)
Biochemistry 40, 1132711337
519. DeMaster, E. G., Quast, B. J., Redfern, B., and
Nagasawa, H. T. (1995) Biochemistry 34,
1149411499
519a. Lai, T. S., Hausladen, A., Slaughter, T. F., Eu, J.
P., Stamler, J. S., and Greenberg, C. S. (2001)
Biochemistry 40, 49044910
520. Gow, A. J., Buerk, D. G., and Ischiropoulos, H.
(1997) J. Biol. Chem. 272, 28412845
520a. Nedospasov, A., Rafikov, R., Beda, N., and
Nudler, E. (2000) Proc. Natl. Acad. Sci. U.S.A.
97, 1354313548
521. Goldstein, S., and Czapski, G. (1996) J. Am.
Chem. Soc. 118, 34193425
522. Caulfield, J. L., Wishnok, J. S., and
Tannenbaum, S. R. (1998) J. Biol. Chem. 273,
1268912695
523. Singh, R. J., Hogg, N., Joseph, J., and
Kalyanaraman, B. (1996) J. Biol. Chem. 271,
1859618603
524. Wong, P. S.-Y., Hyun, J., Fukuto, J. M., Shirota,
F. N., DeMaster, E. G., Shoeman, D. W., and
Nagasawa, H. T. (1998) Biochemistry 37, 5362
5371
525. Ghosh, D. K., and Stuehr, D. J. (1995)
Biochemistry 34, 801807
526. Nathan, C., and Xie, Q.-w. (1994) J. Biol. Chem.
269, 1372513728
527. Rodrguez-Crespo, I., Gerber, N. C., and Ortiz
de Montellano, P. R. (1996) J. Biol. Chem. 271,
1146211467
528. Liu, J., Garca-Cardena, G., and Sessa, W. C.
(1996) Biochemistry 35, 1327713281
529. Bredt, D. S., Hwang, P. M., Glatt, C. E.,
Lowenstein, C., Reed, R. R., and Snyder, S. H.
(1991) Nature (London) 351, 714718
530. Masters, B. S. S., McMillan, K., Sheta, E. A.,
Nishimura, J. S., Roman, L. J., and Martasek,
P. (1996) FASEB J. 10, 552558
531. Hurshman, A. R., and Marletta, M. A. (1995)
Biochemistry 34, 56275634
532. Crane, B. R., Arvai, A. S., Ghosh, D. K., Wu,
C., Getzoff, E. D., Stuehr, D. J., and Tainer, J.
A. (1998) Science 279, 21212126
533. Poulos, T. L., Raman, C. S., and Li, H. (1998)
Structure 6, 255258
534. Villeret, V., Huang, S., Zhang, Y., and Lipscomb,
W. N. (1995) Biochemistry 34, 43074315
535. Gonzlez, D. H., and Andreo, C. S. (1989)
Trends Biochem. Sci. 14, 2427
535a. Reif, A., Frhlich, L. G., Kotsonis, P., Frey, A.,
Bmmel, H. M., Wink, D. A., Pfleiderer, W.,
and Schmidt, H. H. H. W. (1999) J. Biol. Chem.
274, 2492124929
535b. Heller, R., Unbehaun, A., Schellenberg, B.,
Mayer, B., Werner-Felmayer, G., and Werner,
E. R. (2001) J. Biol. Chem. 276, 4047
535c. Hurshman, A. R., and Marletta, M. A. (2002)
Biochemistry 41, 34393456
536. Pufahl, R. A., Wishnok, J. S., and Marletta, M.
A. (1995) Biochemistry 34, 19301941
537. Rusche, K. M., Spiering, M. M., and Marletta,
M. A. (1998) Biochemistry 37, 1550315512
537a. Miranda, K. M., Espey, M. G., Yamada, K.,
Krishna, M., Ludwick, N., Kim, S. M.,
Jourdheuil, D., Grisham, M. B., Feelisch, M.,
Fukuto, J. M., and Wink, D. A. (2001) J. Biol.
Chem. 276, 17201727
538. Schmidt, H. H. H. W., Hofmann, H.,
Schindler, U., Shutenko, Z. S., Cunningham,
D. D., and Feelisch, M. (1996) Proc. Natl. Acad.
Sci. U.S.A. 93, 1449214497
538a. Burner, U., Furtmller, P. G., Kettle, A. J.,
Koppenol, W. H., and Obinger, C. (2000) J.
Biol. Chem. 275, 2059720601
539. Imlay, J. A., and Linn, S. (1988) Science 240,
13021309
540. Fridovich, I. (1997) J. Biol. Chem. 272, 18515
18517
541. Rosen, G. M., Pou, S., Ramos, C. L., Cohen, M.
S., and Britigan, B. E. (1995) FASEB J. 9, 200
209
542. Malmstrom, B. G. (1982) Ann. Rev. Biochem.
51, 2159
543. Naqui, A., and Chance, B. (1986) Ann. Rev.
Biochem. 55, 137166
544. Wood, P. M. (1987) Trends Biochem. Sci. 12,
250251
545. Segal, A. W., and Abo, A. (1993) Trends
Biochem. Sci. 18, 4347
546. Chanock, S. J., Benna, J. E., Smith, R. M., and
Babior, B. M. (1994) J. Biol. Chem. 269, 24519
24522
547. Baggiolini, M., Boulay, F., Badwey, J. A., and
Curnutte, J. T. (1993) FASEB J. 7, 10041010
548. Han, C.-H., Freeman, J. L. R., Lee, T.,
Motalebi, S. A., and Lambeth, J. D. (1998) J.
Biol. Chem. 273, 1666316668
549. Forehand, J. R., Nauseef, W. M., Curnutte, J.
T., and Johnston, R. B., Jr. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 3 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 3995
4028, McGraw-Hill, New York
550. El Benna, J., Dang, P. M.-C., Gaudry, M., Fay,
M., Morel, F., Hakim, J., and Gougerot-
Pocidalo, M.-A. (1997) J. Biol. Chem. 272,
1720417208
551. Huang, J., Hitt, N. D., and Kleinberg, M. E.
(1995) Biochemistry 34, 1675316757
552. Cross, A. R., Rae, J., and Curnutte, J. T. (1995)
J. Biol. Chem. 270, 1707517077
552a. Dang, P. M.-C., Cross, A. R., and Babior, B. M.
(2001) Proc. Natl. Acad. Sci. U.S.A. 98, 3001
3005
553. Isogai, Y., Iizuka, T., and Shiro, Y. (1995) J. Biol.
Chem. 270, 78537857
554. Bouin, A.-P., Grandvaux, N., Vignais, P. V.,
and Fuchs, A. (1998) J. Biol. Chem. 273, 30097
30103
554a. Di-Poi, N., Faur, J., Grizot, S., Molnr, G.,
Pick, E., and Dagher, M.-C. (2001) Biochemistry
40, 1001410022
555. Park, J.-W., Hoyal, C. R., El Benna, J., and
Babior, B. M. (1997) J. Biol. Chem. 272, 11035
11043
Ch 18IND page - E 2/14/03, 11:11 AM 1083
1084 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
References
556. Kramer, I. M., Verhoeven, A. J., van der Bend,
R. L., Weening, R. S., and Roos, D. (1988) J.
Biol. Chem. 263, 23522357
557. Yoshida, L. S., Saruta, F., Yoshikawa, K.,
Tatsuzawa, O., and Tsunawaki, S. (1998) J.
Biol. Chem. 273, 2787927886
558. Segal, A. W., Heyworth, P. G., Cockcroft, S.,
and Barrowman, M. M. (1985) Nature (London)
316, 547549
559. Cox, F. E. G. (1983) Nature (London) 302, 19
559a. Shiose, A., Kuroda, J., Tsuruya, K., Hirai, M.,
Hirakata, H., Naito, S., Hattori, M., Sakaki, Y.,
and Sumimoto, H. (2001) J. Biol. Chem. 276,
14171423
559b. Lee, C.-i, Miura, K., Liu, X., and Zweier, J. L.
(2000) J. Biol. Chem. 275, 3896538972
559c. Bonini, M. G., and Augusto, O. (2001) J. Biol.
Chem. 276, 97499754
559d. Alvarez, B., Ferrer-Sueta, G., Freeman, B. A.,
and Radi, R. (1999) J. Biol. Chem. 274, 842848
559e. Zhang, H., Joseph, J., Feix, J., Hogg, N., and
Kalyanaraman, B. (2001) Biochemistry 40,
76757686
560. Hazen, S. L., Hsu, F. F., dAvignon, A., and
Heinecke, J. W. (1998) Biochemistry 37, 6864
6873
561. Evans, T. J., Buttery, L. D. K., Carpenter, A.,
Springall, D. R., Polak, J. M., and Cohen, J.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 9553
9558
562. Eiserich, J. P., Hristova, M., Cross, C. E., Jones,
A. D., Freeman, B. A., Halliwell, B., and van
der Vliet, A. (1998) Nature (London) 391, 393
397
562a. Henle, E. S., Han, Z., Tang, N., Rai, P., Luo, Y.,
and Linn, S. (1999) J. Biol. Chem. 274, 962971
563. Halliwell, B., and Gutteridge, J. M. C. (1985)
Free Radicals in Biology and Medicine,
Clarendon Press, Oxford
564. Yamazaki, I., and Piette, L. H. (1990) J. Biol.
Chem. 265, 1358913594
565. Wink, D. A., Nims, R. W., Saavedra, J. E.,
Utermahlen, W. E. J., and Ford, P. C. (1994)
Proc. Natl. Acad. Sci. U.S.A. 91, 66046608
566. Pogozelski, W. K., McNeese, T. J., and Tullius,
T. D. (1995) J. Am. Chem. Soc. 117, 64286433
567. Luo, Y., Henle, E. S., and Linn, S. (1996) J. Biol.
Chem. 271, 2116721176
568. Hlavaty, J. J., and Nowak, T. (1997)
Biochemistry 36, 1551415525
569. Weiss, S. J., Test, S. T., Eckmann, C. M., Roos,
D., and Regiani, S. (1986) Science 234, 200203
570. Kanofsky, J. R., Hoogland, H., Wever, R., and
Weiss, S. J. (1988) J. Biol. Chem. 263, 96929696
571. Khan, A. U., and Kasha, M. (1994) Proc. Natl.
Acad. Sci. U.S.A. 91, 1236512367
572. Stratton, S. P., and Liebler, D. C. (1997)
Biochemistry 36, 1291112920
573. Mollinedo, F., Manara, F. S., and Scheider, D.
L. (1986) J. Biol. Chem. 261, 10771082
574. Romeo, D., Skerlavaj, B., Bolognesi, M., and
Gennaro, R. (1988) J. Biol. Chem. 263, 9573
9575
575. Remaley, A. T., Kuhns, D. B., Basford, R. E.,
Glew, R. H., and Kaplan, S. S. (1984) J. Biol.
Chem. 259, 1117311175
576. Heinecke, J. W., Meier, K. E., Lorenzen, J. A.,
and Shapiro, B. M. (1990) J. Biol. Chem. 265,
77177720
577. Shapiro, B. M. (1991) Science 252, 533536
578. Jabs, T., Tschpe, M., Colling, C., Hahlbrock,
K., and Scheel, D. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 48004805
579. Chandra, S., and Low, P. S. (1997) J. Biol.
Chem. 272, 2827428280
580. Imlay, J. A. (1995) J. Biol. Chem. 270, 19767
19777
581. Zhang, L., Yu, L., and Yu, C.-A. (1998) J. Biol.
Chem. 273, 3397233976
582. McCord, J. M. (1985) N. Engl. J. Med. 312,
159163
583. Gonzlez-Flecha, B., and Demple, B. (1995)
J. Biol. Chem. 270, 1368113687
584. Zweier, J. L. (1988) J. Biol. Chem. 263, 1353
1357
585. Karoui, H., Hogg, N., Frjaville, C., Tordo, P.,
and Kalyanaraman, B. (1996) J. Biol. Chem.
271, 60006009
586. Giulivi, C., Poderoso, J. J., and Boveris, A.
(1998) J. Biol. Chem. 273, 1103811043
587. Jousserandot, A., Boucher, J.-L., Henry, Y.,
Niklaus, B., Clement, B., and Mansuy, D.
(1998) Biochemistry 37, 1717917191
588. Berlett, B. S., and Stadtman, E. R. (1997) J. Biol.
Chem. 272, 2031320316
588a. Rauk, A., and Armstrong, D. A. (2000) J. Am.
Chem. Soc. 122, 41854192
588b. Messner, K. R., and Imlay, J. A. (1999) J. Biol.
Chem. 274, 1011910128
588c. Srinivasan, C., Liba, A., Imlay, J. A., Valentine,
J. S., and Gralla, E. B. (2000) J. Biol. Chem. 275,
2918729192
589. Beckman, K. B., and Ames, B. N. (1997) J. Biol.
Chem. 272, 1963319636
590. Chow, C. K., ed. (1988) Cellular Antioxidant
Defense Mechanisms, CRC Press, Boca Raton,
Florida (3 volumes)
591. Halliwell, B., and Gutteridge, J. M. C. (1986)
Trends Biochem. Sci. 11, 372375
592. Dalton, D. A., Russell, S. A., Hanus, F. J.,
Pascae, G. A., and Evans, H. J. (1986) Proc.
Natl. Acad. Sci. U.S.A. 83, 38113815
593. Berger, T. M., Polidori, M. C., Dabbagh, A.,
Evans, P. J., Halliwell, B., Morrow, J. D.,
Roberts, L. J., II, and Frei, B. (1997) J. Biol.
Chem. 272, 1565615660
593a. Kirsch, M., and de Groot, H. (2000) J. Biol.
Chem. 275, 16702 16708
594. Conklin, P. L., Williams, E. H., and Last, R. L.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 9970
9974
594a. Kirsch, M., and de Groot, H. (2001) FASEB J.
15, 15691574
595. Christen, S., Woodall, A. A., Shigenaga, M. K.,
Southwell-Keely, P. T., Duncan, M. W., and
Ames, B. N. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 32173222
596. Stocker, R., Yamamoto, Y., McDonagh, A. F.,
Glazer, A. N., and Ames, B. N. (1987) Science
235, 10431046
597. Peden, D. B., Hohman, R., Brown, M. E.,
Mason, R. T., Berkebile, C., Fales, H. M., and
Kaliner, M. A. (1990) Proc. Natl. Acad. Sci.
U.S.A. 87, 76387642
598. Reiter, R. J. (1995) FASEB J. 9, 526 533
598a. Martn, M., Macas, M., Escames, G., Len, J.,
and Acua-Castroviejo, D. (2000) FASEB J. 14,
16771679
599. Levine, R. L., Mosoni, L., Berlett, B. S., and
Stadtman, E. R. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 1503615040
600. Schultz, J. R., Ellerby, L. M., Gralla, E. B.,
Valentine, J. S., and Clarke, C. F. (1996)
Biochemistry 35, 65956603
600a. Beyer, R. E., Segura-Aguilar, J., Di Bernardo,
S., Cavazzini, M., Fato, R., Fiorentini, D.,
Galli, M. C., Setti, M., Landi, L., and Lenaz, G.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 2528
2532
600b. Lass, A., and Sohal, R. S. (2000) FASEB J. 14,
8794
600c. Suh, J. H., Shigeno, E. T., Morrow, J. D., Cox,
B., Rocha, A. E., Frei, B., and Hagen, T. M.
(2001) FASEB J. 15, 700706
601. Wink, D. A., Hanbauer, I., Krishna, M. C.,
DeGraff, W., Gamson, J., and Mitchell, J. B.
(1993) Proc. Natl. Acad. Sci. U.S.A. 90, 9813
9817
601a. Benaroudj, N., Lee, D. H., and Goldberg, A. L.
(2001) J. Biol. Chem. 276, 2426124267
601b. Sun, Q.-A., Kirnarsky, L., Sherman, S., and
Gladyshev, V. N. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 36733678
601c. Kanzok, S. M., Fechner, A., Bauer, H.,
Ulschmid, J. K., Mller, H.-M., Botella-
Munoz, J., Schneuwly, S., Schirmer, R. H., and
Becker, K. (2001) Science 291, 643646
601d. Lee, S.-R., Bar-Noy, S., Kwon, J., Levine, R. L.,
Stadtman, T. C., and Rhee, S. G. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 25212526
601e. Lowther, W. T., Brot, N., Weissbach, H., and
Matthews, B. W. (2000) Biochemistry 39,
1330713312
601f. St. John, G., Brot, N., Ruan, J., Erdjument-
Bromage, H., Tempst, P., Weissbach, H., and
Nathan, C. (2001) Proc. Natl. Acad. Sci. U.S.A.
98, 99019906
601g. Boschi-Muller, S., Azza, S., Sanglier-
Cianferani, S., Talfournier, F., Van Dorsselear,
A., and Branlant, G. (2000) J. Biol. Chem. 275,
3590835913
601h. Pollock, V. V., and Barber, M. J. (2001)
Biochemistry 40, 14301440
601i. Bieger, B., and Essen, L.-O. (2001) J. Mol. Biol.
307, 18
601j. Seo, M. S., Kang, S. W., Kim, K., Baines, I. C.,
Lee, T. H., and Rhee, S. G. (2000) J. Biol. Chem.
275, 2034620354
602. Deng, H.-X., Hentati, A., Tainer, J. A., Iqbal,
Z., Cayabyab, A., Hung, W.-Y., Getzoff, E. D.,
Hu, P., Herzfeldt, B., Roos, R. P., Warner, C.,
Deng, G., Soriano, E., Smyth, C., Parge, H. E.,
Ahmed, A., Roses, A. D., Hallewell, R. A.,
Pericak-Vance, M. A., and Siddique, T. (1993)
Science 261, 10471051
602a. Yim, M. B., Kang, J.-H., Yim, H.-S., Kwak, H.-
S., Chock, P. B., and Stadtman, E. R. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 57095714
603. Goto, J. J., Gralla, E. B., Valentine, J. S., and
Cabelli, D. E. (1998) J. Biol. Chem. 273, 30104
30109
603a. Estvez, A. G., Crow, J. P., Sampson, J. B.,
Reiter, C., Zhuang, Y., Richardson, G. J.,
Tarpey, M. M., Barbeito, L., and Beckman, J. S.
(1999) Science 286, 24982500
603b. Goto, J. J., Zhu, H., Sanchez, R. J., Nersissian,
A., Gralla, E. B., Valentine, J. S., and Cabelli,
D. E. (2000) J. Biol. Chem. 275, 10071014
604. Gardner, P. R., Raineri, I., Epstein, L. B., and
White, C. W. (1995) J. Biol. Chem. 270, 13399
13405
605. Gaudu, P., and Weiss, B. (1996) Proc. Roy. Soc.
(London) 93, 1009410098
606. Hidalgo, E., Bollinger, J. M., JR, Bradley, T. M.,
Walsh, C. T., and Demple, B. (1995) J. Biol.
Chem. 270, 2090820914
607. Demple, B. (1998) Science 279, 16551656
608. Godon, C., Lagniel, G., Lee, J., Buhler, J.-M.,
Kieffer, S., Perrot, M., Boucherie, H.,
Toledano, M. B., and Labarre, J. (1998) J. Biol.
Chem. 273, 2248022489
609. Zheng, M., slund, F., and Storz, G. (1998)
Science 279, 17181721
610. Ellis, H. R., and Poole, L. B. (1997) Biochemis-
try 36, 1334913356
610a. Fuangthong, M., and Helmann, J. D. (2002)
Proc. Natl. Acad. Sci. U.S.A. 99, 66906695
611. Duranteau, J., Chandel, N. S., Kulisz, A.,
Shao, Z., and Schumacker, P. T. (1998) J. Biol.
Chem. 273, 1161911624
612. Wenger, R. H. (2002) FASEB J. 16, 11511162
613. DeLong, E. F. (2002) Nature (London) 419, 676
677
614. Sinninghe Damst, J. S., Strous, M., Rijpstra,
W. I. C., Hopmans, E. C., Geenevasen, J. A. J.,
van Duin, A. C. T., van Niftrik, L. A., and
Jetten, M. S. M. (2002) Nature (London) 419,
708712
Ch 18IND page - E 2/14/03, 11:11 AM 1084
1085
Study Questions
Concentrations
(mmol/1 of cells)
Abbreviation Metabolite
Before
addition of
antimycin
After
addition of
antimycin
Glucose 6-phosphate G6P 460 124
Fructose 6-phosphate F6P 150 30
Fructose 1,6-bisphosphate FBP 8 33
Triose phosphates TP 18 59
3-Phosphoglycerate 3PGA 45 106
2-Phosphoglycerate 2PGA 26 19
Phosphoenolpyruvate PEP 46 34
Pyruvate Pyr 126 315
Lactate Lac 1125 8750
ATP 2500 1720
ADP 280 855
AMP 36 206
G6P F6P FDP TP etc.
2. The following problem can be solved using stan-
dard reduction potentials (Table 6-8). Use E
(pH 7) values for NAD
+
, enzyme-bound FAD, and
fumarate of 0.32, 0.0, and 0.03 volts, respectively.
Values of numerical constants are given in Table 6-1.
a) Derive an equation relating the equilibrium
constant for a reaction, K
eq
, to differences in E
0
'.
1. Reticulocytes (immature red blood cells) contain
mitochondria that are capable of both aerobic and
anaerobic oxidation of glucose. In an experiment
using these cells, incubated in oxygenated Krebs
Ringer solution with 10 mM glucose, the addition
of antimycin A produced the following changes
in metabolite concentration after 15 min (From
Ghosh, A. K. and Sloviter, H. A. (1973) J. Biol.
Chem. 248, 3035 3040). Interpret the observed
changes in ATP, ADP, and AMP concentrations
(see tabulation). Express the concentration of each
component after addition of antimycin as a per-
centage of that before addition. Then plot the
resulting figures for each compound in the sequence
found in glycolysis, i.e., label the X axis as follows:
b) Calculate the numerical values of K
eq
for the
reactions
Succinate + NAD
+
Fumarate + NADH + H
+
Succinate + FAD Fumarate + FADH
2
at pH 7 and 25C. The values should be calcu-
lated for succinate and the oxidant in the
numerator.
3. Compare the catalytic cycles of the following
enzymes:
Peroxidase
Cytochrome c oxidase
Cytochrome P450
4. What chemical properties are especially important
for the following compounds in the electron trans-
port complexes of mitochondria?
FAD or FMN
Ubiquinone (coenzyme Q)
Cytochrome c
5. Describe the operation of the F
1
F
0
ATP synthase of
mitochondrial membranes.
6. In studies of mitochondrial function the following
stoichiometric ratios have been measured.
a) The P/ O ratio: number of molecules of ATP
formed for each atom of oxygen
(as O
2
) taken up by isolated mitochondria
under specified conditions.
b) The ratio of H
+
ions translocated across a
mitochondrial inner membrane to the
molecules of ATP formed.
c) The ratio of H
+
ions pumped out of a
mitochondrion to the number of
molecules of ATP formed.
Discuss the experimental difficulties in such
measurements. How do uncertainties affect
conclusions about the mechanism of ATP synthase?
Are the ratios in (b) and (c) above necessarily
equal? Explain.
7. Compare P/ O ratios observed for mitochondrial
respiration with the following substrates and
conditions:
Ch 18IND page - E 2/14/03, 11:11 AM 1085
1086 Chapter 18. Electron Transport, Oxidative Phosphorylation, and Hydroxylation
Study Questions
a) Oxidation of NADH by O
2
.
b) Oxidation of succinate by O
2
.
c) Dehydrogenation of ascorbate by O
2
.
How would the ratio of ATP formed to the num-
ber of electrons passing from NADH through the
respiratory chain differ for these three oxidants:
O
2
, fumarate, nitrite?
8. What is the mitochondrial glycerol phosphate
shuttle? Is it utilized by plant cells? Explain.
9. What chemical reactions are included in these two
important components of the nitrogen cycle (see
also Fig. 24-1)?
Nitrification
Denitrification
10. What is the difference between a dioxygenase and a
monooxygenase? What is meant by a cosubstrate
for a monooxygenase?
11. The enzyme p-hydroxybenzoate hydroxylase
utilizes a cosubstrate together with O
2
to form
3,4-dihydroxybenzoate. Indicate the mechanisms
by which the bound FAD cofactor participates in
the reaction.
12. What pterin-dependent hydroxylation reactions
are important to the human body? Point out
similarities and differences between flavin and
pterin hydroxylase mechanisms.
13. Describe the basic properties of nitric oxide syn-
thases (NOSs) and their varied functions in the
body. What are the three different types of NOS?
In what ways do they differ?
14. List several compounds that cause oxidative stress
in cells and describe some chemical and physio-
logical characteristics of each.
15. Propylene glycol is metabolized by several aerobic
bacteria to acetoacetate, which can be catabolized
as an energy source (see references 509a and 509b).
The first step is conversion to an epoxide which
reacts further in coenzyme M-dependent and CO
2
-
dependent reactions to form acetoacetate. Can
you propose chemical mechanisms?
16. A group of slow-growing denitrifying bacteria
obtain energy by oxidizing ammonium ions
anaerobically with nitrite ions.
613,614
NO
2

+ NH
4
+
N
2
+ 2 H
2
O
Intermediate metabolites are hydroxylamine
(H
2
NOH) and hydrazine (N
2
H
4
). The reaction
takes place within internal vesicles known as
anammoxosomes. Unusual cyclobutane- and
cyclohexane-based lipids in their membranes are
thought to partially prevent the escape of the toxic
intermediates from the anammoxosomes.
614
Four protons may move from the cytoplasm
into the vesicles for each ammonium ion oxidized.
Can you write a reaction sequence? What is the
Gibbs energy change for the reaction? How is ATP
generated? See p. 1052.
Ch 18IND page - E 2/14/03, 11:11 AM 1086
1087
Notes
Ch 18IND page - E 2/14/03, 11:11 AM 1087
1088 Chapter 19. The Chemistry of Movement
A. Motility of Bacteria
1. The Structure and Properties of Bacterial Flagella
Quasiequivalence
Growth of flagella
2. Rotation of Flagella
3. Chemotaxis
B. Muscle
1. The Structural Organization of Striated Muscle
2. Muscle Proteins and Their Structures
Actin and the thin filaments
Titin and nebulin
Proteins of the M-line and Z-disc
The regulatory proteins troponin and tropomyosin
Myosins
The thick filaments
3. Actomyosin and Muscle Contraction
The rowing hypothesis
Why two heads?
Structure of the myosin heads
How does actin bind?
Kinesins and other molecular motors
The ATPase cycle of actomyosin and of the kinesins
Observing single molecules
Thinking about chemical mechanisms
4. Control of Muscle Contraction
Calcium ions in muscle
Smooth muscle
5. The Phosphagens
C. Motion in Nonmuscle Cells
1. Actin-Based Motility
2. Transport along Microtubules by Kinesin and
Dynein
3. Eukaryotic Cilia and Flagella
4. Chemotaxis
5. Other Forms of Movement
1089
1089
1090
1091
1091
1093
1096
1096
1096
1096
1099
1099
1099
1099
1101
1104
1104
1105
1105
1106
1107
1108
1108
1110
1114
1114
1117
1117
1118
1118
1119
1121
1122
1122
Boxes
Box 19-A Hereditary Diseases of Muscle
Box 19-B Malignant Hyperthermia and
Stress-Prone Pigs
Box 19-C Actin-Based Motility and Bacterial
Invasion
Tables
Table 19-1 Some Actin-Binding Proteins
1112
1115
1120
1118
References
Study Questions
1122
1127
Contents
Electron micrograph of a thin longitudinal section of a myofibril from pig
muscle. The basic contractile unit is the sarcomere, which extends from one Z
line to the next. Thin actin filaments are anchored at the M lines and the thick
myosin filaments at the Z lines. The (anisotropic) A bands are regions of over-
lap of interdigitated thick and thin filaments, while the I (isotropic) bands are
devoid of thick filaments. The ATP-driven contraction of muscle involves slid-
ing of the interdigitated filaments and shortening of the sarcomere to ~1.8 m.
Micrograph courtesy of Marvin Stromer
Z M Z
I band A band
Sarcomere (~2.5 m)
Ch 19IND page -E- 2/14/03, 11:21 AM 1088
1089
The Chemistry of Movement
19
The swimming of bacteria, the flowing motion of
the ameba, the rapid contraction of voluntary muscles,
and the slower movements of organelles and cytoplasm
within cells all depend upon transduction of chemical
energy into mechanical work.
A. Motility of Bacteria
The smallest organs of propulsion are the bacterial
flagella (Figs. 1-1, 1-3), and we have been able to unravel
some of the mystery of movement by looking at them.
When a cell of E. coli or Salmonella swims smoothly,
each flagellum forms a left-handed superhelix with an
~ 2.3 m pitch. Rotation of these propellors at rates
of 100 200 revolutions / s (100 200 Hz) or more
1,2
in a
counterclockwise direction, as viewed from the distal
end of the flagellum, drives the bacterium forward in
a straight line.
3 8
Several flagella rotate side-by-side as
a bundle.
4
The observed velocities of 20 60 m / s are
remarkably high in comparison with the dimensions
of the bacteria. Also remarkable is the fact that a cell
may travel straight for a few seconds, but then tumble
aimlessly for about 0.1 s before again moving in a
straight line in a different direction. The tumbling
occurs when the flagellum reverses its direction of
rotation and also changes from a left-handed to a
right-handed superhelix, which has just half the
previous pitch.
Such behavior raised many questions. What
causes reversal of direction of the propellor? Why do
the bacteria tumble? How does a bacterium decide
when to tumble? How is the flagellum changed from
a left-handed to a right-handed superhelix? How does
this behavior help the bacterium to find food? Most
intriguing of all, what kind of motor powers the
flagella? The answers are complex, more than 50 genes
being needed to specify the proteins required for
assembly and operation of the motility system of
E. coli or Salmonella typhimurium.
9
1. The Structure and Properties of Bacterial
Flagella
Twenty or more structural proteins are present
from the base to the tip of a complete bacterial flagel-
lum. However, over most of their length the long thin
shafts (Figs. 1-1, 19-1) are composed of subunits of
single proteins called flagellins. Flagellin molecules
have a high content of hydrophobic amino acids and,
in Salmonella, contain one residue of the unusual
N

-methyllysine. The subunits are arranged in a helix


of outside diameter ~ 20 nm in which they also form 11
nearly longitudinal rows or protofilaments.
10 12a
Each
subunit gives rise to one of the projections seen in the
stereoscopic view in Fig. 19-1B. The flagella usually
appear under the electron microscope to be super-
coiled (Fig 19-1CE) with a long wavelength (pitch)
of ~ 2.5 m. The supercoiled structure is essential for
function, and mutant bacteria with straight flagella
are nonmotile. Under some conditions and with some
mutant flagellins, straight flagella, of the type shown
in Fig. 19-1B, are formed. There is a central hole which
is surrounded by what appears to be inner and outer
tubes with interconnecting spokes. However, all of
the 494-residue flagellin subunits presumably have
identical conformations, and each subunit contributes
to both inner and outer tubes as well as to the outer
projections. Basal bodies (Fig. 19-2) anchor the flagella
to the cell wall and plasma membrane and contain the
protic motors (Fig. 19-3) that drive the flagella.
1416
Ch 19IND page -E- 2/14/03, 11:22 AM 1089
1090 Chapter 19. The Chemistry of Movement
Figure 19-1 (A) Axial view of a 5-nm thick cross-section of the flagellar filament shown in (B). The 11 subunits form two
turns of the one-start helix. (B) Stereoscopic oblique view of a 30-nm long section of a flagellum of Salmonella typhimurium.
This is a straight flagellum from a nonmotile strain of bacteria. The structure was determined to a resolution of 0.9 nm by
electron cryomicroscopy. From Mimori et al.
11
Courtesy of Keiichi Namba. (C) Electron micrograph of a cell of S. typhimurium
showing peritrichous (all-around) distribution of flagella. Courtesy of S. Aizawa.
3
(D) Dark-field light micrograph of a flagel-
lated cell of S. typhimurium with flagella dispersed during tumbling (see text). Courtesy of R. M. Macnab.
3
(E) Image of a cell
of Vibrio alginolyticus obtained with dark-field illumination showing the single polar flagellum.
13
Because the illumination
was strong, the size of the cell body and the thickness of the flagellum in the image appear large. Courtesy of Michio Homma.
Quasiequivalence. There are two distinct types
of straight flagella: one (R) in which the protofilaments
have a right-handed twist (as in Fig. 19-1) and the other
(L) in which the protofilaments have a left-handed
twist. These arise from two different conformations
of the subunit proteins. Native supercoiled flagella
contain a mixture of flagellins in the R- and L-states
with all subunits in a given protofilament being in the
same state. The supercoiling of the filament cannot
be explained by stacking of identical subunits but is
thought to arise because of an asymmetric distribution
of protofilaments in a given state around the fila-
ment.
17 19a
Here, as with the icosahedral viruses
(Chapter 7), quasiequivalence permits formation of a
structure that would be impossible with full equiva-
lence of subunits. The corkscrew shape of the flagel-
lum is essential to the conversion of the motors torque
into a forward thrust.
18
Certain mutants of Salmonella
have curly flagella with a superhelix of one-half the
normal pitch. The presence of p-fluorophenylalanine
in the growth medium also produces curly flagella,
and normal flagella can be transformed to curly ones
by a suitable change of pH. More important for
biological function, the transformation from normal
to curly also appears to take place during the tumbling
of bacteria associated with chemotaxis.
17
A
B
C
D
E
5 m
5 m
5 m
10 nm
Ch 19IND page -E- 2/14/03, 11:22 AM 1090
1091
Hook
L ring
P ring
S ring
M ring
C ring
Figure 19-2 (A) Electron micrograph of a flagellum from
E. coli stained with uranyl acetate. The M-and S-rings are
seen at the end. Above them are the P-ring, thought to
connect to the peptidoglycan layer, and the L-ring, thought
to connect to the outer membrane or lipopolysaccharide
layer (see Fig. 8-28). An arrow marks the junction between
hook and thinner filament. From DePamphilis and Adler.
14
The hook is often bent to form an elbow. (B) Average of
~100 electron micrographs of frozen-hydrated preparations
of basal bodies showing the cytoplasmic C-ring (see Fig.
19-3) extending from the thickened M-ring. From
DeRosier.
16
A. Motility of Bacteria
A B
Growth of flagella. Iino added p-fluorophenyl-
alanine to a suspension of bacteria, whose flagella had
been broken off at various distances from the body.
20
Curly ends appeared as the flagella grew out. Unlike
the growth of hairs on our bodies, the flagella grew
from the outer ends. Because no free flagellin was
found in the surrounding medium, it was concluded
that the flagellin monomers are synthesized within
the bacterium, then pass out, perhaps in a partially
unfolded form, through the 2- to 3-nm diameter hole
10,12
in the flagella, and bind at the ends.
21
Flagella of
Salmonella grow at the rate of 1 m in 2 3 min initially,
then more slowly until they attain a length of ~ 15 m.
More recent studies have provided details. The hook
region (Fig. 19-3) grows first to a length of ~ 55 nm by
addition to the basal-body rod of ~ 140 subunits of
protein FlgE. During growth a hook cap formed from
subunits FlgD prevents the FlgE subunits from passing
out into the medium.
22,23
Hook subunits are added
beneath the cap, moving the cap outward. Hook
growth is terminated by protein FlgK (also called
hook-associated protein Hap1). This protein displaces
the hook cap and initiates growth of the main fila-
ment.
24
The first 10 20 subunits added are those of
the FlgK (Fig. 19-3). These are followed by 10 20
subunits of FlgL (Hap3), a modified flagellin whose
mechanical properties can accomodate the stress
induced in the flagella by their rotation.
25
FlgJ is also
needed for rod formation.
25a
Growth of the flagellum to a length of up to 20 m
continues with subunits of FliC that are added at the
tip, which is now covered by a dodecamer of the cap
protein FliD (HAP2).
24,26,26a,b
Its 5-fold rotational
symmetry means that this star-cap does not form
a perfect plug for the 11-fold screw-symmetry of the
flagellum, a fact that may be important in allowing
new flagellin subunits to add at the growing tip. If the
cap protein is missing, as in some FliD mutants, a large
amount of flagellin leaks into the medium.
24
Still unclear is how the protein synthesis that is
taking place on the ribosomes in the bacterial cytoplasm
is controlled and linked to export machinery at the
base of the flagellin. As indicated in Fig. 19-3, the
genetically identified proteins FlhA, FliH, and FliI are
involved in the process that sends the correct flagellin
subunits through the growing flagellum at the appro-
priate time. FliI contains an ATPase domain.
26c
FliS
protein may be an export chaperone.
26d
2. Rotation of Flagella
A variety of experiments showed that the flagellum
is a rigid propellor that is rotated by a motor at the
base. For example, a bacterium, artificially linked by
means of antibodies to a short stub of a flagellum of
another bacterium, can be rotated by the second
bacterium. Rotation of cells tethered to a cover slip
has also been observed. Although it is impossible to
see individual flagella on live bacteria directly, bundles
of flagella and even single filaments (Fig. 19-1C) can
be viewed by dark-field light microscopy.
8,29
Normal
flagella appear to have a left-handed helical form, but
curly Salmonella flagella, which have a superhelix of
one-half the normal pitch, form a right-handed helix.
5
Normal bacteria swim in straight lines but periodically
tumble before swimming in a new random direction.
This behavior is part of the system of chemotaxis by
which the organism moves toward a food supply.
30
Curly mutants tumble continuously. When bacteria
tumble the flagella change from normal to curly. The
pitch is reversed and shortened. A proposed mecha-
nism for the change of pitch involves propagation of
cooperative conformational changes down additional
Ch 19IND page -E- 2/14/03, 11:22 AM 1091
1092 Chapter 19. The Chemistry of Movement
rows of flagellin subunits.
31
There are no muscle-type proteins in the flagella.
By incubating flagellated bacteria with penicillin and
then lysing them osmotically, Eisenbach and Adler
obtained cell envelopes whose flagella would rotate
in a counterclockwise fashion if a suitable artificial
electron donor was added.
32
This and other evidence
showed that ATP is not needed. Rather, the torque
developed is proportional to the protonmotive force
and, under some circumstances, to pH alone. It is
the flow of protons from the external medium into
the cytoplasm that drives the flagella.
8
Movement of
E. coli cells in a capillary tube can also be powered by
an external voltage.
33
In alkalophilic strains of Bacillus
and some Vibrio species a sodium ion gradient will
substitute.
13
Several hundred protons or Na
+
ions
must pass through the motor per revolution.
8
Some
estimates, based on energy balance,
29
are over 1000.
However, Na
+
-dependent rotation at velocities of up
to 1700 Hz has been reported for the polar flagellum
of Vibrio alginolyticus. It is difficult to understand how
the bacterium could support the flow of 1000 Na
+
per
revolution to drive the flagellum.
2
Flh A Fli H,I
Peptidoglycan
Inner
membrane
Outer
membrane
Mot B
Mot A
Flg E (Hook)
Fli F (MS-ring)
Fli G
Flg H (L-ring)
Flg I (P-ring)
Periplasm
C-Ring: FliM, FliN
Rotor
Flg B, C, F,
G, J Rod
Flg L (HAP3)
Flg K (HAP1)
Fli C
(filament, flagellin)
Fli D (HAP2, cap)
Figure 19-3 Schematic drawing of bacterial flagellar motor. Based on drawings
of Berg,
27
Zhou and Blair,
28
and Elston and Oster.
1
What kind of protic motor can
be imagined for bacterial flagella?
Electron microscopy reveals that
the flagellar hook is attached to a
rod that passes through the cell
wall and is, in turn, attached to a
thin disc, the M-ring (or MS-ring),
which is embedded in the cyto-
plasmic membrane both for gram-
positive and gram-negative bacteria
(Fig. 19-3). Two additional rings
are present above the M-ring of
flagella from gram-negative bacteria.
The P-ring interacts with the pepti-
doglycan layer, and the L-ring con-
tacts the outer membrane (lipopoly-
saccharide; Fig. 19-3). A logical
possibility is that the M-ring, which
lies within the plasma membrane, is
the rotor, and a ring of surrounding
protein subunits is the stator for
the motor (Fig. 19-4).
34,35
Glagolev
and Skulachev suggested in 1978
that attraction between COO

and NH
3
+
groups provides the
force for movement.
34
Protons
passing down an H
+
-conducting
pathway from the outer surface
could convert NH
2
groups to
NH
3
+
, which would then be
attracted to the COO

groups on
the stator subunits. When these
two oppositely charged groups
meet, a proton could be transferred
from NH
3
+
to COO

destroying the electrostatic


attraction. At the same time, movement of the M-ring
would bring the next NH
2
group to the H
+
-conducting
pathway from the outside. The COOH of the stator
would now lose its proton through a conducting
pathway to the inside of the bacterium, the proximity
of the new NH
3
+
assisting in this proton release.
Since that time, other models based on electrostatic
interactions have been advanced.
1,29,36
Approximately 40 genes are required for assembly
of the flagella, but mutations in only five motility genes
have produced bacteria with intact flagella that do not
rotate. Among these genes are motA, mot B, FliG, FliM,
and FliN.
16,29,37,37a
Infection with a lambda transducing
bacteriophage carrying functional motB genes restores
motility to motB mutants by inducing synthesis of the
motB protein. Block and Berg observed rotation of
single bacteria tethered to a coverslip by their flagella.
As the synthesis of the motB protein increased,the fla-
gellar rotation rate increased in as many as 16 steps. This
suggested that as many as 16 subunits of the motB gene
product may contribute to the operation of the motor.
38
Later studies suggest eight subunits
39
rather than 16.
Ch 19IND page -E- 2/14/03, 11:22 AM 1092
1093
Both the M-ring and the thin S-ring,
which lies directly above it and is now
usually referred to as the MS-ring, are
formed from ~20 25 subunits of the 61-
kDa FliF protein.
39
Both the MotA and
MotB proteins are embedded in the inner
bacterial membrane and appear to form
a circular array of studs around the
M-ring.
16
MotA has a large cytosolic
domain as well as four predicted trans-
membrane helices
40
while MotB has a
large periplasmic domain and probably
binds to the peptidoglycan.
37,41,41a
The
MotA and MotB proteins, which bind
to each other, are thought to form the
~ 8 functional units in the stator of the
motor.
37
Proteins FliG, FliM, and FliN
are evidently parts of the rotor assembly.
FliM and FliN form an additional ring,
the cytoplasmic or C-ring, which had
been difficult to see in early electron
microscopy. As many as 40 of each of
these subunits may be present in the
ring.
42,43
A ring of FliG subunits joins the
C-ring to the MS-ring (Fig. 19-3). FliE is
also a part of the basal body.
25a
From study of mutants it has been
concluded that three charged residues of
FliG, R279, D286, and D287 are directly
involved in generation of torque by the
motor.
44
Side chains of these residues
may interact with the cytoplasmic
domains of MotA and MotB. Residues
R90 and E98 of MotA may be involved in
controlling proton flow through the motor units.
28,44
The two prolines P173 and P122 are also essential for
torque generation.
28
There are obvious similarities between the flagellar
motors and the protic turbines of ATP synthases (Fig.
18-14), but there are also substantial differences. It
apparently takes about 12 protons for one revolution
of the ATP synthase but about 1000, or ~ 125 per motor
unit, for rotation of a bacterial flagellum. Elston and
Oster propose an ion turbine more complex than that
of ATP synthase. They suggest that the rotor might
contain about 60 slanted rows of positively charged
groups spaced as shown in Fig. 19-4. The motor is
reversible, i.e., it can rotate in either direction. One
possibility is that the subunits alter their conformations
cooperatively in such a way that the slant of the rows
of charged groups is reversed. Other possibilities for
altering the constellation of charges via conformational
changes can be imagined.
1
See also Thomas et al.
44a
A. Motility of Bacteria
Membrane
Mot A
Fli G
C-Ring
(FliM, FliN)
H
+
Rotor
Surface of circumference of C-ring
H
+
Stator
(one of ~8)
Outer
membrane
Periplasmic space
Cytosol
3. Chemotaxis
The flagellar motor is reversible, and in response
to some signal from the bacterium it will turn in the
opposite direction. At the same time, the flagellin sub-
units and those of the hook undergo conformational
changes that change the superhelical twist. Perhaps
synchronous conformational changes in the M-ring
also are associated with the change in direction of
rotation and are induced by interaction with a switch
complex that lies below the M-ring. This consists of
proteins FliG, FliM, and FliN.
44b
Mutations in any
one of these proteins lead to the following four pheno-
types: absence of flagella, paralyzed flagella, or flagella
with the switch biased toward clockwise or toward
counterclockwise rotation.
45
What signals a change in direction of rotation?
The answer lies in the attraction of bacteria to com-
pounds that they can metabolize. Bacteria will swim
toward such compounds but away from repellent
substances, a response known as chemotaxis. Cells
of E. coli swim toward higher concentrations of L-serine
(but not of D-serine), of L-aspartate, or of D-ribose.
Figure 19-4 Schematic drawing of a hypothetical configuration of rotor
and one stator unit in a flagellar motor as proposed by Elston and Oster.
1
The rotor can hold up to 60 positive charges provided by protons flowing
from the periplasm through the stator motor units that surround the
C-ring and hopping from one site to the next along the slanted lines.
The rotor is composed of 15 repeating units, each able to accommodate
four protons. Negative charges on the stator units are 0.5 nm from the
rotor charges at their closest approach. For details see the original paper.
Ch 19IND page -E- 2/14/03, 11:22 AM 1093
1094 Chapter 19. The Chemistry of Movement
Phenol and Ni
2+
ions are repellent.
46 48
By what
mechanism can a minuscule prokaryotic cell sense a
concentration gradient? It is known that the plasma
membrane contains receptor proteins, whose response
is linked to control of the flagella. Since the dimen-
sions of a bacterium are so small, it would probably
be impossible for them to sense the difference in
concentration between one end and the other end of
the cell. The chemotatic response apparently results
from the fact that a bacterium swims for a relatively
long time without tumbling when it senses that the
concentration of the attractant is increasing with time.
When it swims in the opposite direction and the
concentration of attractant decreases, it tumbles
sooner.
49
Koshland
47
proposed that as the membrane
receptors become increasingly occupied with the
attractant molecule, the rate of formation v
f
of some
compound X, within the membrane or within the
bacterium, is increased (Eq. 19-1). When [X] rises
higher than a threshold level, tumbling is induced.
At the same time, X is destroyed at a velocity of v
d
.
Subsequently, a readjustment of v
f
and v
d
occurs such
that the concentration of X falls to its normal steady
state level. X would act directly on the flagellar motor.
The receptors for L-serine
5051a
and L-aspartate
52,53
are 60-kDa proteins encoded by genes tsr and tar in
Salmonella or E. coli.
46,54
These proteins span the inner
plasma membrane of the bacteria as shown in Figs.
11-8 and 19-5. The functioning of the receptor has been
discussed in some detail in Chapter 11. However,
there is still much that is not understood. The sym-
metric head, whose structure is known (Fig. 11-8),
has two binding sites, but the aspartate receptor binds
only one aspartate tightly. There is substantial evidence
that suggests a piston-type sliding of one helix toward
the cytoplasm as part of the signaling mechanism.
54a
While the flagella are distributed around the cell, the
receptors appear to be clustered at the cell poles.
55
Proteins encoded by genes cheA, cheW, cheY and
cheZ, cheB, and ChR are all involved in controlling
chemotaxis.
48,56
Their functions are indicated in the
scheme of Fig. 19-5. All of the corresponding protein
products have been isolated and purified, and the
whole chemotaxis system has been reconstituted in
phospholipid vesicles.
57
Gene CheA encodes a 73-kDa
protein kinase, which binds as a dimer to the cyto-
plasmic domains of the related aspartate, serine, and
ribose/galactose receptors with the aid of a coupling
protein, cheW (Fig. 19-5). A great deal of effort has
been expended in trying to understand how binding
of an attractant molecule to the periplasmic domain of
the receptor can affect the activity of the CheA kinase,
but the explanation is unclear. There is a consensus
v
f
v
d
[X]
(19-1)
that a small but distinct conformational alteration is
transmitted through the receptor.
58 61a
An apparently
-helical region containing methylation sites (Fig. 19-5)
appears to be critically involved in the signaling,
responding not only to occupancy of the receptor site
but also to intracellular pH and temperature and to
methylation. Mutation of the buried Gly 278 found
in this region to branched hydrophobic amino acids,
such as Val or leucine, locks the receptor in state with
a superactivated CheA kinase, while substitution of
Gly 278 with aspartate leaves the kinase inactive.
61
Occupancy of the normal receptor site with ligand
(aspartate, serine, etc.,) dramatically decreases the
kinase activity.
The CheA protein is an autokinase which, upon
activation by the receptor, becomes phosphorylated
on N

of the imidazole ring of His 48. It then transfers


this phospho group from His 48 to the carboxylate of
Asp 57 of the 654-residue protein CheY, which is known
as the response regulator.
62 65d
The unregulated
flagellum rotates counterclockwise (CCW). Phospho-
CheY (CheY-P, which qualifies as X in Eq. 19-1) carries
the message to the flagellar motor to turn clockwise (CW).
This is apparently accomplished through the binding
of CheY-P to the N-terminal portion of protein FliM.
This presumably induces a conformational change,
which is propagated to FliG and to all of the proteins of
the rotor and flagellar rod, hook, and filament.
45,65,66,66a
The flagella fly apart, and the bacterium tumbles and
heads randomly in a new direction.
Tumbling occurs most often when receptors are
unoccupied, and the bacteria change directions often,
as if lost. However, if a receptor is occupied by an
attractant, the activity of CheY is decreased and less
CheY-P will be made. The carboxyl phosphate linkage
in this compound is labile and readily hydrolyzed,
a process hastened by the phosphatase CheZ.
67 69
Consequently, in the presence of a high enough
attractant concentration the tumbling frequency is
decreased, CCW flagellar rotation occurs, and the
bacterium swims smoothly for a relatively long time.
There are still other important factors. Occupancy
of the receptor by a ligand makes the receptor protein
itself a substrate for the chemotaxis-specific methyl-
transferase encoded by the cheR gene.
62,70,71
This
enzyme transfers methyl groups from S-adenosyl-
methionine to specific glutamate side chains of the
receptor to form methyl esters. In the aspartate receptor
there are four such glutamate residues in a large
cytoplasmic domain that includes the C terminus.
Two of these glutamates are initially glutamines and
can undergo methylation only if they are deaminated
first.
72
An esterase encoded by the cheB gene
72
removes
the methyl ester groupings as methanol.
The action of the CheR methyltransferase is
apparently unregulated, but the esterase activity of
CheB is controlled by the phosphorylation state of the
Ch 19IND page -E- 2/14/03, 11:22 AM 1094
1095
Aspartate bound
to receptor
CheY
CheY, response
regulator
P
i
CheA~P
CheY~P H
2
O
CheZ
S-Adenosyl-
methionine
(AdoMet)
CheR
S-Adenosyl-
homocysteine
CH
3
OH
H
2
O
R C
O
O

(Receptor)
R C
OCH
3
O
Induction of
CW rotation
and tumbling
FliM
Methylation
sites
W
CheA
Coupling
protein
Histidine
autokinase
CheW
Inner membrane
CheB
H
2
O
CheB-P
CheA
Signalling path
Figure 19-5 Schematic represen-
tation of an important chemotactic
system of E. coli, S. typhimurium,
and other bacteria. The trans-
membrane receptor activates the
autokinase CheA, which transfers
its phospho group to proteins
CheY and CheB to form CheY-P
and CheB-P. CheY-P regulates
the direction of rotation of the
flagella, which are distributed
over the bacterial surface. CheR
is a methyltransferase which
methylates glutamate carboxyl
groups in the receptor and modu-
lates the CheA activity. CheZ is
a phosphatase and CheB-P a
methylesterase.
autokinase CheA. CheB competes with CheY (Fig.
19-5), and CheB-P is the active form of the esterase.
After a chemotactic stimulus the level of CheA-P falls
and so does the activity of the methylesterase. The
number of methyl groups per receptor rises making
the CheA kinase more active and opposing the de-
crease in kinase activity caused by receptor occupancy.
The system is now less sensitive to the attractant; the
bacterium has adapted to a higher attractant concentra-
tion.
62,73,73a
It tumbles more often unless the attractant
concentration rises; if it is headed toward food tum-
bling is still inhibited. If it is headed away from the
attractant the levels of both CheY-P and ChB-P rise.
A high level of fumarate within the cell also acts on the
switchmotor complex and favors CW rotation.
74
For some bacterial attractants such as D-galactose,
D-ribose, maltose, and dipeptides
75
the corresponding
binding proteins,
38,76
which are required for the sugar
uptake (e.g., Fig. 4-18A), are also necessary for chemo-
taxis. The occupied binding proteins apparently react
with membrane-bound receptors to trigger the chemo-
tactic response. The aspartate receptor (tar gene product)
appears also to be the receptor for the maltose-binding
protein complex,
47
and both the aspartate and the serine
receptor (tsr gene product) also mediate thermotaxis
and pH taxis.
77,77a
Clusters of identical receptors may
function cooperatively to provide high sensitivity and
dynamic range.
77b
A. Motility of Bacteria
Ch 19IND page -E- 2/14/03, 11:22 AM 1095
1096 Chapter 19. The Chemistry of Movement
B. Muscle
There is probably no biological phenomenon that
has excited more interest among biochemists than the
movement caused by the contractile fibers of muscles.
Unlike the motion of bacterial flagella, the movement
of muscle is directly dependent on the hydrolysis of
ATP as its source of energy. Several types of muscle
exist within our bodies. Striated (striped) skeletal
muscles act under voluntary control. Closely related
are the involuntary striated heart muscles, while
smooth involuntary muscles constitute a third type.
Further distinctions are made between fast-twitch and
slow-twitch fibers. Fast-twitch fibers have short
isometric contraction times, high maximal velocities
for shortening, and high rates of ATP hydrolysis.
They occur predominately in white muscle. Because
of the absence of the strong oxidative metabolism
found in red muscles, fast-twitch fibers fatigue rapidly.
Although red muscle sometimes contains fast-twitch
fibers, it more often consists of slow-twitch fibers,
which have a longer contraction time, low shortening
velocity, and low ATPase. They are more resistant to
fatigue
56
than fast-twitch fibers.
78
Embryonic muscle
contains fast-twitch fibers as well as embryonic forms
which contract slowly.
79
Some organisms contain
specialized types of muscle. For example, the asyn-
chronous flight muscles of certain insects cause the
wings to beat at rates of 100 1000 Hz. In these muscles
nerve impulses are used only to start and to stop the
action; otherwise the cycle of contraction and relax-
ation continues automatically.
80
The adductor muscles,
which close the shells of oysters and clams, can sustain
large tensions for long periods of time with little expen-
diture of energy. This is accomplished by a catch
mechanism.
81
1. The Structural Organization of Striated
Muscle
Skeletal muscles consist of bundles of long muscle
fibers, which are single cells of diameter 10 100 m
formed by the fusion of many embryonic cells. The
lengths are typically 2 3 cm in mammals but may
sometimes be as great as 50 cm. Each fiber contains up
to 100 200 nuclei. Typical cell organelles are present
but are often given special names. Thus, the plasma
membrane (plasmalemma) of muscle fibers is called
the sarcolemma. The cytoplasm is sarcoplasm, and
mitochondria may be called sarcosomes. The major
characteristic of muscle is the presence of the contrac-
tile myofibrils, organized bundles of proteins 1 2 m
in diameter and not separated by membranes from the
cytoplasm. Since they occupy most of the cytoplasm,
a substantial number of myofibrils are present in each
muscle fiber.
In the light microscope cross striations with a
repeating distance of ~ 2.5 m can be seen in the myo-
fibrils (Figs. 19-6 and 19-7). The space between two of
the dense Z-discs (Z lines) defines the sarcomere, the
basic contractile unit. In the center of the sarcomere is
a dense A-band (anisotropic band). The name refers
to the intense birefringence of the band when viewed
with plane polarized light. Straddling the Z-discs are
less dense I-bands (the abbreviation stands for iso-
tropic, a misnomer, for although the bands lack bire-
fringence, they are not isotropic). Weakly staining
M-lines (usually visible only with an electron micro-
scope) mark the centers of the A-bands and of the
sarcomeres.
The fine structure of the sarcomere was a mystery
until 1953, when H. E. Huxley, examining thin sections
of skeletal muscle with the electron microscope, dis-
covered a remarkably regular array of interdigitated
protein filaments.
82,83
Thick filaments, 12 16 nm in
diameter and ~ 1.6 m long, are packed in a hexagonal
array on 40- to 50-nm centers throughout the A-bands
(Fig. 19-6B). Between these thick filaments are thin
filaments only 8 nm in diameter and extending from
the Z-line for a length of ~ 1.0 m. When contracted
muscle was examined, it was found that the I-bands
had become so thin that they had nearly disappeared
and that the amount of overlap between the thick and
the thin filaments had increased. This indicated that
contraction had consisted of the sliding movement
of the thick and thin filaments with respect to each
other.
84
In skeletal muscle the sarcomere shortens
to a length of ~ 2 m, but in insect flight muscle a
much smaller shortening occurs repetitively at a very
high rate.
2. Muscle Proteins and Their Structures
The myofibrillar proteins make up 50 60% of the
total protein of muscle cells. Insoluble at low ionic
strengths, these proteins dissolve when the ionic
strength exceeds ~ 0.3 and can be extracted with salt
solutions. Analysis of isolated mammalian myofibrils
86
shows that nine proteins account for 96% or more of
the protein; myosin, which constitutes the bulk of the
thick filaments, accounts for 43% and actin, the prin-
cipal component of the thin filaments, 22%.
Actin and the thin filaments. There are at least
six forms of actin in adult mammalian tissues: -cardiac,
-skeletal muscle, - and -smooth muscle, - and
-cytoplasmic.
87 89
All of them are closely homologous,
e.g., the 42-kDa -skeletal muscle actin differs in only
4 of 375 residues from the -cardiac form and only in 6
residues from the -smooth form. In almost all organ-
isms actins contain one residue of N

-methylhistidine
at position 73.
87,88,90
Actin is an unusual protein in that
Ch 19IND page -E- 2/14/03, 11:22 AM 1096
1097
DB
DB
0.2 m
~30 nm
A
B
B. Muscle
Figure 19-6 (A) The structure of a typical sarcomere of skeletal muscle. The longitudinal section depicted corresponds to
that of the electron micrograph, Fig. 19-7A. The titin molecules in their probable positions are colored green. The heads of
only a fraction of the myosin molecules are shown protruding toward the thin actin filaments with which they interact.
A magnified section at the top is after Spudich.
85
It shows the interactions of the myosin heads with the thin filaments at
the right-hand edge. (B) A sketch showing the arrangement of thick and thin filaments as seen in a transverse section of a
striated muscle fiber. (C) Left: electron micrograph of a transverse section of a glycerated rabbit psoas muscle. The hexagonal
arrangement of six thin filaments around one thick filament can be seen in the center of the circle. Six other thick filaments
form a larger concentric circle as in (B). Right: transverse section of a smooth muscle fiber. Notice the irregular arrangement
of thick and thin filaments. Filaments of intermediate diameter are also present, as are dense bodies (DB). The latter are
characteristically present in smooth muscle.
C
Thick filaments,
1.5 m ~12 nm
Z disc M disc Z disc
A band I band
Sarcomere length 2.5 m
contracts to 1.71.8 m
Thin filaments
0 200 nm 1 m
N
Titin
CC
N N
N
3
Myosin monomer
Actin monomer
Actin
Ch 19IND page -E- 2/14/03, 11:22 AM 1097
1098 Chapter 19. The Chemistry of Movement
Figure 19-7 (A) Electron micrograph of a longitudinal section of a mammalian
skeletal muscle (pig biceps muscle). The tissue was doubly fixed, first with
formaldehyde and glutaraldehyde, then with osmium tetroxide. It was then
stained with uranyl acetate and lead citrate. The section shows a white muscle
fiber containing few mitochondria and narrow Z-lines. The Z-discs (marked
Z), M-line, A- and I-bands, and thick and thin filaments can all be seen clearly.
The periodicity of ~40 nm along the thin filaments corresponds to the length
of the tropomyosin molecules, and the cross striation is thought to represent
bound tropomyosin and troponin. The numerous dense particles in the upper
part of the micrograph are glycogen granules, while the horizontal membranous
structures are longitudinal tubules of the sarcoplasmic reticulum (endoplasmic
reticulum). These come into close apposition to the T tubules leading from the
surface of the muscle fiber. The T tubules (T) are visible in longitudinal section
at the upper left of the micrograph on both sides of the Z-line and in cross-
section in the upper right-hand corner. There a T tubule is seen lying between
two lateral cisternae of the sarcoplasmic reticulum. (B) Longitudinal section
of smooth muscle (chicken gizzard) fixed as in A. Thick filaments (Th), which
are considerably thicker than those in striated muscle and less regular, can be
seen throughout the section. They are surrounded by many thin filaments,
which are often joined to dense bodies (DB). A mitochondrion (Mi) is seen in
the center of the micrograph, and at the lower edge is a boundary between
two adjacent cells. Notice the caveolae (C), which are present in large numbers
in the plasma membrane and which are extremely active in smooth muscle.
Micrographs courtesy of Marvin Stromer.
A
B
it can exist in both a filamentous
and a soluble state. The intercon-
version between them is of great
physiological importance. Actin
filaments dissolve in a low ionic
strength medium containing ATP
to give the soluble, monomeric
G-actin. Each G-actin monomer
usually contains one molecule of
bound ATP and a calcium ion.
Because of its tendency to poly-
merize, G-actin has been difficult to
crystallize. However, it forms crys-
talline complexes with several other
proteins, e.g., deoxyribonuclease I,
91
a fragment of gelsolin, and profilin,
92
which block polymerization and it
has recently been crystallized as the
free ADP complex.
92a
The three-
dimensional structure of the actin
is nearly the same in all cases. The
molecule folds into four domains,
the ATP binding site being buried in
a deep cleft. The atomic structure
(Fig. 7-10) resembles that of hexo-
kinase, of glycerol kinase, and of an
ATP-binding domain of a chaperonin
of the Hsp 70 family.
90
As with the
kinases, actin can exist in a closed
and more open conformations, one
of which is seen in the profilin com-
plex. Addition of 1 mM Mg
2+
or 0.1
M KCl to a solution of G-actin leads
to spontaneous transformation into
filaments of F-actin (Figs. 7-10 and
19-9) each containing 340 380 actin
monomers and resembling the thin
filaments of muscle.
93-94a
The ATP
bound in the F-actin filament is
hydrolyzed within ~ 100 s to ADP
and P
i
. However, the hydrolysis is
not as rapid as polymerization so
that a cap of ATP-containing
monomers may be found at each
end of the filament.
94,95,95a
There is a
striking similarity to the binding of
nucleotides to microtubule subunits
(Fig. 7-33) and in the contractile tail
sheath of bacteriophage (Box 7-C).
The two ends of the F-actin
filaments have different surfaces of
the monomer exposed and grow at
different rates. This has been dem-
onstrated by allowing the myosin
fragment called heavy meromyosin
(HMM; see Fig. 19-10) to bind to (or
decorate) an actin filament. The
0.4 m
0.4 m C
Ch 19IND page -E- 2/14/03, 11:23 AM 1098
1099
myosin heads bind at an angle, all pointed in one
direction. This gives a pointed appearance to one
end and a barbed appearance at the other. When
monomeric actin is added to such an HMM-decorated
F-actin filament the barbed ends grow much faster
than the pointed ends.
94,96
In the intact sarcomere the
ends that become pointed when decorated are free,
while the opposite barbed ends of the filaments are
attached at the Z-line (Fig. 19-6A). The existence in the
cytoplasm of proteins that cap the fast-growing end
of actin filaments thereby preventing further growth
96,97
suggests that cap proteins may be present at the ends
of the thin filaments of the myofibrils.
Titin and nebulin. The third most abundant
protein (10%), titin (also called connectin),
98 100a
is one
of the largest of known proteins. Titin cDNA from
human cardiac muscle encodes a 26,926-residue chain.
Several tissue-specific isoforms of the protein are
created by alternative mRNA splicing.
101
A single titin
molecule stretches ~ 1200 nm from the Z-disc, where
the N terminus is bound, to the M-line, where the C-
terminal domain is attached (Fig. 19-8A). Thoughout
much of the A-band titin binds to the thick filament
and appears to be part of a scaffold for maintenance
of the sarcomere structure. The I-band portion of titin
has elastic properties that allow it to lengthen greatly
or to shorten as the sarcomere changes length.
98,100,102
Under the electron microscope titin appears as a
flexible beaded string ~4 nm in diameter. Most of the
molecule is made up of repetitive domains of two
types. In human cardiac titin there are 132 folded
domains that resemble type III fibronectin repeats and
112 immunoglobulin-like domains.
98
In a PEVK
region, between residues 163 and 2174, 70% of the
residues are Pro, Glu, Val, or Lys. The titin molecule
may be organized as polyproline helices in this elastic
region.
102a
At the C terminus of titin 800 residues,
including a Ser / Thr protein kinase domain, are found
within the M-line.
Another very large protein, nebulin (3% of the
total protein),
103
appears to be stretched alongside the
thin filaments. In the electron microscope it appears as
a flexible, beaded string ~ 4 nm in diameter. Ninety-
seven percent of the 6669-residue human nebulin is
organized as 185 copies of an ~ 35-residue module.
104,105
Nebulin has a proline residue at about every 35th
position, possibly corresponding in length to the pitch
of the actin helix (Fig. 7-10). At the C terminus is an
SH3 domain (see Fig. 11-14), which is preceded by a
120-residue segment rich in potential phosphorylation
sites.
106
This part of the peptide chain is anchored in the
Z-discs (Fig. 19-8B, C). The three extreme N-terminal
modules of nebulin bind to tropomodulin, which caps
the pointed ends of thin filments.
106a
Avian cardiac
muscle contains a much shorter 100-kDa protein called
nebulette, which resembles the C-terminal parts of
nebulin. Nebulin has been described as encoding a
blueprint for thin filament architecture.
99,103
Proteins of the M-line and Z-disc. The M-line
region contains the structural protein myomesin,
which binds to both titin and myosin and holds the
two together.
107
Fast skeletal and cardiac fibers also
contain another M-protein, which may bridge between
myosin filaments. Both the C-terminal region of nebu-
lin and the N termini of pairs of titin molecules meet
in the Z-disc, where they are bound into a lattice con-
taining -actinin
98,108-109b
and other proteins (Fig.
19-8B). The dimeric -actinin, a member of the spec-
trin family, has a subunit mass of ~ 97 kDa.
109a
Found
primarily in the Z-discs, it is also present in nonmuscle
cells in stress fibers and at other locations in the cyto-
skeleton (Chapter 7). It may anchor actin filaments
to various structures outside of the sarcomere.
110
In
the dense Z-disc of insect flight muscle a regular hex-
agonal lattice of -actinin
111
and a large (500 700 kDa)
modular protein called kettin
112,112a,b
bind the thin
filaments of opposite polarity together.
The C-protein (thick filaments), myomesin (M-line
protein), and -actinin (Z-line protein)
110,113,114
each
provide 2% of the protein in the myofibril. Less than 1%
each of 11 or more other proteins may also be present
within the sarcomere.
86,115
Several of these, including
the cytoskeletal proteins desmin and vimentin, and
synemin surround the Z-discs.
116,116a
The regulatory proteins troponin and tropo-
myosin. These two proteins are also associated with
the filaments, each one contributing ~ 5% to the total
protein of myofibrils. Tropomyosin is an elongated
-helical coiled-coil molecule, each molecule of which
associates with seven actin subunits of an actin fila-
ment. Troponin consists of three subunits known as
troponins C, T, and I. The elongated troponin T binds
to tropomyosin. Troponin I is an inhibitor of the inter-
action of myosin and actin necessary for muscle con-
traction. Troponin C, a member of the calmodulin
family (Fig. 6-8), binds Ca
2+
and induces conforma-
tional changes that relieve the inhibition and allow
contraction to occur. Nebulin is also thought to bind
to tropomyosin. A possible arrangement of one of the
tropomyosintroponinnebulin complexes that lie
along the length of the thin filaments is shown sche-
matically and as a three-dimensional model in Figs.
19-8C and D. These proteins are discussed further in
Section 4. Figure 19-9 shows a model of the thin fila-
ments with tropomyosin coiled-coil molecules on each
side. The troponin subunits are not shown.
Myosins. There are 15 distinct families of proteins
within the myosin superfamily.
117120
They vary greatly
in size, but all of them bind and hydrolyze ATP, and all
bind to actin. Most have C-terminal tails. At their N
B. Muscle
Ch 19IND page -E- 2/14/03, 11:23 AM 1099
1100 Chapter 19. The Chemistry of Movement
Figure 19-8 (A) Schematic drawing showing one molecule of titin (connectin) in a half sarcomere and its relationship to
thick myosin filaments and thin actin filaments. The complex repeat patterns of fibronectin type III, immunoglobulin, in
the three zones D, C, and P are also indicated.
98
See Maruyama.
98,98a
(B) Schematic drawing of the molecular structure of the
sarcomere Z-disc. Titin, which is thought to parallel the thin filaments through the I-band, consists of various modules that
are numbered from the N termini. In the Z-disc titin binds to -actin, shown here as three vertical rods, and also to actin or
actin-binding proteins. The SH3 domain ( shown as a sphere) of nebulin and the N terminus of titin may interact. Regulatory
phosphorylation sites are marked P. From Young et al.
108
Courtesy of Mathias Gautel. (C) Hypothetical model of a composite
regulatory complex containing nebulin, tropomyosin, and troponin on the thin filaments of a skeletal muscle sarcomere. Each
seven-module nebulin super-repeat (squares with graded shading) binds one tropomyosin, possible through the seven charge
clusters along the length of each tropomyosin, and one troponin complex (shaded spheres with a tail). This complex consists
of TnT, TnI, and TnC in orientations indicated by the N and C termini. Each nebulin super-repeat binds to seven actin monomers
(open circles) along the thin filament. Tropomodulin caps the pointed ends of actin filaments and Cap Z, the barbed ends.
From Wang et al.
103
Actin filament
PEVK N
C
Kinase
Myosin filament Titin
190
Z
674-1400 1900
210
kDa
A band I band
M
D zone C zone P zone
6 11
Fibronectin type III domain (motif I)
Immunoglobulin domain (motif II)
MLCK region
Interdomain
A
7 6
5
4
3
P
P
2 1
7 6
5
4
3
P
P
2 1
Actin
P
Titin immunoglobulin domain
Titin Z-repeat
-Actinin
Titin non-modular sequences
Nebulin
Phosphorylation site
Titin
N C
Actin
Tropomodulin Cap Z
(-actinin)
Troponin
C N
TnT
TnCTnI
Tropomyosin
N
Nebulin
C
Z
C
B
Ch 19IND page -E- 2/14/03, 11:23 AM 1100
1101
termini are one or two globular heads, which contain
the catalytic centers in which ATP hydrolysis occurs.
Sizes vary from 93 kDa for a myosin with a very short
tail from Toxoplasma
118
to over 300 kDa. Myosins I,
found in ameboid organisms and also in our own
bodies (for example in the microvilli of the brush
border of intestinal epithelial cells), are small single-
headed molecules.
117,121
Myosins II are the conven-
tional myosins of myofibrils and are often referred to
simply as myosin. However, each of the three muscle
types (skeletal, cardiac, and smooth) has its own kind
of myosin II.
121ac
Likewise, at least six different genes
have been identified for the light chains of the myosin
heads.
122
Fast and slow muscle as well as embryonic
muscle have their own light chains. Each myosin II
molecule consists of two identical ~ 230-kDa heavy
chains, which are largely -helical, together with two
pairs of smaller 16- to 21-kDa light chains. Human
skeletal muscle heavy chains contain 1938 residues
of which the first ~ 850 are folded into pear-shaped
heads, which contain the catalytic sites involved in
harnessing ATP cleavage to movement. Following
proline 850 nearly all of the remaining 1088 residues
form an -helical coiled-coil rod of dimensions ~160 x
2 nm (Fig. 19-10) in which the two chains coil around
each other. The two heavy chains are parallel, each
having its N terminus in one of the two heads and its
C terminus bound in the shaft of the thick filament.
Myosins II from other sources have similar struc-
tures. For example, analysis of the DNA sequence for
a heavy chain gene from the nematode Caenorhabditis
showed that the protein contains 1966 residues, 1095
of which contain an amino acid sequence appropriate
for a 160-nm long coiled coil.
123
There are no prolines
within this sequence, which lies between Pro 850 and
B. Muscle
Figure 19-9 Stereoscopic ribbon drawing of the
proposed structure of a thin actin filament with
tropomyosin coiled-coils bound on opposing sides.
124
Five actin nomomers are assembled in the structure as
is also illustrated in Fig. 7-10. From Lorenz et al.
125
Courtesy of Michael Lorenz.
Pro 1944. Although there are many bands containing
positively and negatively charged side chains along
the myosin rod, the interactions between the two
coiled helices are largely nonpolar. In Drosophila 15
different heavy-chain isoforms are created by splicing
of a single mRNA.
123a
While the C-terminal portions of the two parallel
myosin heavy chains form a rod, the N-terminal por-
tions fold into two separate heads. Each head also
contains two smaller 16- to 21-kDa peptide chains
which belong to the calmodulin family. One of these,
the essential light chain, is tightly bound to the
heavy chain. The second, the regulatory light chain,
is able to bind Ca
2+
and is less tightly bound to the rest
of the head. A short treatment with trypsin or papain
cuts the myosin molecule into two pieces. The tail end
gives rise to light meromyosin (LMM), a molecule
~ 90 nm in length. The remainder of the molecule
including the heads is designated heavy meromysin
(HMM). A longer trypsin treatment leads to cleavage
of HMM into one ~ 62-kDa S2 fragment 40 nm long,
and two ~ 130-kDa S1 fragments, each of the latter
representing one of the two heads (Fig. 19-10).
The junction of the head and tail portions of myo-
sin appears rigid in Fig. 19-10A. However, there must
be considerable conformational flexibility and perhaps
some uncoiling of the helices to allow the two heads
to interact with a single thin filament as is observed
by electron microscopy.
126,128
There also appears to be
a hinge between the S2 and LMM segments (Figs.
19-10A and 19-14).
The thick filaments. Dissociated myosin mole-
cules can be induced to aggregate into rods similar to
the thick filaments of muscle.
129
Since the filaments
Ch 19IND page -E- 2/14/03, 11:23 AM 1101
1102 Chapter 19. The Chemistry of Movement
100 nm
Heads emerging at this
level have their C-terminal
ends pointed towards the
M line
Levels of heads are
14.4 nm apart
Bare zone
~160 nm
~160 nm
M M
Figure 19-10 (A) An approximate scale drawing of the myosin molecule. The hinge is a
region that is rapidly attacked by trypsin to yield the light and heavy meromyosins (LMM and
HMM). Total length ~160 nm, molecular mass, 470 kDa; two ~ 200-kDa heavy chains; two pairs
of 16- to 21-kDa light chains; heads: ~15 x 4 x 3 nm. (B) Electron micrograph of rabbit myosin
monomers that became dissociated from thick filaments in the presence of ATP, fixed and
shadowed with platinum.
127
Courtesy of Tsuyoshi Katoh.
A
B
Figure 19-11 (A) Radial projection illustrating packing of myosin rods as suggested by Squire
130
for thick filaments of vertebrate
skeletal muscle. The region of the bare zone at the M-line is shown. The filled circles represent the head ends of the myosin
molecules and the arrowheads represent the other end of the rod, i.e., the end of the LMM portion. Antiparallel molecules
interacting with overlaps of 43 and 130 nm are shown joined by single and triple cross-lines, respectively. Positions where
two arrowheads meet are positions of end-to-end butting. O is an up molecule (thin lines) and A a down molecule (thick
lines). The molecules move from the core at the C-terminal end to the filament surface at the head end. The levels marked B
may be the levels of attachment of M-bridge material to the myosin filament. The level M-M is the center of the M-line and of
the whole filament. The lateral scale is exaggerated more than threefold. (B) A segment of titin showing the 43-nm 11-domain
super-repeat. (C) Model of a segment of a thick filament showing the 43-nm repeat, the C-protein, also bound at 43 nm inter-
vals.
99
(B) and (C) Courtesy of John Trinick.
43 nm
43 nm
M-line
Z-line
N
22
11
1
C
A B C
C-terminal ends
Adjacent
myosin
HMM
S1 heads
Points of slow
trypsin cleavage
Hinge
(rapid trypsin cleavage)
S2, 50 nm LMM, 90 nm
~43 nm
Ch 19IND page -E- 2/14/03, 11:23 AM 1102
1103
have a diameter of ~ 14 to 20 nm, a large number of the
thin 2-nm myosin molecules must be packed together.
Electron microscopy reveals the presence of the heads
projecting from the thick filaments at intervals of ~ 43.5
nm. However, there is a bare zone centered on the
M-line, a fact that suggests tail-to-tail aggregation of
the myosin monomers at the M-line in the centers of
the thick filaments (see magnified section of Fig. 19-6,
A). A helical packing arrangement involving about
300 myosin molecules (up to 30 rods in a single cross
section) in close packing with a small central open core
has been proposed for skeletal muscle myosin
130,131
and is illustrated in Fig. 19-11A,B. There are approxi-
mately three heads per turn of the helix, each group
of three heads spaced 14.3 nm from the preceding one
along the thick filament. It is apparently the zones of
positive and negative charge, which are especially
prevalent in the LMM segment toward the C termini,
that lock the successive myosin molecules into this
14.3-nm spacing.
116,132
Titin also binds to the LMM
segment of the myosin rod,
99,133
and its 11-domain
super repeat of IgG-like and fibronectin-like modules
are also 43 nm in length.
98,101
There are typically 47 49
of these super repeats in titin, and if each fits to a turn
of the helix, as shown in Fig. 19-11B, there would be
147 myosin molecules in one-half of the thick filament.
Not all muscles have the thick filament structure
Figure 19-12 (A) Stereo-
scopic views of computer-
assisted reconstructions of
images of myosin heads
attached to an F-actin filament
centered between two thick
filaments. Atomic structures
of actin (Fig. 7-10) and of
myosin heads (Fig. 19-15)
have been built into the
reconstructed images obtained
by electron microscopy. (A)
With the nonhydrolyzable
ATP analog ATPPNP bound
in the active sites. (B) Rigor.
Two myosin heads are appar-
ently bound to a single actin
filament in (A). If they belong
to the same myosin molecule
the two C-terminal ends must
be pulled together from the
location shown here. In (B) a
third head is attached, pre-
sumably from another myosin
rod. This configuration is
often seen in rigor. From
Winkler et al.
134
Courtesy of
K. A. Taylor.
of Fig. 19-11. In the tarantula muscle, which has a
particularly well-ordered structure, there are four
myosins per turn.
135,135a
Figure 19-13 shows a recon-
struction of scallop myosin which has a 7-fold rota-
tional symmetry. The thick filaments often contain
Figure 19-13 Superimposed sections for the 14 nm thick-
ness of a computer-assisted reconstruction of the myosin
filaments of the scallop adductor muscle. From Vibert and
Craig.
137
A
B
B. Muscle
Ch 19IND page -E- 2/14/03, 11:23 AM 1103
1104 Chapter 19. The Chemistry of Movement
other proteins in addition to myosin. Thus, skeletal
muscle contains the C-protein in a series of helical bands
along the thick rod.
135b,c
In nematodes, molluscs, and
insects the thick filament has a cylindrical core of
paramyosin, another protein with a structure resem-
bling that of the myosin rod. A minor component of
Drosophila myosin, the myosin rod protein, lacks heads
but is transcribed from the myosin heavy chain gene.
136
3. Actomyosin and Muscle Contraction
That actin and myosin are jointly responsible for
contraction was demonstrated long before the fine
structure of the myofibril became known. In about
1929, ATP was recognized as the energy source for
muscle contraction, but it was not until 10 years later
that Engelhardt and Ljubimowa showed that isolated
myosin preparations catalyzed the hydrolysis of
ATP.
138
Szent-Gyrgi
139,140
showed that a combination
of the two proteins actin (discovered by F. Straub
141
)
and myosin was required for Mg
2+
-stimulated ATP
hydrolysis (ATPase activity). He called this combina-
tion actomyosin.
Under the electron microscope the myosin heads
can sometimes be seen to be attached to the nearby
thin actin filaments as crossbridges. When skeletal
muscle is relaxed (not activated by a nerve impulse),
the crossbridges are not attached, and the muscle can
be stretched readily. The thin filaments are free to
move past the thick filaments, and the muscle has
some of the properties of a weak rubber band. How-
ever, when the muscle is activated and under tension,
the crossbridges form more frequently. When ATP is
exhausted (e.g., after death) muscle enters the state of
rigor in which the crossbridges can be seen by electron
microscopy to be almost all attached to thin filaments,
accounting for the complete immobility of muscle in
rigor (Figs. 19-12, 14).
134
In rigor the crossbridges are almost all firmly
attached to the thin actin filaments, making an approx-
imately 45 angle to the actin filaments.
142 144
However,
the addition of ATP causes their instantaneous release
and the relaxation of the muscle fiber. In contrast,
activation by a nerve impulse, with associated release
of calcium ions (Section B,4), causes the thin filaments
to slide between the thick filaments with shortening
of the muscle. An activated muscle shortens if a low
tension is applied to the muscle, but at a higher tension
it maintains a constant length. Because the maximum
tension developed is proportional to the length of
overlap between the thick and thin filaments, it was
natural to identify the individual crossbridges as the
active centers for generation of the force needed for
contraction.
The rowing hypothesis. H. E. Huxley
145,146
and
A. F. Huxley and R. M. Simmons
147
independently
proposed that during contraction the myosin heads
attach themslves to the thin actin filaments. The hy-
drolysis of ATP is then coupled to the generation of a
tension that causes the thick and thin filaments to be
pulled past each other. The heads then release them-
selves and become attached at new locations on the
actin filament. Repetition of this process leads to the
sliding motion of the filaments (Fig. 19-14). The evi-
dence in favor of this rowing or swinging bridge
hypothesis was initially based largely on electron
microscopy. For example, contracting muscle was
frozen rapidly and fixed for microscopy in the frozen
state.
148
Relaxed muscle shows no attached cross-
bridges, but contracting muscle has many. However,
Figure 19-14 A model for the coupling of ATP hydrolysis to force production in muscle based on proposals of H. E. Huxley,
and A. F. Huxley and Simmons. The power stroke is depicted here as a rotation of the crossbridge from a 90 to a 45 configu-
ration. Four representative stages are shown: (1) the rigor complex, (3) the dissociated myosin ATP complex, (4) the actomyo-
sin ADP pre-power stroke state in which the actinmyosin band has reformed but with a different actin subunit, which may be
distant from that in (1), and (6) the actomyosin ADP post-power stroke state. Force production and contraction result from
crossbridges passing cyclically through the steps depicted from left to right. Numbering of the stages corresponds approxi-
mately to that in Fig. 19-18. After H. Huxley.
146
Myosin
Actin
3
S
1
1
ATP
S
2
4
ADP + P
i
ADP
6
Ch 19IND page -E- 2/14/03, 11:23 AM 1104
1105 B. Muscle
their appearance was distinct from that seen in rigor.
The model was also supported by indirect physical
methods.
An impressive demonstration that myosin heads
do move along the actin filaments was provided by
Sheetz and Spudich, who found that myosin-coated
fluorescent beads ~0.7 m in diameter will move along
actin filaments from cells of the alga Nitella in an ATP-
dependent fashion at velocities similar to those required
in muscle.
149
The myosin heads literally glide along
the thick cables of parallel actin filaments present in
these algae.
Why two heads? The actin filament is a two-start
helix, and it is natural to ask whether the two myosin
heads bind to just one or simultaneously to both of the
actin strands. Most evidence supports a 1:1 interaction
of a single head with just one strand of actin. However,
the other actin strand may associate with heads from
a different thick filament. Another question concerns
the role of the pairs of myosin heads. Could the two
heads bind sequentially to the actin and exert their
pull in a fixed sequence? In the reconstruction of the
actomyosin complex in rigor (Fig. 19-12B) two different
images are seen for the crossbridges. This suggests
the existence of two different conformations for the
attached myosin heads. Similar images for smooth
muscle heavy meromyosin in its inactive (resting)
dephosphorylated state (see p. 1116) show the two
heads in very different orientations with one binding
to the other of the pair and blocking its movement.
121b
Perhaps one head is tightly bound at the end of the
power stroke while the other is at a different stage of
the catalytic cycle. Nevertheless, single-headed myosin
from Acanthamoeba will propell organelles along actin
filaments,
150
and actin filaments will slide across a
Figure 19-15 Ribbon representation of chicken skeletal myosin subfragment-1 showing the major domains and tryptic
fragments. Prepared with the program MolScript. From Rayment.
157
surface coated with single-headed myosin formed by
controlled proteolysis.
151
The additional interactions
seen in rigor may be peculiar to that state.
Structure of the myosin heads. Myosin and
myosin fragments can be isolated in large quantities,
but they have been difficult to crystallize. However,
Rayment and coworkers purified S1 heads cleaved
from chicken myosin by papain and subjected them to
reductive methylation (using a dimethylamineborane
complex; see also Eq. 3-34). With most of the lysine
side chain amino groups converted to dimethylamine
groups, high-quality crystals were obtained, and a
structure was determined by X-ray diffraction.
152
Since that time various forms of both modified and
unmodified myosin heads from several species have
been studied by X-ray crystallography.
153160
Especially
clear results were obtained with unmodified myosin
from the ameba Dictyostelium discoideum. The head
structure, shown in Fig. 19-11, includes a 95-kDa piece
of the heavy chain and both light chains. A clearer
picture of the neck region containing the light chains
was provided by the structure of the regulatory
domain of scallop myosin.
161
Unlike mammalian or
avian myosins, molluscan myosins are regulated by
binding of Ca
2+
to a site in the essential light chain,
but the structures are similar to those in Figs. 19-10
and 19-15.
Cleavage of the ~850-residue S1 heads with trypsin
yields mainly three large fragments that correspond
to structural domains of the intact protein as shown in
Fig. 19-15. They are known as the 25-kDa (N-terminal),
50-kDa, and 20-kDa fragments, and for myosin from
D. discoideum correspond to residues 1 to 204, 216 to
626, and 647 to 843, respectively. The ATP-binding
site is in a deep cleft between the 20-kDa and 50-kDa
Upper domain
of 50 kDa region
Lower domain
of 50 kDa region
50 kDa cleft
Nucleotide
pocket
C-terminal 20 kDa region
Essential
light chain
Regulatory
light chain
Attachment to
myosin rod
N-Terminal
25 kDa region
Ch 19IND page -E- 2/14/03, 11:23 AM 1105
1106 Chapter 19. The Chemistry of Movement
Figure 19-16 (A) The nucleotide binding site of myosin
with MgADPBeF
x
bound in a conformation thought to
mimic that of ATP prior to hydrolysis. The -sheet strands
are contributed by both the 25-kDa and 50-kDa domains.
The P-loop lies between T178 and E187. The conserved
N233 to G240 loop, which also contributes important ATP-
binding residues, comes from the 50-kDa region. (B) Stereo-
scopic view of the -phospho group binding pocket with the
bound MgADPVO
4
(vanadate) complex. The coordinated
Mg
2+
and associated water molecules are seen clearly.
Courtesy of Ivan Rayment.
157
regions. Figure 19-16 illustrates the binding of an ATP
analog, the beryllium fluoride complex of MgADP, in
the active site. As can be seen, the ATP binds to loops
at the C termini of the strands of the 8-stranded
sheet from the 25-kDa domain. The conserved P-loop
(Chapter 12, E), which lies between T178 and E187,
curls around the and phospho groups, and has the
sequence G(179)ESGAGKT. A second conserved loop
N(233)SNSSR-G(240) from the 50-kDa domain contrib-
utes to the binding of ATP.
The actin-binding region of the myosin head is
formed largely by the 50-kDa segment, which is split
by a deep cleft into two separate domains (Fig. 19-15),
both of which are thought to participate in binding to
actin. A surface loop (loop 1) near the ATP-binding
site at the junction of the 25- and 50-kDa regions
affects the kinetic properties of myosin, probably by
influencing product release. A second loop (loop 2,
residues 626 647) at the junction of the 50- and 20-kDa
regions interact with actin. Loop 2 contains a GKK
sequence whose positive charges may interact with
negative charges in the N-terminal part of actin.
162 164
The C-terminal fragment of myosin contains a
globular domain that interacts with both the 20-kDa
and 50-kDa regions and contains an -helical neck
that connects to the helix of the coiled-coil myosin rod.
This helical region is surrounded by the two myosin
light chains (Fig. 19-15).
157
A pair of reactive thiol
groups (from C697 and C707) in the globular domain
are near the active site. Crosslinking of these cysteines
by an S S bridge has been utilized to trap nucleotide
analogs in the active site.
165
How does actin bind? The actin monomer con-
sists of four subdomains, 1, 2, 3, and 4 numbered from
the N terminus (Fig. 7-10). The negatively charged
N-terminal region of actin contains the sequence
A
B
D E
4 25 24
D E D D.
K185
T186
457
S237
3
1
4
G182
W3
2
S236
E459
R238
K185
T186
457
S237
3
1
4
W3
2
G182
E459
S236
R238
Mg
++
Mg
++
Ch 19IND page -E- 2/14/03, 11:23 AM 1106
1107
It may interact with loop 1 of myosin, which contains
five lysines. However, to form a strong interaction
with the myosin head a conformational change must
occur in the myosin. A change may also occur in actin.
Modeling suggests that a large nonpolar contact region
involves actin residues A144, I341, I345, L349, and
F352 and myosin residues P529, M530, I535, M541,
F542, and P543. A conformational change in actin,
which might involve largely the highly conserved
actin subdomain 2, may also be required for tight
interaction.
142,166 168
Kinesins and other molecular motors. Before
considering further how the myosin motor may work,
we should look briefly at the kinesins, a different
group of motor molecules,
168a
which transport various
cellular materials along microtubule rails. They also
participate in organization of the mitotic spindle and
other microtubule-dependent activities.
168a,b,c
See
Section C,2 for further discussion. More than 90 mem-
bers of the family have been identified. Kinesin heads
have much shorter necks than do the myosin heads.
A myosin head is made up of ~ 850 residues, but the
motor domain of a kinesin contains only ~ 345. Like
Figure 19-17 Ribbon drawing of human kinesin with
bound MgADP. From Gulick et al.
174
Courtesy of Ivan
Rayment and Andy Gulick.
myosin, the 950- to 980-residue kinesins have a long
coiled-coil C- terminal region that forms a neck of
~ 50 residues, a stalk of ~ 190 and ~ 330 residue seg-
ments with a Pro / Gly-rich hinge between them, and
an ~ 45 residue tail.
169171
Crystal structures are known for motor domains of
human kinesin
172
and of a kinesin from rat brain.
169,173
The structures of one of six yeast kinesins,
174
a protein
called Kar3, and also of a Drosophila motor molecule
designated Ncd have also been determined.
175
The last
was identified through study of a Drosophila mutant
called non-claret disjunctional (Ncd). The motor
domains of various members of the kinesin family
show ~ 40% sequence identity and very close structural
identity (Fig. 19-17).
174
Although the sequences are
different from those of the myosin heads or of G proteins,
the folding pattern in the core structures is similar in
all cases. An 8-stranded sheet is flanked by three
helices on each side and a P-loop crosses over the ATP-
binding site as in Fig. 19-16. Further similarity is found
in the active site structures, which, for a monomeric
kinesin KIF1A,
174a
have been determined both with
bound ADP and with a nonhydrolyzable analog of
ATP.
174b,174c
Although there is little similarity in amino
acid sequences the structures in the catalytic core
are clearly related to each other, to those of dimeric
kinesins,
174d
to those of myosins, and to those of the
GTP-hydrolyzing G proteins.
A puzzling discovery was that the motor domain
of kinesin, which binds primarily to the subunits of
tubulin (Fig. 7-34) and moves toward the fast growing
plus end of the microtubule,
176
is located at the N ter-
minus of the kinesin molecule, just as is myosin. How-
ever, the Ncd and Kar3 motor domains are at the C-
terminal ends of their peptide chains and move their
cargos toward the minus ends of microtubules.
174
Nevertheless, the structures of all the kinesin heads
are conserved as are the basic chemical mechanisms.
The differences in directional preference are determined
by a short length of peptide chain between the motor
domain and the neck, which allows quite different geo-
metric arrangements when bound to microtubules.
173,177
Like Ncd, myosin VI motor domains also move back-
wards toward the pointed (minus) ends of actin
filaments.
178179a
Other major differences between kinesins and myo-
sin II heads involve kinetics
180,181
and processivity.
173
Dimeric kinesin is a processive molecule. It moves
rapidly along microtubules in 8-nm steps but remains
attached.
182,182a
Myosins V and VI are also proces-
sive
183183e
but myosin II is not. It binds, pulls on actin,
and then releases it. The many myosin heads interact-
ing with each actin filament accomplish muscle con-
traction with a high velocity in spite of the short time
of attachment. Ncd and Kar3 are also nonprocessive
and slower than the plus end-oriented kinesins.
184
B. Muscle
Ch 19IND page -E- 2/14/03, 11:23 AM 1107
1108 Chapter 19. The Chemistry of Movement
The ATPase cycles of actomyosin and of the
kinesins. The properties of the protein assemblies
found in muscle have been described in elegant details,
but the most important question has not been fully
answered. How can the muscle machinery use the
Gibbs energy of hydrolysis of ATP to do mechanical
work? Some insight has been obtained by studying
the ATPase activity of isolated myosin heads (S1) alone
or together with actin. Results of numerous studies of
ATP binding, hydrolysis, and release of products using
fast reaction techniques
185191
and cryoenzymology
191a
are summarized in Fig. 19-18. In resting muscle the
myosin heads swing freely in the ~ 20-nm space be-
tween the thick and thin filaments. However, in acti-
vated muscle some heads are bound tightly to actin as
if in rigor (complex AM in Fig. 19-18). When ATP is
added MgATP binds into the active site of the myosin
(Fig. 19-18, step a) inducing a conformational change
to form AM*ATP in which the bond between actin
and myosin is weakened greatly, while that between
myosin and ATP is strengthened. The complex disso-
ciates (step b) to give free actin and (M*ATP), which
accumulates at 15C. However, at higher tempera-
tures the bound ATP is hydrolyzed rapidly (step c) to a
form M**ADPP
i
in which the ATP has been cleaved
to ADP + P
i
but in which the split products remain
bound at the active site.
116,192,192a,b
All of these reac-
tions are reversible. That is, the split products can
recombine to form ATP. This fact suggests that most
of the Gibbs energy of hydrolysis of the ATP must be
stored, possibly through a conformational change in
the myosin head or through tighter bonding to ATP.
As long as calcium ions are absent, there is only a slow
release of the bound ADP and P
i
and replacement with
fresh ATP takes place. Thus, myosin alone shows a
very weak ATPase activity.
On the other hand, in activated muscle the head
with the split ATP products will bind to actin (step d),
probably at a new position. The crossbridges that
form appear to be attached almost at right angles to
the thin filaments. In step e, P
i
is released following
a conformational alteration that is thought to open a
back door to allow escape of the phosphate ion.
193
In the final two steps ( f and g) the stored energy in the
myosin head (or in the actin) is used to bring about
another conformational change that alters the angle
of attachment of myosin head to the thin filament from
~ 90 to ~ 45. At least some indication of such a change
can be observed directly by electron microscopy.
144
Such a change in angle is sufficient to cause the actin
filament to move ~ 10 nm with respect to the thick
filaments to complete the movement cycle (Fig. 19-18),
if the head is hinged at the correct place. However,
the existence of at least four different conformational
states suggests a more complex sequence.
193a,193b
Examination of the three-dimensional structures avail-
able also suggest a complex sequence of alterations in
structure and geometry. X-ray crystallographic struc-
tures of myosin heads, in states thought to correspond
to states 1 and 3 of Figs. 19-14 and 19-18, are also in
agreement on an ~10 (5 12) nm movement of the lever
arm.
194,195
Six states of the actomyosin complex are
depicted in Fig. 19-18, but a complete kinetic analysis
requires at least eight and possibly 12 states.
196,197
Observing single molecules. A major advance in
the study of molecular motors has been the develop-
ment of ways to observe and study single macromole-
cules. The methods make use of optical traps (optical
tweezers) that can hold a very small (~ 1 m diameter)
polystyrene or silica bead near the waist of a laser
beam focused through a microscope objective.
198 202
In one experimental design an F-actin filament is
stretched between two beads, held in a pair of optical
traps. The filament is pulled taut and lowered onto a
stationary silica bead to which a few myosin HMM
fragments have been attached (Fig. 19-19). If ATP is
present, short transient movements along the filament
are detected by observation of displacements of one
of the beads when the actin filament contacts HMM
heads. An average lateral displacement of 11 nm was
observed. Each HMM head exerted a force of 3 4 pN,
a value consistent with expectations for the swinging
bridge model.
200
From the duration of a single displace-
ment ( 7 ms) and an estimated k
cat
for ATP hydrolysis
of 10 s
1
, the fraction of time that the head was attached
during one catalytic cycle of the head was therefore
only 0.07. This ratio, which is called the duty ratio,
is low for actomyosin. However, many myosin heads
bind to each actin filament in a muscle. Each head
exerts its pull for a short time. but the actin is never
totally unattached.
203
Similar measurements with
smooth muscle revealed similar displacements but
with a 10-fold slower sliding velocity and a 4-fold
increase in the duty ratio. This may perhaps account
for an observed 3-fold increase in force as compared
with skeletal muscle.
204,204a
Other single molecule techniques involve direct
observation of motor molecules or of S1 myosin frag-
ments tagged with highly fluorescent labels.
205,206
All measurements of single molecule movement are
subject to many errors. Brownian motion of the beads
makes measurements difficult.
207
Not all results are in
agreement, and some are difficult to understand.
207a
Most investigators agree that there is a step size of
~410 nm. Kitamura et al. found 5.3 nm as the aver-
age.
206
However, they also reported the puzzling obser-
vation that some single S1 molecules moved 11 30 nm
in two to five rapid successive steps during the time of
hydrolysis of a single molecule at ATP. They suggested
that some of the energy of ATP hydrolysis may be
stored in S1 or in the actin filament and be released
in multiple steps. Veigel et al.
208
observed that a
brush border myosin I from chicks produced ~ 6 nm
Ch 19IND page -E- 2/14/03, 11:23 AM 1108
1109
Figure 19-18 Simplified view of the ATP hydrolysis cycle for actomyosin. A similar cycle can be written for kinesins and
dyneins. Here A stands for fibrous actin and M, M*, M**, and M*** for four different conformations of the myosin heads.
As indicated by the numbers in squares, four of the six states of actomyosin shown here can also be correlated with those in
Fig. 19-14.
movements, each of which was followed by an addi-
tional ~ 5.5 nm step within ~ 5 ms. They attribute
these steps to two stages in the power stroke, e.g., to
steps f and g of Fig. 19-18. A value of ~10nm was
reported recently by Piazzesi et al.
208a
Myosin V moves
along actin filaments with very large 36-nm steps.
208b
Motion of kinesin heads has been observed by
movement of microtubules over biotinylated kinesin
fixed to a steptavidin-coated surface,
209
by direct obser-
vation of fluorescent kinesin moving along microtu-
bules,
171
and by optical trap interferometry.
210
Kinesin
heads move by 8-nm steps, evidently the exact length
B. Muscle
a
b
c
d
e f
g
ATP
A M* ATP M* ATP
H
2
O
H
+
A M
A M ADP
ADP
(Tightly bound)
Power stroke
AM*** ADP
P
i
A M** ADP P
i
M** ADP P
i
Myosin head
attaches at a
new location on
actin filament
In all states ATP
represents MgATP
A
Myosin
head is
detached
from actin
6
4
3
1
Myosin
P
P
P
M Tight binding to
actin as in rigor
Myosin
P
P
P
M* Weak binding to actin
Tight binding to ATP
M** Closed active site.
ATP is in equilibrium
with ADP and P
i
.
Tight binding to actin.
P
P
P
Myosin
Myosin
P
P
P
P
P
M*** Metastable state;
energy stored in
myosin head
Power stroke
Myosin
P
P
P
2
slow
fast
5
fast
Ch 19IND page -E- 2/14/03, 11:23 AM 1109
1110 Chapter 19. The Chemistry of Movement
interactions bind the nucleotide. This is shown for a
kinase in Fig. 12-32 and for myosin in Fig. 19-16. Dur-
ing the actomyosin reaction several conformational
changes must occur. Not only does the affinity for the
bound nucleotide vary, but also the binding of actin to
myosin can be strong, as in the nucleotide-free state or
of an tubulin dimer in the microtubule structure
(Fig. 7-33). One ATP is apparently hydrolyzed for each
8-nm step. However, shorter steps of ~ 5 nm have
sometimes been seen.
211,212
The movement is processive, kinesin motors typi-
cally taking 100 steps before dissociating from the
microtubule.
201,212a
Kinesin is bound
to the microtubule continuously. Its
duty ratio is nearly 1.0 (the same is
true for the bacterial flagellar motor;
Fig. 19-4).
212b
However, single kine-
sin heads, which lack the coiled-coil
neck region, have a duty ratio of
< 0.45. The movement is nonpro-
cessive.
213
The Ncd motor is also
nonprocessive.
214 216
As mentioned
previously, the Ncd and kinesin
motor domains are at opposite ends
of the peptide chain, and the motors
move in opposite directions along
microtubules.
217,218
The critical
difference between the two motor
molecules was found in the neck
domains, which gave rise to differ-
ing symmetrics in the two heads.
219
The latter are shown in Fig. 19-20, in
which they have been docked onto
the tubulin protofilament structure.
One head, both of ncd and of kinesin,
occupies a similar position on the
microtubule, but the other head
points toward the microtubular plus end for kinesin
but toward the minus end for Ncd. Cryoelectron
microscopy also supports this interpretation.
220
Still not fully understood is the processive action
of kinesin.
221 224
It is often assumed that this protein
moves in a hand-over-hand fashion with the two heads
binding alternatively to the microtubule. Some sub-
stantial reorganization of the peptide chain in the
hinge region at the end of the neck is presumably
involved.
173
An alternative inchworm mechanism
has been suggested.
220a
Thinking about chemical mechanisms. We
have now examined the active sites of kinases that
cleave ATP (Chapter 12), ATPases that pump ions by
cleaving ATP, ATP synthases that form ATP from ADP
and P
i
(Chapter 18), and GTP hydrolyzing enzymes
that cause movement and shape changes that control
metabolic processes (Chapter 11). It is striking that the
active site regions where the ATP or GTP bind have
such a highly conserved structure.
225
This suggests
that the secret of movement can be found in the very
strong interactions of the nucleotides and their split
products with the proteins. In every case there is at
least one tight binding or closed conformation in
which a large number of hydrogen bonds and ionic
Figure 19-20 Model showing the ncd and kinesin dimer
structures docked onto a tubulin protofilament. The bound
ncd and kinesin heads are positioned similarly. Because of
the distinct architectures of the kinesin and ncd necks, the
unbound kinesin head points toward the plus end, whereas
the unbound ncd head is tilted toward the minus end of the
protofilament. From Sablin et al.
219
Courtesy of Ronald Vale.
Figure 19-19 Schematic drawing (not to scale) illustrating the use of two
optical traps that are focused on beads attached to a single actin filament.
The filament is lowered onto a stationary silica bead sparsely coated with
HMM fragments of myosin. In the presence of ATP the myosin heads bind
transiently for a few milliseconds to the actin, moving it in one direction and
displacing the beads from their positions in the optical traps. An image of one
of the beads is projected onto photodiode detectors capable of measuring small
displacements. The displacing force can also be recorded. For details of the
experiments and of the optical traps and measuring devices see Finer et al.
200
Courtesy of J. A. Spudich.
Ch 19IND page -E- 2/14/03, 11:24 AM 1110
1111
in the presence of bound ADP. Binding is weak when
ATP or the split product ADP + P
i
are in the active site.
To understand these differences we should look at
the structure of ATP itself. The triphosphate group has
many negative charges repelling each other. What
must happen to allow the binding of ATP to myosin
to break the actin-myosin bond? The electrostatic
attraction of these phospho groups for active site
groups is doubtless one cause of the observed confor-
mational changes. Could it be that electrostatic repul-
sion, via a proton shuttle mechanism, is also induced
at the right point in the actinmyosin interface? Many
studies with analogs of ATP have contributed to our
understanding. Neither the purine nor the ribose
ring of ATP is absolutely essential. The compound
2-[(4-azido-2-nitrophenyl) amino] diphosphate
(NANDP) and related nonnucleotide analogs
165,196,226
support muscle contraction and relaxation in the same
way as does ATP. An analog with a rigid five-membered
ring, 2',3'-dideoxydidehydro-ATP, is also active.
227
A
comparison of kinetic data and X-ray structural data
supports the proposal that the ATP must be bound
in the conformation shown for MgADPBeF
x
in Fig.
19-16A.
196
When the two SH groups of C697 and
C707 of the myosin head are crosslinked by various
reagents,
227a
this NANDP analog can be trapped at the
active site. Because of the presence of its azide ( N
3
)
group the trapped compound can serve as a photo-
affinity label, attaching itself to a tryptophan side
chain upon activation with visible light (Eq. 23-27).
How can cleavage of ATP to bound ADP + P
i
create a metastable high-energy state of the myosin
head ready to hold onto and pull the actin chain? This
may be compared with the inverse problem of gener-
ating ATP in oxidative phosphorylation, in which ADP
and P
i
coexist in equilibrium with ATP in a closed
active site (Fig. 18-14). Comparison should also be made
with the GTP-hydrolyzing G proteins (Fig. 11-7).
227b,c
During hydrolysis of GTP by the Ras protein binding
to the protein induces a shift of negative charge from
the - to -oxygens of GTP facilitating bond cleavage
as in Eq. 19-2. G proteins also couple substrate hydrol-
ysis to mechanical motion. We should also think
O
P
O
P
O
P
O
O O O O O O CH
2
CH
2
N
H
N
O
O
N
3

2
NANTP

about the fact that when ATP is cleaved within myosin


there will necessarily be a flow of electrical charge
from the water to the ADP (Eq. 19-2). This will be
followed by some accomodation of the protein to the
new charge constellation. As we have seen previously,
movement of protons is often the key to conformational
changes. In this case, the initial change must be to
create a high-energy state of myosin which, following
loss of the orthophosphate ion, can cause the major
conformational change that swings the lever arm of
the myosin. The conformational changes may occur
in several steps in which the packing of groups within
the myosin head is always tight in some places and
rather loose in others. Movement within the head is
being observed not only by X-ray crystallography but
by fluorescence resonance energy transfer (FRET;
Chapter 23)
227d
and by the newer luminescence
resonance energy transfer (LRET). For example, a
terbium chelate of azide-ATP was photochemically
bound in the active site, and a fluorescent dye was
attached to Cys 108 in the regulatory light chain. The
terbium ion was irradiated, and fluorescence of the
dye was observed. Distance changes, measured in
the absence and presence of ATP, were consistent with
the swinging arm model.
228
Dyes have been attached
to SH groups engineered into various locations in the
myosin molecule to permit other distance measure-
ments.
229,230
In another elegant application of the FRET
technique the green fluorescent protein of Aequoria
(Box 23-A) was fused to the C terminus of the motor
domain of myosin giving a fluorescent lever arm.
Energy transfer to blue fluorescent protein fused to
the N terminus of the S1 head was measured. The
distance between these was estimated by the FRET
technique and was also consistent with expectations
for the rowing model.
231
The rowing model is generally accepted, but
other quite different processes have been proposed to
account for the elementary cycle of muscle contraction.
Muscle contracts nearly isovolumetrically; thus, anything
that expands the sarcomere will cause a contraction.
It has been suggested that the hydrolysis of ATP deposits
negatively charged phospho groups on the actin filaments,
and that the electrostatic repulsion is responsible for
B. Muscle
(19-2)
P
O
P
O
O P
O
O
O
O
Adenosine
O
O
O
Mg
2
H
O
H
O
P
O
O P
O
O
O
O
Adenosine

P
O
O
O
O


+ +
H
H
+
Ch 19IND page -E- 2/14/03, 11:24 AM 1111
1112 Chapter 19. The Chemistry of Movement
Considering the numerous specialized proteins in
muscle it is not surprising that many rare hereditary
muscle diseases are known. The most frequent and most
studied of these is Duchenne muscular dystrophy.
An X-linked disease of boys, it may not be recognized
until two to three years of age, but victims are usually
in a wheelchair by age 12 and die around age 20.
Individual muscle fibers disintegrate, die, and are
replaced by fibrous or fatty tissue.
a d
The disease
strikes about 1 out of 3500 boys born. The less serious
Becker muscular dystrophy arises from defects in
the same gene but affects only 1 in 30,000 males, some
of whom have a normal life span. Because of its
frequency and the knowledge that the gene must lie
in the X-chromosome, an intensive search for the gene
was made. It was found in 1986 after a five-year
search.
a,e
This was the first attempt to find a faulty
gene whose protein product was totally unknown.
The project, which relied upon finding restriction
fragment polymorphisms (Chapter 26) that could
serve as markers in the genome, made use of the
DNA from patients with a range of related diseases.
The very rare female patients in whom the faulty
gene had been translocated from the X-chromosome
to an autosome also provided markers. DNA probes
obtained from a young man with a large X-chromo-
some deletion that included genes related to retinitis
pigmentosa and several other diseases provided
additional markers. The result was a triumph of
reverse genetics which has since been applied to
the location of many other disease genes.
e
The muscular dystrophy gene may be the largest
human gene. It consists of 2.3 million base pairs,
which include 79 exons which encode a huge 427 kDa
protein named dystrophin. The protein consists of
four main domains.
a,f,g
The N-terminal domain binds
to actin and is homologous to -actinin. The central
domain is an elongated rod resembling spectrin. It
contains repetitive coiled-coil segments and four
hinge regions. The third domain is rich in cysteine
and binds Ca
2+
, while the fourth domain has a struc-
ture that is shared by several other proteins of the
dystrophin family. Dystrophin is quantitatively a
minor protein of muscle. It forms part of the cyto-
skeleton, lying adjacent to the sarcolemma (cell mem-
brane) along with -spectrin and vinculin (see Fig.
8-16).
While one end of the dystrophin molecule binds
to actin filaments, the C-terminal domain associates
with several additional proteins to form a dystrophin
glycoprotein complex (see figure).
f,h k
Dystrophin
is linked directly to the membrane-spanning protein
-dystroglycan, which in the outer membrane sur-
faces associates with a glycoprotein -dystroglycan.
The latter binds to laminin-2 (Fig. 8-33), a protein
that binds the cell to the basal lamina. Four
BOX 19-A HEREDITARY DISEASES OF MUSCLE
other membrane-spanning proteins, -, -, -, and
-sarcoglycans, are among additional members of
the complex.
km
Patients with Duchenne muscular dystrophy are
deficient not only in dystrophin but also in the dystro-
glycan and sarcoglycan proteins.
f,n
Evidently, dystro-
phin is needed for formation of the complex which
plays an essential role in muscle. In both types of
X-linked muscular dystrophy there are individuals
with a wide range of point mutations, frame-shift
mutations, and deletions in the dystrophin gene.
d
The essential function of dystrophin and associated
proteins is uncertain but may be related to the linkage
from actin filaments through the membrane to lami-
nin. Individuals with Becker muscular dystrophy
also have defects in dystrophin, but the protein is
partially functional. Some other muscular dystro-
phies are caused by defects in the autosomal genes
of any of the four sarcoglycan subunits.
j,k,o
or in that
of laminin 1 chain.
p,q
The arrows in the accompany-
ing drawing indicate chromosome locations of the
sarcoglycan subunits, which are sites for mutations
causing limb girdle muscular dystrophy.
k
Dystrophin, shorter isoforms, and related pro-
teins are found in many tissues including the brain.
s
One related protein, utrophin (dystrophin-related
Schematic model of the dystrophinglycoprotein complex.
Courtesy of Kevin P. Campbell. See Lim and Campbell
r
Abbreviations: LGMD, Limb Girdle muscular dystrophy;
CMD, congenital muscular dystrophy; DMD / BMD,
Duchenne / Becker muscular dystrophies.
Ch 19IND page -E- 2/14/03, 11:24 AM 1112
1113
BOX 19-A (continued)
protein 1), is present in the neuromuscular junction
of adult skeletal muscle. One approach to therapy of
Duchenne muscular dystrophy is to stimulate a higher
level of expression of the utrophin gene.
t
Because the
dystrophin gene is so large treatment by gene transfer
is not practical, but transfer of parts of the gene may
be. Myoblast transfer has not been successful, but
new approaches will be devised.
d
Myotonic dystrophy is a generalized adult-onset
disorder with muscular spasms, weakness, and many
other symptoms.
u y
It is one of the triple-repeat
diseases (Table 26-4). The affected gene encodes a
protein kinase of unknown function. The correspond-
ing mRNA transcript has ~ 2400 nucleotides. The
gene has a CTG repeat (CTG)
n
near the 3'-end with
n < 30 normally. For persons with the mildest cases
of myotonic dystrophy n may be over 50 while in
severe cases it may be as high as 2000. As in other
triple-repeat diseases the repeat number tends to
increase in successive generations of people as does
the severity of the disease.
x
For some individuals, muscular dystrophy causes
no obvious damage to skeletal muscle but affects the
heart producing a severe cardiomyopathy, and
a
Anderson, M. S., and Kunkel, L. M. (1992) Trends Biochem. Sci.
17, 289292
b
Emery, A. E. H., and Emery, M. L. H. (1995) The History of a
Genetic Disease: Duchenne Muscular Dystrophy or Meryons
Disease, Royal Society of Medicine, London
c
Brown, S. C., and Lucy, J. A., eds. (1997) Dystrophin: Gene,
Protein and Cell Biology, Cambridge Univ. Press, London
d
Worton, R. G., and Brooke, M. H. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 3 (Scriver, C.
R., Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 4195
4226, McGraw-Hill, New York
e
Rowland, L. P. (1988) N. Engl. J. Med. 318, 13921394
f
Tinsley, J. M., Blake, D. J., Zuellig, R. A., and Davies, K. E.
(1994) Proc. Natl. Acad. Sci. U.S.A. 91, 83078313
g
Fabbrizio, E., Bonet-Kerrache, A., Leger, J. J., and Mornet, D.
(1993) Biochemistry 32, 1045710463
h
Ervasti, J. M., and Campbell, K. P. (1991) Cell 66, 11211131
i
Madhavan, R., and Jarrett, H. W. (1995) Biochemistry 34,
1220412209
j
Sweeney, H. L., and Barton, E. R. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 1346413466
k
Jung, D., and 13 others. (1996) J. Biol. Chem. 271, 3232132329
l
Winder, S. J. (2001) Trends Biochem. Sci. 26, 118124
m
McDearmon, E. L., Combs, A. C., and Ervasti, J. M. (2001)
J. Biol. Chem. 276, 3507835086
n
Durbeej, M., and Campbell, K. P. (1999) J. Biol. Chem. 274,
2660926616
o
Yang, B., Ibraghimov-Beskrovnaya, O., Moomaw, C. R.,
Slaughter, C. A., and Campbell, K. P. (1994) J. Biol. Chem. 269,
60406044
p
Noguchi, S., and 17 others. (1995) Science 270, 819822
q
Tiger, C.-F., Champliaud, M.-F., Pedrosa-Domellof, F., Thor-
nell, L.-E., Ekblom, P., and Gullberg, D. (1997) J. Biol. Chem.
272, 2859028595
r
Lim, L. E., and Campbell, K. P. (1998) Curr. Opin. Neurol. 11,
443452
B. Muscle
s
Dixon, A. K., Tait, T.-M., Campbell, E. A., Bobrow, M., Roberts,
R. G., and Freeman, T. C. (1997) J. Mol. Biol. 270, 551558
t
Campbell, K. P., and Crosbie, R. H. (1996) Nature (London) 384,
308309
u
Harper, P. S. (1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 3 (Scriver, C. R., Beaudet, A. L.,
Sly, W. S., and Valle, D., eds), pp. 42274252, McGraw-Hill,
New York
v
Ptacek, L. J., Johnson, K. J., and Griggs, R. C. (1993) N. Engl.
J. Med. 328, 482489
w
Pearson, C. E., and Sinden, R. R. (1996) Biochemistry 35, 5041
5053
x
Tapscott, S. J., and Thornton, C. A. (2001) Science 293, 816817
y
Miller, J. W., Urbinati, C. R., Teng-umnuay, P., Stenberg, M. G.,
Byrne, B. J., Thornton, C. A., and Swanson, M. S. (2000) EMBO
J. 19, 44394448
z
Olson, T. M., Michels, V. V., Thibodeau, S. N., Tai, Y.-S., and
Keating, M. T. (1998) Science 280, 750755
aa
Rayment, I., Holden, H. M., Sellers, J. R., Fananapazir, L., and
Epstein, N. D. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 38643868
bb
Roopnarine, O., and Leinwand, L. A. (1998) Biophys. J. 75,
30233030
cc
Yanaga, F., Morimoto, S., and Ohtsuki, I. (1999) J. Biol. Chem.
274, 88068812
dd
Martinsson, T., Oldfors, A., Darin, N., Berg, K., Tajsharghi, H.,
Kyllerman, M., and Wahlstrm, J. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 1461414619
ee
Miller, T., Szczesna, D., Housmans, P. R., Zhao, J., de Freitas,
F., Gomes, A. V., Culbreath, L., McCue, J., Wang, Y., Xu, Y.,
Kerrick, W. G. L., and Potter, J. D. (2001) J. Biol. Chem. 276,
37433755
ff
Kobayashi, K., and 17 others. (1998) Nature (London) 394,
388392
gg
Pelin, K., and 19 others. (1999) Proc. Natl. Acad. Sci. U.S.A. 96,
23052310
hh
Weil, D., and 18 others. (1995) Nature (London) 374, 6061
persons with Duchenne muscular dystrophy often die
from heart failure. Heart failure from other causes,
some hereditary, is a major medical problem, especially
among older persons. Hereditary forms are often
autosomal dominant traits that may cause sudden
death in young persons. At least seven genes for
cardiac sarcomeric proteins including actin,
z
myosin,
both heavy and light chains,
aa dd
three subunits of
troponin,
ee
tropomyosin, and protein C (p. 1104) may
all carry mutations that cause cardiomyopathy.
A hereditary disease common in Japan results
from a defect in migration of neurons and is associated
with brain malformation as well as muscular dystrophy.
ff
In nemaline myopathy a defect in nebulin leads to
progressive weakness and often to death in infancy.
A characteristic is the appearance of nemaline bodies
or thickened Z-discs containing Z-disc proteins.
gg
Some hereditary diseases involve nonmuscle myosins.
Among these is Usher syndrome, the commonest
cause of deaf-blindness. The disease, which results
from a defect in the myosin VIA gene, typically causes
impairment of hearing and retinitis pigmentosa
(Chapter 23).
hh
Ch 19IND page -E- 2/14/03, 11:24 AM 1113
1114 Chapter 19. The Chemistry of Movement
Extracellular
Cytoplasm
T-tubule
membrane
SR lumen
SR
membrane
lateral expansion of the sarcomere.
232
Still other ideas
have been advanced.
233,234
4. Control of Muscle Contraction
Skeletal muscle must be able to rest without
excessive cleavage of ATP but able to act rapidly with
a high expenditure of energy upon nervous excitation.
Even a simple physical activity requires that a persons
muscles individually contract and relax in rapid re-
sponse to nerve impulses from the brain. To allow for
this control the endoplasmic reticulum (sarcoplasmic
reticulum, SR) of striated muscle fibers is organized
in a striking regular manner.
235 237
Interconnecting
tubules run longitudinally through the fibers among
the bundles of contractile elements. At regular intervals
they come in close contact with infoldings of the outer
cell membrane (the T system of membranes, Fig. 19-21;
see also Fig. 19-7A). A nerve impulse enters the muscle
fiber through the neuromuscular junctions and travels
along the sarcolemma and into the T tubules. At the
points of close contact the signal is transmitted to the
longitudinal tubules of the sarcoplasmic reticulum,
which contain a high concentration of calcium ions.
Calcium ions in muscle. A nerve signal arriving
at a muscle causes a sudden release of the calcium ions
into the cytoplasm from cisternae of the sarcoplasmic
reticulum (SR) that are located adjacent to the T-tubules.
Diffusion of the Ca
2+
into the myofibrils initiates con-
traction. In smooth muscle the signals do not come
directly from the nervous system but involve hormonal
regulation.
238
Again, calcium ions play a major role,
which is also discussed in Chapter 6, Section E, and
in Box 6-D. Muscle contains a large store of readily
available Ca
2+
in lateral cisternae of the SR. The free
intracellular Ca
2+
concentration is kept low by a very
active ATP-dependent calcium ion pump (Fig. 8-26),
which is embedded in the membranes of the SR.
238a
Within the vesicles Ca
2+
is held loosely by the ~ 63-kDa
protein calsequestrin, which binds as many as 50
calcium ions per molecule. When the cytoplasmic
concentration of free Ca
2+
falls below ~ 10
6
M, contrac-
tion ceases. In fast-contracting skeletal muscles the
Ca
2+
-binding protein parvalbumin (Fig. 6-7) may
Figure 19-21 (A) Electron micrograph showing two transverse tubules (T-tubules) that are formed by infolding of the plas-
ma membrane. They wrap around a skeletal muscle fiber and carry nerve impulses to all parts of the fiber. From Alberts et
al.
237
Courtesy of Clara Franzini-Armstrong. (B) Three-dimensional surface representation of the calcium release channels
known as ryanodine receptors, type RyR1 based on cryoelectron microscopy and image reconstruction at a resolution of 4.1
nm. The image to the left shows the surface that would face the cytoplasm while that to the right shows the surface that
would interact with the sarcoplasmic reticulum, TA representing the transmembrane portion. Notice the fourfold symmetry of
the particle, which is composed of four 565-kDa subunits. From Sharma et al.
239
Courtesy of Manjuli Rani Sharma. (C), (D)
Model showing proposed arrangement of ryanodine receptors and dihydropyridine receptors (round) in the T tubule and SR
membranes. From Serysheva et al.
245a
B
Myofibril
Transverse (T) tubules
formed from invaginations
of plasma membrane
Sarcoplasmic reticulum
lateral cisternum 500 nm
Ca
2+
release channels
A C
D
Ch 19IND page -E- 2/14/03, 11:24 AM 1114
1115
assist in rapid removal of free Ca
2+
from the cytoplasm.
Contraction is activated when Ca
2+
is released from
the SR through the calcium release channels,
240 244
which are often called ryanodine receptors. The
name arises from their sensitivity to the insecticidal
plant alkaloid ryanodine, which at low concentration
( 60 M) causes the channels to open, but a high
concentration closes the channels.
243
These calcium
release channels consist of tetramers of ~ 5000-residue
proteins. The bulk of the 565-kDa polypeptides are on
the cytosolic surface of the SR membranes, where they
form a complex foot structure (Fig. 19-21B) that
spans the ~ 12 nm gap between the SR vesicles and the
T-tubule membrane.
239,240
Ryanodine receptor function
is modulated by NO, which apparently binds to SH
groups within the Ca
2+
channel.
243a,243b
Some ryano-
dine receptors are activated by cyclic ADP-ribose
(cADPR, p. 564).
243c
Some have an oxidoreductase-
like structural domain.
243d
The release channels open in response to an in-
completely characterized linkage to the voltage sensor
that is present in the T-tubule membrane and is known
as the dihydropyridine receptor.
240,245
This too, is a
Ca
2+
channel, which opens in response to arrival of an
action potential (nerve impulse; see Chapter 30) that
move along the T-tubule membrane. Because the
action potential arrives almost simultaneously
throughout the T-tubules of the muscles, the dihydro-
pyridine receptors all open together. It isnt clear
whether the linkage to the calcium channels is via
stimulation from released Ca
2+
passing from the dihy-
dropyridine receptor to the surfaces of the feet of the
release channels, or is a result of depolarization of the
T-tubule membrane, or involves direct mechanically
linked conformational changes.
240,245
The close coop-
eration of the Ca
2+
release channel and the voltage
sensor is reflected in their close proximity. In the
sarcoplasmic reticulum every second release channel
is adjacent to a voltage sensor in the opposing T-tubule
membrane.
240,245a
The essential nature of the voltage
B. Muscle
N
H
O
O
H
3
C
CH
3
CH
3
O
OH
CH
3
H
OH
OH
HO
H
OH
HO
Ryanodine
BOX 19-B MALIGNANT HYPERTHERMIA AND STRESS-PRONE PIGS
Very rarely during surgery the temperature
of a patient suddenly starts to rise uncontrollably.
Even when heroic measures are taken, sudden
death may follow within minutes. This malignant
hyperthermia syndrome is often associated with
administration of halogenated anesthetics such as a
widely used mixture of halothane (2-bromo-2-chloro-
1,1,1-trifluoroethane) and succinylcholine.
a d
There
is often no warning that the patient is abnormally
sensitive to anesthetic. However, development of
an antidote together with increased alertness to
the problem has greatly decreased the death rate.
Nevertheless, severe damage to nerves and kidneys
may still occur.
c
Biochemical investigation of the
hyperthermia syndrome has been facilitated by the
discovery of a similar condition that is prevalent
among certain breeds of pigs. Such stress-prone
pigs are likely to die suddenly of hyperthermia
induced by some stress such as shipment to market.
The sharp rise in temperature with muscles going
into a state of rigor is accompanied by a dramatic
lowering of the ATP content of the muscles.
The problem, both in pigs and in humans
susceptible to malignant hyperthermia, was found
in the Ca
2+
release channels (ryanodine receptors).
Study of inheritance in human families together
a
Gordon, R. A., Britt, B. A., and Kalow, W., eds. (1973) Interna-
tional Symposium on Malignant Hyperthermia, Thomas, Spring-
field, Illinois
b
Clark, M. G., Williams, C. H., Pfeifer, W. F., Bloxham, D. P.,
Holland, P. C., Taylor, C. A., and Lardy, H. A. (1973) Nature
(London) 245, 99 101
c
MacLennan, D. H., and Phillips, M. S. (1992) Science 256, 789 794
d
Simon, H. B. (1993) N. Engl. J. Med. 329, 483 487
e
Fujii, J., Otsu, K., Zorzato, F., de Leon, S., Khanna, V. K., Weiler,
J. E., OBrien, P. J., and MacLennan, D. H. (1991) Science 253,
448 451
f
MacLennan, D. H., Duff, C., Zorzato, F., Fujii, J., Phillips, M.,
Korneluk, R. G., Frodis, W., Britt, B. A., and Worton, R. G.
(1990) Nature (London) 343, 559 561
g
Sakube, Y., Ando, H., and Kagawa, H. (1997) J. Mol. Biol. 267,
849 864
with genetic studies in pigs led to the finding that
the stress-prone pigs have cysteine replacing argin-
ine 615 in the Ca
2+
channel protein. This modifica-
tion appears to facilitate opening of the channels but
to inhibit their closing.
e
A similar mutation has
been found in some human families in which the
condition has been recognized. However, there is
probably more than one site of mutation in humans.
c,f
Similar mutations in the nematode C. elegans are
being investigated with the hope of shedding light
both on the problem of hyperthermia and on the
functioning of the Ca
2+
release channels.
g
Ch 19IND page -E- 2/14/03, 11:24 AM 1115
1116 Chapter 19. The Chemistry of Movement
sensor is revealed by a lethal mutation (muscular
disgenesis) in mice. Animals with this autosomal
recessive trait generate normal action potentials in the
sarcolemma but Ca
2+
is not released and no muscular
contraction occurs. They lack a 170-kDa dihydro-
pyridine-binding subunit of the sensor.
246
Some aspects of regulation by calcium ions are
poorly understood. The frequent observations of
oscillations in [Ca
2+
] in cells is described in Box 6-D.
Another phenomenom is the observation of Ca
2+
sparks, detected with fluorescent dyes and observa-
tion by confocal microscopy.
247
These small puffs of
Ca
2+
have been seen in cardiac muscle
247
and in a
somewhat different form in smooth muscle.
248
They
may represent the release of Ca
2+
from a single release
channel or a small cluster of channels. When the calcium
release channels open, Ca
2+
ions flow from the cisternae
of the SR into the cytoplasm, where they activate both
the troponintropomyosin system and also the Ca
2+
-
calmodulin-dependent light chain protein kinase,
which acts on the light chains of the myosin head.
These light chains resemble calmodulin in their Ca
2+
-
binding properties. The function of light chain phos-
phorylation of skeletal muscle myosin is uncertain but
it is very important in smooth muscle.
248a
The regulatory complex of tropomyosin and tro-
ponin is attached to the actin filaments as indicated
in Fig. 19-8D and also in Fig. 19-9. The latter shows
a model at near atomic resolution but without side
chains on the tropomyosin and without the troponin
components. When the regulatory proteins are com-
pletely removed from the fibrils, contraction occurs
until the ATP is exhausted. However, in the presence
of the regulatory proteins and in the absence of calcium,
both contraction and hydrolysis of ATP are blocked.
Tropomyosin (Tm) is a helical coiled-coil dimer, a 40-nm
rigid rod, in which the two 284-residue 33-kDa mono-
mers have a parallel orientation (Fig. 19-9)
249
as in the
myosin tail. However, an 8 to 9 residue overlap at
the ends may permit end-to-end association of the Tm
molecules bound to the actin filament. As with other
muscle proteins there are several isoforms,
250,251
whose
distribution differs in skeletal and smooth muscle and
in platelets. The elongated Tm rods appear to fit into
the grooves between the two strands of actin mono-
mers in the actin filament.
252 254
In resting muscle the
Tm is thought to bind to actin near the site at which
the S1 portion of the myosin binds. As a consequence,
the Tm rod may block the attachment of the myosin
heads to actin and prevent actin-stimulated hydrolysis
of ATP. The 40-nm Tm rod can contact about seven actin
subunits at once (Fig. 19-9). Thus, one Tmtroponin
complex controls seven actin subunits synchronously.
Troponin (Tn) consists of three polypeptides (TnC,
TnI, TnT) that range in mass from 18 to 37 kDa. The
complex binds both to Tm and to actin.
255,256
Peptide
TnT binds tightly to Tm and is thought to link the
TnITnC complex to Tm.
256,257
TnI interacts with
actin and inhibits ATPase activity in the absence of
Ca
2+
.
258 261
It may work with the other two peptides
to keep the Tm in the proper position to inhibit ATP
hydrolysis. TnC binds calcium ions. This ~160-residue
protein has a folding pattern almost identical to that of
calmodulin (Fig. 6-8) with four Ca
2+
-binding domains
arranged in two pairs at the ends of a long 9-turn helix.
When Ca
2+
binds to TnC, a conformational change
occurs
258,259,262-265
(p. 313). This induces changes in
the troponintropomyosinactin complex, releases the
inhibition of actomyosin ATPase, and allows contrac-
tion to occur.
265a
In the heart additional effects are
exerted by -adrenergic stimulation, which induces
phosphorylation of two sites on TnI by the action of
the cAMP-dependent protein kinase PKA. Dephos-
phorylation by protein phosphatase 2A completes a
regulatory cycle in which the doubly phosphorylated
TnI has a decreased sensitivity to calcium ions.
266
Cardiac muscle also contains a specialized protein
called phospholamban. An oligomer of 52-residue
subunits, it controls the calcium ion pump in response
to -adrenergic stimulation. Unphosphorylated phos-
pholamban inhibits the Ca ATPase, keeping [Ca
2+
]
high in the cytoplasm. Phosphorylation of phospho-
lamban by cAMP and/or calmodulin-dependent protein
kinase activates the Ca
2+
pump,
267268a
removing Ca
2+
and ending contraction.
X-ray diffraction and electron microscopy in the
1970s suggested that when calcium binds to troponin
the tropomyosin moves through an angle of ~ 20 away
from S1, uncovering the active site for the myosin
ATPactin interactions.
252,253
Tropomyosin could be
envisioned as rolling along the surface of the actin,
uncovering sites on seven actin molecules at once.
Side-chain knobs protruding from the tropomyosin
like teeth on a submicroscopic gear might engage
complementary holes in the actin. At the same time a
set of magnesium ion bridges between zones of nega-
tive charge on tropomyosin and actin could hold the
two proteins together. This proposal has been difficult
to test. Although the older image reconstruction is
regarded as unreliable, recent work still supports this
steric blocking model.
255,269-270c
Image reconstruction
from electron micrographs of thin filaments shows
that, in the presence of Ca
2+
, the tropomyosin does
move 25 away from the position in low [Ca
2+
]. How-
ever, instead of two states of the thin filament, on
and off, there may be at least three, which have been
called blocked, closed, and open.
255,269,271,271a
The closed state may be attained in rigor.
269
In addi-
tion, the possibility that changes in the conformation
of actin as well as of myosin occur during the contrac-
tion cycle must be considered.
255
Ch 19IND page -E- 2/14/03, 11:24 AM 1116
1117
Smooth muscle. The primary regulation of smooth
muscle contraction occurs via phosphorylation of the
Ser 19 OH group in the 20-kDa regulatory light chains
of each myosin head.
121b,160,272 274
The phosphorylated
form is active, participating in the contraction process.
Removal of Ca
2+
by the calcium pump and dephospho-
rylation of the light chains by a protein phosphatase
275
restores the muscle to a resting state. The N-terminal
part of the myosin light chain kinase binds to actin,
while the catalytic domain is in the center of the pro-
tein. The C-terminal part binds to myosin, and this
binding also has an activating effect.
276
Another protein, caldesmon, binds to smooth
muscle actin and blocks actomyosin ATPase.
271,277278a
It is present in smooth muscle in a ratio of actin:tropo-
myosin:caldesmon of ~14:2:1. Inhibition can be reversed
by Ca
2+
, but there is no agreement on the function of
caldesmon.
277
It is an elongated ~ 756-residue protein
with N-terminal domain, which binds to myosin, and
a C-terminal domain, which binds to actin, separated
by a long helix.
278
Caldesmon may be a substitute for
troponin in a tropomyosin-type regulatory system, or
it may promote actomyosin assembly. Another possi-
bility is that it functions in a latch state, an energy-
economic state of smooth muscle at low levels of ATP
hydrolysis.
278,279
In molluscan muscles Ca
2+
binds to a
myosin light chain and activates contraction directly.
Some molluscan smooth muscles (catch muscles)
also have a latch state, which enables these animals to
maintain muscular tension for long periods of time,
e.g., holding their shells tightly closed, with little
expenditure of energy.
279a
Catch muscles contain
myosin plus a second protein, catchin, which is
formed as a result of alternative mRNA splicing.
Catchin contains an N-terminal sequence that may
undergo phosphorylation as part of a regulatory
mechanism.
280
However, recent experiments indicate
that twitchin (see next paragraph), rather than catchin,
is essential to the catch state and is regulated by phos-
phorylation.
280a
Regulation of the large groups of
unconventional myosins is poorly understood. Phos-
phorylation of groups on the myosin heavy chains is
involved in ameba myosins and others.
281
An unexpected aspect of regulation was discovered
from study of the 40 or more genes of Caenorhabditis
elegans needed for assembly and function of muscle.
The mutants designated unc-22 showed a constant
twitch arising from the muscles in the nematodes
body. The gene was cloned using transposon tagging
(Chapter 27) and was found to encode a mammoth
753-kDa 6839-residue protein which has been named
twitchin.
282 285
Twitchin resembles titin (Fig. 19-8) and
like titin has a protein kinase domain, which is nor-
mally inhibited by the end of its peptide chain, which
folds over the active site of the kinase. Perhaps the
protein kinase activities of twitchin, titin, and related
proteins
285
are required in assembly of the sarcomere.
5. The Phosphagens
ATP provides the immediate source of energy for
muscles but its concentration is only ~ 5 mM. As dis-
cussed in Chapter 6, Section D, phosphagens, such
as creatine phosphate, are also present and may
attain a concentration of 20 mM in mammalian mus-
cle. This provides a reserve of high-energy phospho
groups and keeps the adenylate system buffered at a
high phosphorylation state ratio
286
(see Eq. 6-67).
The concentration of both ATP and creatine phos-
phate as well as their rates of interconversion can be
monitored by
31
P NMR within living muscles (Figs.
6-4 and 19-22). Phospho groups were observed to be
B. Muscle
Creatine-P + ADP Creatine + ATP
Figure 19-22 Phosphorus-31 magnetic resonance spectra of
wrist flexor muscles of the forearm of a trained long-distance
runner at rest and during contraction at three different levels
of exercise. Ergometer measurements indicating the percent
of initial maximum strength (% max) were recorded over
each 6-min period. Spectra were obtained during the last 3
min of each period. Times of spectral data collection: A,
resting; B, 46 min; C, 1012 min; and D, 1618 min. The pH
ranged from 6.9 to 7.0. From Park et al.
288
20 10 0 -10
Chemical shift (ppm)
Resting
20%
40%
60%
maximum
effort
P
i
PCr


ATP
A
B
C
D
Ch 19IND page -E- 2/14/03, 11:24 AM 1117
1118 Chapter 19. The Chemistry of Movement
transferred from creatine phosphate to ADP to form
ATP with a flux of 13 mmol kg
1
s
1
in rat legs.
287
The
reverse reaction must occur at about the same rate
because little cleavage of ATP to P
i
was observed in the
anaesthetized rats. Use of surface coils has permitted
direct observation of the operation of this shuttle
system in human muscle (Fig. 19-22)
288
as well as in
animal hearts (see Chapter 6). Only a fraction of the
total creatine present within cells participates in the
shuttle, however.
289
C. Motion in Nonmuscle Cells
At one time actin and myosin were thought to be
present only in muscles, but we know now that both
actin proteins of the myosin family are present in all
eukaryotic cells. Ameboid movement, the motion of
cilia and flagella, and movement of materials along
microtubules within cells also depend upon proteins
of this group.
1. Actin-Based Motility
Ameboid movements of protozoa and of cells
from higher organisms, the ruffling movements of cell
membranes, phagocystosis, and the cytoplasmic
streaming characteristic of many plant cells
289a
have
all been traced to actin filaments or actin cables rather
Bind and stabilize Profilin
a,b,c,d,e,f,g,h
monomeric actin ADF/Cofilin
i,j,h,k
Thymosin
f,l
Cap actin filament ends CapZ
m,n
barbed end Fragmin
o,p
pointed end -Actin
q
Tropomodulin
r
Arp2/3, a complex
of seven polypeptides
Sever or dissociate Gelsolin
s,t,u,v,w
actin filament Depactin
Profilin
d,e
ADF/cofilin
h,k,x,y
TABLE 19-1
Some Actin-Binding Proteins
Function Name
a
Aderem, A. (1992) Trends Biochem. Sci. 17, 438 443
b
Mannherz, H. G. (1992) J. Biol. Chem. 267, 11661 11664
c
Way, M., and Weeds, A. (1990) Nature (London) 344, 292 293
d
Eads, J. C., Mahoney, N. M., Vorobiev, S., Bresnick, A. R., Wen, K.-K.,
Rubenstein, P. A., Haarer, B. K., and Almo, S. C. (1998) Biochemistry 37,
11171 11181
e
Vinson, V. K., De La Cruz, E. M., Higgs, H. N., and Pollard, T. D. (1998)
Biochemistry 37, 10871 10880
f
Kang, F., Purich, D. L., and Southwick, F. S. (1999) J. Biol. Chem. 274, 36963
36972
g
Gutsche-Perelroizen, I., Lepault, J., Ott, A., and Carlier, M.-F. (1999) J. Biol.
Chem. 274, 6234 6243
h
Nodelman, I. M., Bowman, G. D., Lindberg, U., and Schutt, C. E. (1999) J.
Mol. Biol. 294, 1271 1285
i
Lappalainen, P., Fedorov, E. V., Fedorov, A. A., Almo, S. C., and Drubin, D.
G. (1997) EMBO J. 16, 5520 5530
j
Rosenblatt, J., and Mitchison, T. J. (1998) Nature (London) 393, 739 740
k
Chen, H., Bernstein, B. W., and Bamburg, J. R. (2000) Trends Biochem. Sci. 25,
19 23
l
Carlier, M.-F., Didry, D., Erk, I., Lepault, J., Van Troys, M. L.,
Vandekerckhove, J., Perelroizen, I., Yin, H., Doi, Y., and Pantaloni, D. (1996)
J. Biol. Chem. 271, 9231 9239
m
Barron-Casella, E. A., Torres, M. A., Scherer, S. W., Heng, H. H. Q., Tsui, L.-
C., and Casella, J. F. (1995) J. Biol. Chem. 270, 21472 21479
n
Kuhlman, P. A., and Fowler, V. M. (1997) Biochemistry 36, 13461 13472
o
Steinbacher, S., Hof, P., Eichinger, L., Schleicher, M., Gettemans, J.,
Vandekerckhove, J., Huber, R., and Benz, J. (1999) EMBO J. 18, 2923 2929
p
Khaitlina, S., and Hinssen, H. (1997) Biophys. J. 73, 929 937
q
See main text
r
Gregorio, C. C., Weber, A., Bondad, M., Pennise, C. R., and Fowler, V. M.
(1995) Nature (London) 377, 83 86
s
Azuma, T., Witke, W., Stossel, T. P., Hartwig, J. H., and Kwiatkowski, D. J.
(1998) EMBO J. 17, 1362 1370
t
De Corte, V., Demol, H., Goethals, M., Van Damme, J., Gettemans, J., and
Vandekerckhove, J. (1999) Protein Sci. 8, 234 241
u
McGough, A., Chiu, W., and Way, M. (1998) Biophys. J. 74, 764 772
v
Sun, H. Q., Yamamoto, M., Mejillano, M., and Yin, H. L. (1999) J. Biol. Chem.
274, 33179 33182
w
Robinson, R. C., Mejillano, M., Le, V. P., Burtnick, L. D., Yin, H. L., and
Choe, S. (1999) Science 286, 1939 1942
x
Carlier, M.-F., Ressad, F., and Pantaloni, D. (1999) J. Biol. Chem. 274, 33827
33830
y
McGough, A., and Chiu, W. (1999) J. Mol. Biol. 291, 513 519
z
Markus, M. A., Matsudaira, P., and Wagner, G. (1997) Protein Sci. 6, 1197
1209
aa
Vardar, D., Buckley, D. A., Frank, B. S., and McKnight, C. J. (1999) J. Mol.
Biol. 294, 1299 1310
bb
Matsudaira, P. (1991) Trends Biochem. Sci. 16, 87 92
cc
Ono, S., Yamakita, Y., Yamashiro, S., Matsudaira, P. T., Gnarra, J. R.,
Obinata, T., and Matsumura, F. (1997) J. Biol. Chem. 272, 2527 2533
dd
McLachlan, A. D., Stewart, M., Hynes, R. O., and Rees, D. J. G. (1994) J.
Mol. Biol. 235, 1278 1290
ee
Tsukita, S., Yonemura, S., and Tsukita, S. (1997) Trends Biochem. Sci. 22, 53
58
ff
Tsukita, S., and Yonemura, S. (1999) J. Biol. Chem. 274, 34507 34510
Crosslink actin filaments
or monomers
Tight bundles Villin
c,z,aa
Loose bundles -Actinin
bb
Spectrin
bb
Fascin
cc
MARCKS
a
Network Filamin
bb,c
Gelactins
Bind actin filaments Talin
dd
to membrane ERM proteins
ee,ff
Function Name
Ch 19IND page -E- 2/14/03, 11:24 AM 1118
1119
than to microtubules.
289b
Actin is one of the most
abundant proteins in all eukaryotes. Its network of
filaments is especially dense in the lamellipodia of cell
edges, in microvilli, and in the specialized stereocilia
and acrosomal processes (see also pp. 369-370).
289c
Actin filaments and cables are often formed rapidly
and dissolve quickly. When actin filaments grow, the
monomeric subunits with bound ATP are added most
rapidly at the barbed end and dissociate from the
filament at the pointed end (see Section B,2).
94,290
The rate of growth may be ~ 20 200 nm / s, which
requires the addition of 10 100 subunits / s.
291
Various
actin-binding proteins control the growth and stability
of the filaments. The actin-related proteins Arp2 and
Arp3, as a complex Arp2/3, together with recently
recognized formins.
291a
, provide nuclei for rapid
growth of new actin filaments as branches near the
barbed ends.
290,292293c
Growth of the barbed ends of actin filaments is
stimulated by phosphoinositides and by members of
the Rho family of G proteins (p. 559)
293d
through inter-
action with proteins of the WASp group.
293b,d,e
The
name WAS comes from the immune deficiency disorder
WiskottAldrich Syndrome. Yet another family, the
Ena/VASP proteins, are also implicated in actin dy-
namics. They tend to localize at focal adhesions and
edges of lamellipodia.
293e,f
Profilin (Table 19-1) stabiliz-
es a pool of monomeric actin when the barbed ends of
actin filaments are capped. However, it catalyzes both
the addition of actin monomers to uncapped barbed
ends and rapid dissociation of subunits from pointed
ends, leading to increased treadmilling.
294,295
Actin-
severing proteins such as the actin depolymerizing
factor (ADF or cofilin, Table 19-1) promote break-
down of the filaments.
296297a
Treadmilling in the
actin filaments of the lamellipods of crawling cells or
pseudopods of amebas provides a motive force for
many cells
291,298299
ranging from those of Dictyostelium
to human leukocytes. A series of proteins known as
the ezrin, radixin, moesin (ERM) group attach actin
to integral membrane proteins (Fig. 8-17)
292,300,301
and
may interact directly with membrane lipids.
301a,b
Bound ATP in the actin subunits is essential for poly-
merization, and excess ATP together with crosslinking
proteins stabilize the filaments. However, when the
bound ATP near the pointed ends is hydrolyzed to
ADP the filaments become unstable and treadmilling
is enhanced. Thus, as in skeletal muscle, ATP provides
the energy for movement.
Bacteria also contain filamentous proteins that
resemble F-actin and which may be utilized for cell-
shape determination.
301c
Actin-based motility is used
by some bacteria and other pathogens during invasion
of host cells (Box 19-C). It is employed by sperm cells
of Ascaris and of C. elegans, which crawl by an ameboid
movement that utilizes treadmilling of filaments
formed from a motile sperm protein, which does not
resemble actin.
302,302a
Cells are propelled on a glass
surface at rates up to ~1 m / s.
Various nonmuscle forms of myosin also interact
with actin without formation of the myofibrils of
muscle.
299
In most higher organisms nonmuscle myo-
sins often consist of two ~ 200-kDa subunits plus two
pairs of light chains of ~ 17 and 24 kDa each. These
may form bipolar aggregates, which may bind to pairs
of actin filaments to cause relative movement of two
parts of a cell.
303
Movement depending upon the
cytoskeleton is complicated by the presence of a bewil-
dering array of actin-binding proteins, some of which
are listed in Table 19-1.
2. Transport along Microtubules by Kinesin
and Dynein
Many materials are carried out from the cell bodies
of neurons along microtubules in the axons, which in
the human body may be as long as 1 m. The rates of
this fast axonal transport in neurons may be as high
as 5 m / s or 0.43 m / day. The system depends upon
ATP and kinesin (Fig. 19-17) and permits small vesi-
cles to be moved along single microtubules.
304305b
Movement is from the minus end toward the plus end
of the microtubule as defined in Figs. 7-33, 7-34. Slow-
er retrograde axonal transport carries vesicles from
the synapses at the ends of the axons (Fig. 30-8) back
toward the cell body. This retrograde transport depends
upon the complex motor molecule cytoplasmic dynein
which moves materials from the plus end of the micro-
tubule toward the minus end.
305,305c
In addition to
these movements, as mentioned in Chapter 7, micro-
tubules often grow in length rapidly or dissociate into
their tubulin subunits. Growth occurs at one end by
addition of tubulin subunits with their bound GTP.
The fast growing plus-ends of the microtubules are
usually oriented toward the cell periphery, while the
minus-ends are embedded in the centrosome or
microtubule-organizing center (p. 372).
306
Just as
with actin, in which bound ATP is hydrolyzed to ADP,
the bound GTP in the -tubulin subunits of microtubules
is hydrolyzed to GDP
307 310
decreasing the stability of
the microtubules, a phenomenon described as dynamic
instability. Various microtubule-associated proteins
(MAPs) have strong effects on this phenomenon.
311
The MAPs are often regulated by phosphorylation
dephosphorylation cycles involving serine / threonine
kinases. Microtubules also undergo posttranslational
alterations not seen in other proteins. These include
addition or removal of tyrosine at the C terminus.
312
Polyglycyl groups containing 3 34 glycine residues
may be bound covalently to -carboxyl groups of
glutamate side chains in both - and -tubulins.
312,313
This stabilizes the microtubules and is important to the
long-lived microtubules of the axonomes of flagella and
C. Motion in Nonmuscle Cells
Ch 19IND page -E- 2/14/03, 11:24 AM 1119
1120 Chapter 19. The Chemistry of Movement
BOX 19-C ACTIN-BASED MOTILITY AND BACTERIAL INVASION
(vasodilator-stimulated phosphoprotein). The pro-
line-rich region of the bacterial ActA may bind to
VASP to initiate polymerization.
g
Both profilin
(Table 19-1) and the crosslinking protein -actinin
also stimulate comet tail growth. Myosin does not
participate in actin-based motility, but the hydrolysis
of ATP drives the process through its link to actin
polymerization.
i
Although Listeria has been studied most, actin-
based motility is employed by other pathogens as
well, e.g., Shigella flexneri (the dysentery bacterium),
k
Rickettsia,
l
and vaccinia virus.
l
Although entero-
pathogenic E. coli do not use this method of move-
ment, they induce accumulation of actin beneath the
bacteria. They also promote formation of actin-rich
adherent pseudopods and highly organized cyto-
skeletal structures that presumably assist the bacteria
in entering a cell.
m
a
Cossart, P., and Lecuit, M. (1998) EMBO J. 17, 37973806
b
Sechi, A. S., Wehland, J., and Small, J. V. (1997) J. Cell Biol. 137,
155167
c
Pantaloni, D., Le Clainche, C., and Carlier, M.-F. (2001) Science
292, 15021506
d
Mogilner, A., and Oster, G. (1996) Biophys. J. 71, 30303045
e
Mourrain, P., Lasa, I., Gautreau, A., Gouin, E., Pugsley, A., and
Cossart, P. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 1003410039
f
Southwick, F. S., and Purich, D. L. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 51685172
g
Niebuhr, K., Ebel, F., Frank, R., Reinhard, M., Domann, E., Carl,
U. D., Walter, U., Gertler, F. B., Wehland, J., and Chakraborty, T.
(1997) EMBO J. 16, 54335444
h
Welch, M. D., Rosenblatt, J., Skoble, J., Portnoy, D. A., and
Mitchison, T. J. (1998) Science 281, 105108
i
Loisel, T. P., Boujemaa, R., Pantaloni, D., and Carlier, M.-F.
(1999) Nature (London) 401, 613616
j
Machesky, L. M., and Cooper, J. A. (1999) Nature (London) 401,
542543
k
Bourdet-Sicard, R., Rdiger, M., Jockusch, B. M., Gounon, P.,
Sansonetti, P. J., and Tran Van Nhieu, G. (1999) EMBO J. 18,
58535862
l
Cudmore, S., Cossart, P., Griffiths, G., and Way, M. (1995)
Nature (London) 378, 636638
m
Rosenshine, I., Ruschkowski, S., Stein, M., Reinscheid, D. J.,
Mills, S. D., and Finlay, B. B. (1996) EMBO J. 15, 26132624
Listeria monocytogenes is a dangerous food-borne
bacterium that has become a major problem in the
United States. This is one of the best understood
intracellular pathogens. It is able to enter cells, escape
from phagocytic vesicles, spread from cell to cell, and
cross intestinal, bloodbrain, and placental barriers.
a c
Within cells these bacteria move using actin-based
motility. Actin subunits polymerize at one end of
a bacterium leaving a comet tail of crosslinked
fibrous actin behind (see micrographs). Actin poly-
merization occurs directly behind the bacteria with
subunits of monomeric actin adding to the fast
growing barbed end (see Section B,2) of the actin
strands. Growth has been described as a Brownian
ratchet.
c,d
Continual Brownian movement opens
up spaces behind the bacteria, spaces that are imme-
diately filled by new actin subunits. This provides a
propulsive force adequate to move the bacteria ahead
at velocities of about 0.3 m / s.
Polymerization of actin is induced by interaction
of a dimer of a 610-residue bacterial protein ActA
with proteins of the host cell.
a,e h
ActA is a composite
protein with an N-terminal region that protrudes
from the bacterial cell, a central region of proline-rich
repeats that appear to be essential for recognition by
host cells, and a C-terminal hydrophobic membrane
anchor. There are also regions of close sequence
similarity to the human actin-binding proteins vincu-
lin and zyxin. The number of host proteins needed
in addition to monomeric actin are:
i,j
the two actin-
related proteins, Arp2 and Arp3, which stimulate
actin polymerization and branching;
h
ADF/ cofilin,
which increases the rates, both of growth at the barbed
ends and dissociation from the pointed ends of the
filaments; and Cap Z, which caps barbed ends (Table
19-1). The need for ADF/ cofilin and Cap Z seems
paradoxical. Cap Z may cap mostly older and
slower growing filaments, restricting rapid filament
assembly to the region closest to the bacterium. The
need for ADF/ cofilin is unclear.
i
Growth rates are
also enhanced by the human protein called VASP
(A) Listeria cell with comet tail of cross-linked actin filaments. From
Kocks et al. (1992) Cell 68, 521531. Courtesy of Pascale Cossart.
(B) Enlarged section of a thin comet tail of high resolution showing
the actin filaments. From Sechi et al.
b
Courtesy of Antonio Sechi.
A
B
0.2 m
Ch 19IND page -E- 2/14/03, 11:24 AM 1120
1121
cilia. Polyglutamyl groups of 6 7 glutamates are also
often added.
314
Both dynein and several kinesins act as motors for
formation of the spindle and for movement of chromo-
somes toward the minus ends of spindle microtubules
during mitosis and meiosis (Fig. 26-11).
314a
In the
genome of Saccharomyces cerevisiae there is only one
dynein gene, but genes for six different kinesin-type
motor molecules are present.
315
In higher organisms
there may be even more genes for kinesins but there
is apparently only one dynein in most species.
316
Axonemal dyneins drive the motion of eukaryotic
flagella and cilia. As with the cytoplasmic dyneins a
complete molecule consists of two or three heavy chains
with molecular mass ~520 kDa, some localized in the
dynein tail, and several lighter chains.
305a,317 321
Like
myosin dynein is an ATPase.
3. Eukaryotic Cilia and Flagella
The motion of eukaryotic flagella (Fig. 1-8) involves
a sliding of the microtubular filaments somewhat
analogous to the sliding of muscle filaments.
305,322 325
Figure 19-23 (A) Diagram of a cross-sectional view of the
outer portion of a lamellibranch gill cilium. This has the
9+2 axoneme structure as shown in Fig. 1-8 and in (B). The
viewing direction is from base to tip. From M. A. Sleigh.
329
(B, C) Thin-section electron micrographs of transverse (B)
and longitudinal (C) sections of wild-type Chlamydomonas
axonemes. In transverse section labels A and B mark A and
B subtubules of microtubule doublets; oa, ia, outer and inner
dynein arms, respectively; sp, spokes; cpp, central pair
projections; bk, beaks. From Smith and Sale.
329a
Sliding between the outer doublet microtubules (Fig.
19-23) via their inner and outer arms (dynein com-
pounds) is thought to provide the characteristic bend-
ing waves.
325a,b
The movement is powered by dynein
and ATP hydrolysis. Force and displacement measure-
ments made by optical trapping nanometry suggest
that the characteristic rhythmic beating of flagella
results from an oscillatory property of the dynein.
326
The extremely complex structure of flagella is illustrated
in Fig. 19-23. About 250 individual axonemal proteins
have been detected in flagella of the alga Chlamydo-
monas (Fig. 1-11),
327
and a large number of mutants
with various defects in their flagella have been isolated.
The radial spokes (Fig. 19-23) alone contain 17 different
proteins. These spokes protrude at ~ 29-nm intervals
while the dynein molecules lie between pairs of the
outer microtubule doublets at ~ 24-nm intervals. The
dynein arms protrude from the A microtubule of
each outer doublet and make contact with the incom-
plete B microtubule of the next doublet (Fig. 19-12).
Although the shapes of the molecules are quite differ-
ent, the basic chemistry of the ATPase activity of the
dyneinmicrotubule system resembles that of actomy-
osin. However, the complexity of the dynein arms,
328
A B
C
C. Motion in Nonmuscle Cells
Ch 19IND page -E- 2/14/03, 11:24 AM 1121
1122 Chapter 19. The Chemistry of Movement
1. Elston, T. C., and Oster, G. (1997) Biophys. J.
73, 703721
2. Magariyama, Y., Sugiyama, S., Muramoto, K.,
Maekawa, Y., Kawagishi, I., Imae, Y., and
Kudo, S. (1994) Nature (London) 371, 752
3. Macnab, R. M. (1987) in Escherichia coli and
Salmonella typhimurium (Niedhardt, F. C., ed),
pp. 7083, Am. Soc. for Microbiology,
Washington, D.C.
4. Berg, H. C. (1975) Sci. Am. 233(Aug), 3644
5. Shimada, K., Kamiya, R., and Asakura, S.
(1975) Nature (London) 254, 332334
which exist in two types, inner and outer, suggests a
complex contraction cycle.
4. Chemotaxis
As described in Box 11-C, the ameboid cells of the
slime mold Dictyostelium discoideum are attracted to
nutrients such as folic acid during their growth stage.
Later, as the cells undergo developmental changes
they become attracted by pulses of cyclic AMP.
330
Occupancy of 7-helix receptors for cAMP on the outer
plasma membrane appears to induce methylation of
both proteins and phospholipids and a rise in cyto-
solic Ca
2+
and changes in the cytoskeleton that result
in preferential extension of the actin-rich pseudopods
toward the chemoattractant.
331
In a similar manner, human ameboid leukocytes
are attracted to sites of inflammation by various
chemotactic factors.
332
These include the 74-residue
cleavage product C5a formed from the fifth component
of complement (Chapter 31),
333
various lymphokines
(Chapter 31) secreted by lymphocytes, and peptides
such as VGSE and AGSE, as well as larger peptides
released by mast cells, basophils, or stimulated mono-
cytes
334
and oxoicosenoids.
335
Polymorphonuclear
leukocytes, upon engulfing sodium urate crystals in
gouty joints, release an 8.4-kDa chemotactic protein
which may cause a damaging response in this arthritic
condition. Leukotriene B is a potent chemotactic agent
as are a series of specific bacterial products, formylated
peptides such as N-formyl-MLF.
332,336
Neutrophils, monocytes, macrophages, eosinophils,
basophils, and polymorphonuclear leukocytes are all
affected by several or all of these factors. Binding to
specific receptors results in a variety of changes in the
cells. These include alterations in membrane potential,
cyclic nucleotide levels, and ion fluxes (Na
+
, K
+
, Ca
2+
)
as well as increased methylation of specific proteins.
A reorganization of microtubules and actin fibrils
occurs, probably in response to an altered gradient of
Ca
2+
. The morphology of the cells changes, and they
begin immediately to crawl toward the chemoattractants.
It appears that these ameboid cells detect a gradient of
attractant concentration between one end of the cell
and the other, even though the anticipated difference
may amount to only 0.1% of the total.
337,338
5. Other Forms of Movement
Movement is characteristic of life and is caused not
only by motor proteins but by various springs and
ratchets which may be energized in a number of ways.
339
A striking example, which any one with a microscope
and some fresh pond water can observe, is contraction
of the stalk of protozoa of the genus Vorticella. Appar-
ently first reported in 1676 by Leeuwenhoek the organ-
isms 2 3 mm-long stalk contracts into a coiled spring
(see p. 1 and also p. 281) when the animal is disturbed.
Application of calcium ions causes contraction within
a few milliseconds to ~ 40% of the original length. The
process reverses slowly after a few seconds. Contrac-
tion is caused by a spring-like organ the spasmoneme,
which is a bundle of short 2 nm-diameter fibrils inside
the stalk. The fibrils are thought to be weakly cross-
linked and held in the extended state by electrostatic
repulsion between the negatively charged rods. Addi-
tion of Ca
2+
neutralizes the charges permitting an
entropy-driven collapse of the fibers.
339
Another remarkable example is extension of the
acrosomal process from a sperm cell of the horseshoe
crab Limulus polyphemus at fertilization. A bundle of
actin filaments in a crystalline state lies coiled around
the base of the nucleus. At fertilization the bundle
uncoils and slides through a tunnel in the nucleus to
form a 60 m-long acrosomal process within a few
seconds. The uncoiled bundle is also crystalline. The
coiled bundle is apparently overtwisted and an actin
crosslinking protein scruin mediates the conforma-
tional alteration that takes place.
339
A somewhat related
process may be involved in contraction of bacteriophage
tails (pp. 363, 364)
Some bacteria glide with a twitching movement
induced by rapid retraction of pili.
340
Another type
of movement involves the pinching off of vesicles,
e.g., of clathrin-coated pits (Fig. 8-27). This is a GTP-
driven process that requires a mechanoenzyme called
dynamin.
341,342
References
6. Berg, H. C. (1975) Nature (London) 254, 389392
7. Macnab, R. M. (1985) Trends Biochem. Sci. 10,
185188
8. Macnab, R. M., and Aizawa, S.-J. (1984) Annu
Rev Biophys Bioeng. 13, 5183
9. Sharp, L. L., Zhou, J., and Blair, D. F. (1995)
Biochemistry 34, 91669171
10. Morgan, D. G., Owen, C., Melanson, L. A., and
DeRosier, D. J. (1995) J. Mol. Biol. 249, 88110
11. Mimori, Y., Yamashita, I., Murata, K.,
Fujiyoshi, Y., Yonekura, K., Toyoshima, C.,
and Namba, K. (1995) J. Mol. Biol. 249, 6987
12. Yamashita, I., Vonderviszt, F., Mimori, Y.,
Suzuki, H., Oosawa, K., and Namba, K. (1995)
J. Mol. Biol. 253, 547558
12a. Samatey, F. A., Imada, K., Nagashima, S.,
Vonderviszt, F., Kumasaka, T., Yamamoto, M.,
and Namba, K. (2001) Nature (London) 410,
331337
13. Muramoto, K., Kawagishi, I., Kudo, S.,
Magariyama, Y., Imae, Y., and Homma, M.
(1995) J. Mol. Biol. 251, 5058
14. DePamphilis, M. L., and Adler, J. (1971) J.
Bacteriol. 105, 396407
Ch 19IND page -E- 2/14/03, 11:24 AM 1122
1123
15. Stallmeyer, M. J. B., Aizawa, S.-I., Macnab, R.
M., and DeRosier, D. J. (1989) J. Mol. Biol. 205,
519528
16. DeRosier, D. J. (1998) Cell 93, 17 20
17. Trachtenberg, S., and DeRosier, D. J. (1992) J.
Mol. Biol. 226, 447454
18. Trachtenberg, S., DeRosier, D. J., Zemlin, F.,
and Beckmann, E. (1998) J. Mol. Biol. 276, 759
773
19. Hasegawa, K., Yamashita, I., and Namba, K.
(1998) Biophys. J. 74, 569575
19a. Macnab, R. M. (2001) Nature (London) 410,
321322
20. Iino, T. (1969) Bacteriol. Rev. 33, 454475
21. Kubori, T., Shimamoto, N., Yamaguchi, S.,
Namba, K., and Aizawa, S.-I. (1992) J. Mol.
Biol. 226, 433446
22. Vonderviszt, F., Zvodszky, P., Ishimura, M.,
Uedaira, H., and Namba, K. (1995) J. Mol. Biol.
251, 520532
23. Muramoto, K., Makishima, S., Aizawa, S.-I.,
and Macnab, R. M. (1998) J. Mol. Biol. 277,
871882
24. Ikeda, T., Oosawa, K., and Hotani, H. (1996) J.
Mol. Biol. 259, 679686
25. Fahrner, K. A., Block, S. M., Krishnaswamy,
S., Parkinson, J. S., and Berg, H. C. (1994) J.
Mol. Biol. 238, 173186
25a. Hirano, T., Minamino, T., and Macnab, R. M.
(2001) J. Mol. Biol. 312, 359369
26. Maki, S., Vonderviszt, F., Furukawa, Y.,
Imada, K., and Namba, K. (1998) J. Mol. Biol.
277, 771777
26a. Yonekura, K., Maki, S., Morgan, D. G.,
DeRosier, D. J., Vonderviszt, F., Imada, K., and
Namba, K. (2000) Science 290, 21482152
26b. Macnab, R. M. (2000) Science 290, 20862087
26c. Minamino, T., Tame, J. R. H., Namba, K., and
Macnab, R. M. (2001) J. Mol. Biol. 312, 1027
1036
26d. Auvray, F., Thomas, J., Fraser, G. M., and
Hughes, C. (2001) J. Mol. Biol. 308, 221229
27. Berg, H. C. (1996) Proc. Natl. Acad. Sci. U.S.A.
93, 1422514228
28. Lloyd, S. A., Whitby, F. G., Blair, D. F., and
Hill, C. P. (1999) Nature (London) 400, 472475
29. Schuster, S. C., and Khan, S. (1994) Annu. Rev.
Biophys. Biomol. Struct. 23, 509539
30. Adler, J. (1976) Sci. Am. 234 (Apr), 4047
31. Calladine, C. R. (1975) Nature (London) 255,
121124
32. Eisenbach, M., and Adler, J. (1981) J. Biol.
Chem. 256, 88078814
33. Fung, D. C., and Berg, H. C. (1995) Nature
(London) 375, 809812
34. Glagolev, A. N., and Skulachev, V. P. (1978)
Nature (London) 273, 280282
35. Chun, S. Y., and Parkinson, J. S. (1988) Science
239, 276278
36. Lager, P. (1990) Commun. Theor. Biol. 2, 99
123
37. Tang, H., Braun, T. F., and Blair, D. F. (1996) J.
Mol. Biol. 261, 209221
37a. Ko, M., and Park, C. (2000) J. Mol. Biol. 303,
371382
38. Block, S. M., and Berg, H. C. (1984) Nature
(London) 309, 470472
39. Ueno, T., Oosawa, K., and Aizawa, S.-I. (1994)
J. Mol. Biol. 236, 546555
40. Zhou, J., Fazzio, R. T., and Blair, D. F. (1995) J.
Mol. Biol. 251, 237242
41. Garza, A. G., Biran, R., Wohlschlegel, J. A.,
and Manson, M. D. (1996) J. Mol. Biol. 258,
270285
41a. Van Way, S. M., Hosking, E. R., Braun, T. F.,
and Manson, M. D. (2000) J. Mol. Biol. 297,
724
42. Zhao, R., Pathak, N., Jaffe, H., Reese, T. S.,
and Khan, S. (1996) J. Mol. Biol. 261, 195208
43. Marykwas, D. L., Schmidt, S. A., and Berg, H.
C. (1996) J. Mol. Biol. 256, 564576
44. Zhou, J., Lloyd, S. A., and Blair, D. F. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 64366441
44a. Thomas, D. R., Morgan, D. G., and DeRosier,
D. J. (1999) Proc. Natl. Acad. Sci. U.S.A. 96,
1013410139
44b. Lux, R., Kar, N., and Khan, S. (2000) J. Mol.
Biol. 298, 577583
45. Toker, A. S., and Macnab, R. M. (1997) J. Mol.
Biol. 273, 623634
46. Kehry, M. R., Doak, T. G., and Dahlquist, F. W.
(1984) J. Biol. Chem. 259, 1182811835
47. Koshland, D. E., Jr. (1988) Biochemistry 27,
58295834
48. Stock, J., and Stock, A. (1987) Trends Biochem.
Sci. 12, 371375
49. Segall, J. E., Block, S. M., and Berg, H. C.
(1986) Proc. Natl. Acad. Sci. U.S.A. 83, 8987
8991
50. Jeffery, C. J., and Koshland, D. E., Jr. (1993)
Protein Sci. 2, 559566
51. Li, J., Li, G., and Weis, R. M. (1997) Biochemis-
try 36, 1185111857
51a. Isaac, B., Gallagher, G. J., Balazs, Y. S., and
Thompson, L. K. (2002) Biochemistry 41, 3025
3036
52. Scott, W. G., Milligan, D. L., Milburn, M. V.,
Priv, G. G., Yeh, J., Koshland, D. E., Jr., and
Kim, S.-H. (1993) J. Mol. Biol. 232, 555573
53. Yeh, J. I., Biemann, H.-P., Priv, G. G., Pandit,
J., Koshland, D. E., Jr., and Kim, S.-H. (1996) J.
Mol. Biol. 262, 186201
54. Foster, D. L., Mowbray, S. L., Jap, B. K., and
Koshland, D. E., Jr. (1985) J. Biol. Chem. 260,
1170611710
54a. Falke, J. J., and Hazelbauer, G. L. (2001) Trends
Biochem. Sci. 26, 257265
55. Maddock, J. R., and Shapiro, L. (1993) Science
259, 17171723
56. Wolfe, A. J., Conley, M. P., and Berg, H. C.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 6711
6715
57. Ninfa, E. G., Stock, A., Mowbray, S., and
Stock, J. (1991) J. Biol. Chem. 266, 97649770
58. Kim, S.-H. (1994) Protein Sci. 3, 159165
59. Chervitz, S. A., and Falke, J. J. (1995) J. Biol.
Chem. 270, 2404324053
60. Ottemann, K. M., Thorgeirsson, T. E.,
Kolodziej, A. F., Shin, Y.-K., and Koshland, D.
E., Jr. (1998) Biochemistry 37, 70627069
61. Trammell, M. A., and Falke, J. J. (1999)
Biochemistry 38, 329336
61a. Hirschman, A., Boukhvalova, M.,
VanBruggen, R., Wolfe, A. J., and Stewart, R.
C. (2001) Biochemistry 40, 1387613887
62. Spiro, P. A., Parkinson, J. S., and Othmer, H.
G. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 7263
7268
63. Volz, K. (1993) Biochemistry 32, 1174111753
64. Bellsolell, L., Cronet, P., Majolero, M., Serrano,
L., and Coll, M. (1996) J. Mol. Biol. 257, 116
128
65. Jiang, M., Bourret, R. B., Simon, M. I., and
Volz, K. (1997) J. Biol. Chem. 272, 1185011855
65a. Halkides, C. J., McEvoy, M. M., Casper, E.,
Matsumura, P., Volz, K., and Dahlquist, F. W.
(2000) Biochemistry 39, 52805286
65b. Lee, S.-Y., Cho, H. S., Pelton, J. G., Yan, D.,
Berry, E. A., and Wemmer, D. E. (2001) J. Biol.
Chem. 276, 1642516431
65c. Cho, H. S., Lee, S.-Y., Yan, D., Pan, X.,
Parkinson, J. S., Kustu, S., Wemmer, D. E., and
Pelton, J. G. (2000) J. Mol. Biol. 297, 543551
65d. Kim, C., Jackson, M., Lux, R., and Khan, S.
(2001) J. Mol. Biol. 307, 119135
66. Bren, A., and Eisenbach, M. (1998) J. Mol. Biol.
278, 507514
References
66a. Bren, A., and Eisenbach, M. (2001) J. Mol. Biol.
312, 699709
67. Sanna, M. G., and Simon, M. I. (1996) J. Biol.
Chem. 271, 73577361
68. Blat, Y., and Eisenbach, M. (1996) J. Biol. Chem.
271, 12261231
69. Silversmith, R. E., Appleby, J. L., and Bourret,
R. B. (1997) Biochemistry 36, 1496514974
70. Weis, R. M., Chasalow, S., and Koshland, D.
E., Jr. (1990) J. Biol. Chem. 265, 68176826
71. Wu, J., Li, J., Li, G., Long, D. G., and Weis, R.
M. (1996) Biochemistry 35, 49844993
72. West, A. H., Martinez-Hackert, E., and Stock,
A. M. (1995) J. Mol. Biol. 250, 276290
73. Alon, U., Surette, M. G., Barkai, N., and
Leibler, S. (1999) Nature (London) 397, 168171
73a. Bray, D. (2002) Proc. Natl. Acad. Sci. U.S.A. 99,
79
74. Prasad, K., Caplan, S. R., and Eisenbach, M.
(1998) J. Mol. Biol. 280, 821828
75. Nickitenko, A. V., Trakhanov, S., and Quiocho,
F. A. (1995) Biochemistry 34, 1658516595
76. Lux, R., Jahreis, K., Bettenbrock, K., Parkin-
son, J. S., and Lengeler, J. W. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 1158311587
77. Gebert, J. F., Overhoff, B., Manson, M. D., and
Bods, W. (1988) J. Biol. Chem. 263, 16652
16660
77a. Nishiyama, S.-i, Maruyama, I. N., Homma,
M., and Kawagishi, I. (1999) J. Mol. Biol. 286,
12751284
77b. Kim, S.-H., Wang, W., and Kim, K. K. (2002)
Proc. Natl. Acad. Sci. U.S.A. 99, 1161111615
78. Unsworth, B. R., Witzmann, F. A., and Fitts, R.
H. (1982) J. Biol. Chem. 257, 1512915136
79. Kelly, A. M., and Rubinstein, N. A. (1980)
Nature (London) 288, 267269
80. Bullard, B. (1983) Trends Biochem. Sci. 8, 6870
81. Cohen, C. (1982) Proc. Natl. Acad. Sci. U.S.A.
79, 31763178
82. Huxley, H., and Hanson, J. (1954) Nature
(London) 173, 973976
83. Huxley, H. E. (1958) Sci. Am. 199, 6782
84. Huxley, H. E. (1990) J. Biol. Chem. 265, 8347
8350
85. Glickson, J. D., Phillips, W. D., and Rupley, J.
A. (1971) J. Am. Chem. Soc. 93, 40314038
86. Yates, L. D., and Greaser, M. L. (1983) J. Mol.
Biol. 168, 123141
87. Yao, X., Grade, S., Wriggers, W., and Ruben-
stein, P. A. (1999) J. Biol. Chem. 274, 37443
37449
88. Rubenstein, P. A. (1990) BioEssays 12, 309315
89. Allen, P. G., Shuster, C. B., Ks, J., Chaponnier,
C., Janmey, P. A., and Herman, I. M. (1996)
Biochemistry 35, 1406214069
90. Kabsch, W., and Holmes, K. C. (1995) FASEB
J. 9, 167174
91. Kabsch, W., Mannherz, H. G., Suck, D., Pai, E.
F., and Holmes, K. C. (1990) Nature (London)
347, 3744
92. Chik, J. K., Lindberg, U., and Schutt, C. E.
(1996) J. Mol. Biol. 263, 607623
92a. Otterbein, L. R., Graceffa, P., and Dominguez,
R. (2001) Science 293, 708711
93. Pollard, T. D., and Craig, S. W. (1982) Trends
Biochem. Sci. 7, 5558
94. Teubner, A., and Wegner, A. (1998) Biochemis-
try 37, 75327538
94a. Orlova, A., Galkin, V. E., VanLoock, M. S.,
Kim, E., Shvetsov, A., Reisler, E., and
Egelman, E. H. (2001) J. Mol. Biol. 312, 95106
95. Carlier, M.-F., and Pantaloni, D. (1988) J. Biol.
Chem. 263, 817825
95a. Sablin, E. P., Dawson, J. F., VanLoock, M. S.,
Spudich, J. A., Egelman, E. H., and Fletterick,
R. J. (2002) Proc. Natl. Acad. Sci. U.S.A. 99,
1094510947
Ch 19IND page -E- 2/14/03, 11:24 AM 1123
1124 Chapter 19. The Chemistry of Movement
96. Maruta, H., Knoerzer, W., Hinssen, H., and
Isenberg, G. (1984) Nature (London) 312, 424
427
97. Pollard, T. D., and Cooper, J. A. (1986) Ann.
Rev. Biochem. 55, 9871035
98. Maruyama, K. (1997) FASEB J. 11, 341345
98a. Maruyama, K. (2002) Trends Biochem. Sci. 27,
264266
99. Trinick, J. (1994) Trends Biochem. Sci. 19, 405
409
100. Labeit, S., and Kolmerer, B. (1995) Science 270,
293296
100a. Amodeo, P., Fraternali, F., Lesk, A. M., and
Pastore, A. (2001) J. Mol. Biol. 311, 283296
101. Kolmerer, B., Olivieri, N., Witt, C. C.,
Herrmann, B. G., and Labeit, S. (1996) J. Mol.
Biol. 256, 556563
102. Tskhovrebova, L., and Trinick, J. (2001) J. Mol.
Biol. 310, 755771
102a. Ma, K., Kan, L.-s, and Wang, K. (2001)
Biochemistry 40, 34273438
103. Wang, K., Knipfer, M., Huang, Q.-Q., van
Heerden, A., Hsu, L. C.-L., Gutierrez, G.,
Quian, X.-L., and Stedman, H. (1996) J. Biol.
Chem. 271, 43044314
104. Kalverda, A. P., Wymenga, S. S., Lomman, A.,
van de Ven, F. J. M., Hilbers, C. W., and
Canters, G. W. (1994) J. Mol. Biol. 240, 358371
105. Millevoi, S., Trombitas, K., Kolmerer, B.,
Kostin, S., Schaper, J., Pelin, K., Granzier, H.,
and Labeit, S. (1998) J. Mol. Biol. 282, 111123
106. Politou, A. S., Millevoi, S., Gautel, M.,
Kolmerer, B., and Pastore, A. (1998) J. Mol.
Biol. 276, 189202
106a. McElhinny, A. S., Kolmerer, B., Fowler, V. M.,
Labeit, S., and Gregorio, C. C. (2001) J. Biol.
Chem. 276, 583592
107. Obermann, W. M. J., Gautel, M., Weber, K.,
and Frst, D. O. (1997) EMBO J. 16, 211220
108. Young, P., Ferguson, C., Banuelos, S., and
Gautel, M. (1998) EMBO J. 17, 16141624
109. Sorimachi, H., Freiburg, A., Kolmerer, B.,
Ishiura, S., Stier, G., Gregorio, C. C., Labeit,
D., Linke, W. A., Suzuki, K., and Labeit, S.
(1997) J. Mol. Biol. 270, 688695
109a. Tang, J., Taylor, D. W., and Taylor, K. A. (2001)
J. Mol. Biol. 310, 845858
109b. Joseph, C., Stier, G., OBrien, R., Politou, A. S.,
Atkinson, R. A., Bianco, A., Ladbury, J. E.,
Martin, S. R., and Pastore, A. (2001)
Biochemistry 40, 49574965
110. Isobe, Y., Warner, F. D., and Lemanski, L. F.
(1988) Proc. Natl. Acad. Sci. U.S.A. 85, 6758
6762
111. Deatherage, J. F., Cheng, N., and Bullard, B.
(1989) J. Cell Biol. 108, 17751782
112. van Straaten, M., Goulding, D., Kolmerer, B.,
Labeit, S., Clayton, J., Leonard, K., and
Bullard, B. (1999) J. Mol. Biol. 285, 15491562
112a. Kolmerer, B., Clayton, J., Benes, V., Allen, T.,
Ferguson, C., Leonard, K., Weber, U., Knekt,
M., Ansorge, W., Labeit, S., and Bullard, B.
(2000) J. Mol. Biol. 296, 435448
112b. Fukuzawa, A., Shimamura, J., Takemori, S.,
Kanzawa, N., Yamaguchi, M., Sun, P.,
Maruyama, K., and Kimura, S. (2001) EMBO
J. 20, 48264835
113. Yamaguchi, M., Izumimoto, M., Robson, R.
M., and Stromer, M. H. (1985) J. Mol. Biol. 184,
621644
114. Baron, M. D., Davison, M. D., Jones, P., Patel,
B., and Critchley, D. R. (1987) J. Biol. Chem.
262, 25582561
115. Pan, K.-M., Roelke, D. L., and Greaser, M. L.
(1986) J. Biol. Chem. 261, 99229928
116. McLachlan, A. D. (1984) Ann. Rev. Biophys.
Bioeng. 13, 167189
References
116a. Bellin, R. M., Huiatt, T. W., Critchley, D. R.,
and Robson, R. M. (2001) J. Biol. Chem. 276,
3233032337
117. Carragher, B. O., Cheng, N., Wang, Z.-Y.,
Korn, E. D., Reilein, A., Belnap, D. M.,
Hammer, J. A., III, and Steven, A. C. (1998)
Proc. Natl. Acad. Sci. U.S.A. 95, 1520615211
118. Heintzelman, M. B., and Schwartzman, J. D.
(1997) J. Mol. Biol. 271, 139146
119. Baker, J. P., and Titus, M. A. (1997) J. Mol. Biol.
272, 523535
120. Hasson, T., and Mooseker, M. S. (1996) J. Biol.
Chem. 271, 1643116434
121. Jontes, J. D., and Milligan, R. A. (1997) J. Mol.
Biol. 266, 331342
121a. Ajtai, K., Garamszegi, S. P., Park, S., Dones, A.
L. V., and Burghardt, T. P. (2001) Biochemistry
40, 1207812093
121b. Wendt, T., Taylor, D., Trybus, K. M., and
Taylor, K. (2001) Proc. Natl. Acad. Sci. U.S.A.
98, 43614366
121c. Andersen, J. L., Schjerling, P., and Saltin, B.
(2000) Sci. Am. 283(Sep), 4855
122. Weiss, A., Schiaffino, S., and Leinwand, L. A.
(1999) J. Mol. Biol. 290, 6175
123. Robillard, G., and Schulman, R. G. (1972) J.
Mol. Biol. 71, 507511
123a. Swank, D. M., Bartoo, M. L., Knowles, A. F.,
Iliffe, C., Bernstein, S. I., Molloy, J. E., and
Sparrow, J. C. (2001) J. Biol. Chem. 276, 15117
15124
124. Ferrin, T. E., Huang, C. C., Jarvis, L. E., and
Langridge, R. (1988) J. Mol. Graphics 6, 1327
125. Lorenz, M., Poole, K. J. V., Popp, D., Rosen-
baum, G., and Holmes, K. C. (1995) J. Mol.
Biol. 246, 108119
126. Knight, P. J. (1996) J. Mol. Biol. 255, 269274
127. Katoh, T., Konishi, K., and Yazawa, M. (1998)
J. Biol. Chem. 273, 1143611439
128. Offer, G., and Knight, P. (1996) J. Mol. Biol.
256, 407416
129. Davis, J. S. (1988) Ann. Rev. Biophys. Biophys.
Chem. 17, 217239
130. Squire, J. M. (1973) J. Mol. Biol. 77, 291323
131. Padrn, R., Alamo, L., Murgich, J., and Craig,
R. (1998) J. Mol. Biol. 275, 3541
132. Sohn, R. L., Vikstrom, K. L., Strauss, M.,
Cohen, C., Szent-Gyorgyi, A. G., and Lein-
wand, L. A. (1997) J. Mol. Biol. 266, 317330
133. Bennett, P. M., and Gautel, M. (1996) J. Mol.
Biol. 259, 896903
134. Winkler, H., Reedy, M. C., Reedy, M. K.,
Tregear, R., and Taylor, K. A. (1996) J. Mol.
Biol. 264, 302322
135. Crowther, R. A., Padron, R., and Craig, R.
(1985) J. Mol. Biol. 184, 429439
135a. Offer, G., Knight, P. J., Burgess, S. A., Alamo,
L., and Padrn, R. (2000) J. Mol. Biol. 298, 239
260
135b. Flavigny, J., Souchet, M., Sbillon, P., Berrebi-
Bertrand, I., Hainque, B., Mallet, A., Bril, A.,
Schwartz, K., and Carrier, L. (1999) J. Mol.
Biol. 294, 443456
135c. Witt, C. C., Gerull, B., Davies, M. J., Centner,
T., Linke, W. A., and Thierfelder, L. (2001) J.
Biol. Chem. 276, 53535359
136. Standiford, D. M., Davis, M. B., Miedema, K.,
Franzini-Armstrong, C., and Emerson, C. P. J.
(1997) J. Mol. Biol. 265, 4055
137. Vibert, P. and Craig, R. (1983) J. Mol. Biol. 165,
303320
138. Engelhardt, W. A., and Ljubimowa, M. N.
(1939) Nature (London) 144, 668669
139. Szent-Gyorgyi, A. (1941) Studies Inst. Med.
Chem., Univ. Szeged 1, 1726 (reprinted in H.
M. Kalckar ed., Biological Phosphorylations, pp.
465472, Prentice-Hall, Englewood Cliffs,
New Jersey 1969)
140. Szent-Gyorgyi, A. (1947) Chemistry of Muscu-
lar Contraction, Academic Press, New York
141. Straub, F. B. (1969) in Biological Phosphorylation
(Kalckar, H. M., ed), pp. 474483, Prentice-
Hall, Englewood Cliffs, New Jersey (reprinted
from Studies Inst. Med. Chem. Univ. Szeged,
2, 315)
142. Milligan, R. A. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 2126
143. Schmitz, H., Reedy, M. C., Reedy, M. K.,
Tregear, R. T., Winkler, H., and Taylor, K. A.
(1996) J. Mol. Biol. 264, 279301
144. Katayama, E. (1998) J. Mol. Biol. 278, 349367
145. Huxley, H. (1969) Science 114, 13561366
146. Huxley, H. E. (1998) Trends Biochem. Sci. 23,
8487
147. Huxley, A. F., and Simmons, R. M. (1969)
Nature (London) 233, 533538
148. Tsukita, S., and Yano, M. (1985) Nature
(London) 317, 182184
149. Sheetz, M. P., and Spudich, J. A. (1983) Nature
(London) 303, 3135
150. Adams, R. J., and Pollard, T. D. (1986) Nature
(London) 322, 754756
151. Harada, Y., Noguchi, A., Kishino, A., and
Yanagida, T. (1987) Nature (London) 326, 805
808
152. Rayment, I., Rypniewski, W. R., Schmidt-
Bse, K., Smith, R., Tomchick, D. R., Benning,
M. M., Winkelmann, D. A., Wesenberg, G.,
and Holden, H. M. (1993) Science 261, 5058
153. Fisher, A. J., Smith, C. A., Thoden, J. B., Smith,
R., Sutoh, K., Holden, H. M., and Rayment, I.
(1995) Biochemistry 34, 89608972
154. Smith, C. A., and Rayment, I. (1995) Biochem-
istry 34, 89738981
155. Fisher, A. J., Smith, C. A., Thoden, J., Smith,
R., Sutoh, K., Holden, H. M., and Rayment, I.
(1995) Biophys. J. 68, 19s 29s
156. Rayment, I., Holden, H. M., Whittaker, M.,
Yohn, C. B., Lorenz, M., Holmes, K. C., and
Milligan, R. A. (1993) Science 261, 5865
157. Rayment, I. (1996) J. Biol. Chem. 271, 15850
15853
158. Smith, C. A., and Rayment, I. (1996) Biochem-
istry 35, 54045417
159. Gulick, A. M., Bauer, C. B., Thoden, J. B., and
Rayment, I. (1997) Biochemistry 36, 11619
11628
160. Murphy, R. A. (1994) FASEB J. 8, 311318
161. Xie, X., Harrison, D. H., Schlichting, I., Sweet,
R. M., Kalabokis, V. N., Szent-Gyrgyi, A. G.,
and Cohen, C. (1994) Nature (London) 368,
306312
162. Furch, M., Geeves, M. A., and Manstein, D. J.
(1998) Biochemistry 37, 63176326
163. Van Dijk, J., Fernandez, C., and Chaussepied,
P. (1998) Biochemistry 37, 83858394
164. Yengo, C. M., Chrin, L., Rovner, A. S., and
Berger, C. L. (1999) Biochemistry 38, 14515
14523
165. Grammer, J. C., Kuwayama, H., and Yount, R.
G. (1993) Biochemistry 32, 57255732
166. Miller, C. J., Wong, W. W., Bobkova, E.,
Rubenstein, P. A., and Reisler, E. (1996)
Biochemistry 35, 1655716565
166a. Hansen, J. E., Marner, J., Pavlov, D.,
Rubenstein, P. A., and Reisler, E. (2000)
Biochemistry 39, 17921799
166b. Bertrand, R., Derancourt, J., and Kassab, R.
(2000) Biochemistry 39, 1462614637
166c. Sasaki, N., Ohkura, R., and Sutoh, K. (2000) J.
Biol. Chem. 275, 3870538709
167. Orlova, A., Chen, X., Rubenstein, P. A., and
Egelman, E. H. (1997) J. Mol. Biol. 271, 235
243
167a. Prochniewicz, E., and Thomas, D. D. (2001)
Biochemistry 40, 1393313940
Ch 19IND page -E- 2/14/03, 11:24 AM 1124
1125
168. Belmont, L. D., Orlova, A., Drubin, D. G., and
Egelman, E. H. (1999) Proc. Natl. Acad. Sci.
U.S.A. 96, 2934
168a. Higgins, S. J., and Banting, G., eds. (2000)
Molecular Motors, Vol. Essays in Biochemistry
No. 35, Portland Press, London
168b. Vale, R. D., and Milligan, R. A. (2000) Science
288, 8895
168c. Verhey, K. J., and Rapoport, T. A. (2001) Trends
Biochem. Sci. 26, 545550
169. Sack, S., Mller, J., Marx, A., Thormhlen, M.,
Mandelkow, E.-M., Brady, S. T., and Man-
delkow, E. (1997) Biochemistry 36, 16155
16165
169a. Song, Y.-H., Marx, A., Mller, J., Woehlke, G.,
Schliwa, M., Krebs, A., Hoenger, A., and
Mandelkow, E. (2001) EMBO J. 20, 62136225
169b. Gilbert, S. P. (2001) Nature (London) 414, 597
598
170. Stock, M. F., Guerrero, J., Cobb, B., Eggers, C.
T., Huang, T.-G., Li, X., and Hackney, D. D.
(1999) J. Biol. Chem. 274, 1461714623
171. Vale, R. D., Funatsu, T., Pierce, D. W.,
Romberg, L., Harada, Y., and Yanagida, T.
(1996) Nature (London) 380, 451453
172. Kull, F. J., Sablin, E. P., Lau, R., Fletterick, R. J.,
and Vale, R. D. (1996) Nature (London) 380,
550555
173. Mandelkow, E., and Johnson, K. A. (1998)
Trends Biochem. Sci. 23, 429433
174. Gulick, A. M., Song, H., Endow, S. A., and
Rayment, I. (1998) Biochemistry 37, 17691776
174a. Miki, H., Setou, M., Kaneshiro, K., and
Hirokawa, N. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 70047011
174b. Kikkawa, M., Sablin, E. P., Okada, Y., Yajima,
H., Fletterick, R. J., and Hirokawa, N. (2001)
Nature (London) 411, 439445
174c. Schliwa, M., and Woehlke, G. (2001) Nature
(London) 411, 424425
174d. Yun, M., Zhang, X., Park, C.-G., Park, H.-W.,
and Endow, S. A. (2001) EMBO J. 20, 2611
2618
175. Sablin, E. P., Kull, F. J., Cooke, R., Vale, R. D.,
and Fletterick, R. J. (1996) Nature (London) 380,
555559
176. Thormhlen, M., Marx, A., Mller, S. A., Song,
Y.-H., Mandelkow, E.-M., Aebi, U., and
Mandelkow, E. (1998) J. Mol. Biol. 275, 795
809
177. Block, S. M. (1998) Cell 93, 58
178. Wells, A. L., Lin, A. W., Chen, L.-Q., Safer, D.,
Cain, S. M., Hasson, T., Carragher, B. O.,
Milligan, R. A., and Sweeney, H. L. (1999)
Nature (London) 401, 505508
179. Schliwa, M. (1999) Nature (London) 401, 431
432
179a. Homma, K., Yoshimura, M., Saito, J., Ikebe,
R., and Ikebe, M. (2001) Nature (London) 412,
831834
180. Ma, Y.-Z., and Taylor, E. W. (1995) Biochemistry
34, 1324213251
181. Ma, Y.-Z., and Taylor, E. W. (1997) J. Biol.
Chem. 272, 717723
182. Visscher, K., Schnitzer, M. J., and Block, S. M.
(1999) Nature (London) 400, 184189
182a. Tomishige, M., Klopfenstein, D. R., and Vale,
R. D. (2002) Science 297, 22632267
183. Mehta, A. D., Rock, R. S., Rief, M., Spudich, J.
A., Mooseker, M. S., and Cheney, R. E. (1999)
Nature (London) 400, 590593
183a. Rief, M., Rock, R. S., Mehta, A. D., Mooseker,
M. S., Cheney, R. E., and Spudich, J. A. (2000)
Proc. Natl. Acad. Sci. U.S.A. 97, 94829486
183b. Trybus, K. M., Krementsova, E., and Freyzon,
Y. (1999) J. Biol. Chem. 274, 2744827456
183c. De La Cruz, E. M., Wells, A. L., Sweeney, H.
L., and Ostap, E. M. (2000) Biochemistry 39,
1419614202
183d. Rock, R. S., Rice, S. E., Wells, A. L., Purcell, T.
J., Spudich, J. A., and Sweeney, H. L. (2001)
Proc. Natl. Acad. Sci. U.S.A. 98, 1365513659
183e. Karcher, R. L., Roland, J. T., Zappacosta, F.,
Huddleston, M. J., Annan, R. S., Carr, S. A.,
and Gelfand, V. I. (2001) Science 293, 1317
1320
184. Goldman, Y. E. (1998) Cell 93, 14
185. Taylor, E. W. (1991) J. Biol. Chem. 266, 294302
186. Houadjeto, M., Travers, F., and Barman, T.
(1992) Biochemistry 31, 15641569
187. White, H. D., Belknap, B., and Webb, M. R.
(1997) Biochemistry 36, 1182811836
188. Spudich, J. A. (1994) Nature (London) 372,
515518
189. Cooke, R. (1993) FASEB J. 9, 636642
190. Ostap, E. M., Barnett, V. A., and Thomas, D.
D. (1995) Biophys. J. 69, 177188
191. Kambara, T., Rhodes, T. E., Ikebe, R., Yamada,
M., White, H. D., and Ikebe, M. (1999) J. Biol.
Chem. 274, 1640016406
191a. Lionne, C., Stehle, R., Travers, F., and Barman,
T. (1999) Biochemistry 38, 85128520
192. Harrington, W. F., and Rodgers, M. E. (1984)
Ann. Rev. Biochem. 53, 3573
192a. Uyeda, T. Q. P., Tokuraku, K., Kaseda, K.,
Webb, M. R., and Patterson, B. (2002)
Biochemistry 41, 95259534
192b. Himmel, D. M., Gourinath, S., Reshetnikova,
L., Shen, Y., Szent-Gyrgyi, A. G., and Cohen,
C. (2002) Proc. Natl. Acad. Sci. U.S.A. 99,
1264512650
193. Yount, R. G., Lawson, D., and Rayment, I.
(1995) Biophys. J. 68, 445495
193a. Burghardt, T. P., Park, S., and Ajtai, K. (2001)
Biochemistry 40, 48344843
193b. Borejdo, J., Ushakov, D. S., Moreland, R.,
Akopova, I., Reshetnyak, Y., Saraswat, L. D.,
Kamm, K., and Lowey, S. (2001) Biochemistry
40, 37963803
194. Dominguez, R., Freyzon, Y., Trybus, K. M.,
and Cohen, C. (1998) Cell 94, 559571
195. Highsmith, S. (1999) Biochemistry 38, 9791
9797
196. Gulick, A. M., Bauer, C. B., Thoden, J. B., Pate,
E., Yount, R. G., and Rayment, I. (2000) J. Biol.
Chem. 275, 398408
197. Geeves, M. A., and Holmes, K. C. (1999) Ann.
Rev. Biochem. 68, 687728
198. Block, S. M. (1992) Nature (London) 360, 493
495
199. Mehta, A. D., Rief, M., Spudich, J. A., Smith,
D. A., and Simmons, R. M. (1999) Science 283,
16891695
200. Finer, J. T., Simmons, R. M., and Spudich, J. A.
(1994) Nature (London) 368, 113119
201. Molloy, J. E., Burns, J. E., Kendrick-Jones, J.,
Tregear, R. T., and White, D. C. S. (1995)
Nature (London) 378, 209212
202. Mehta, A. D., Rief, M., and Spudich, J. A.
(1999) J. Biol. Chem. 274, 1451714520
203. Howard, J. (1997) Nature (London) 389, 561
567
204. Guilford, W. H., Dupuis, D. E., Kennedy, G.,
Wu, J., Patlak, J. B., and Warshaw, D. M.
(1997) Biophys. J. 72, 10061021
204a. Rosenfeld, S. S., Xing, J., Whitaker, M.,
Cheung, H. C., Brown, F., Wells, A., Milligan,
R. A., and Sweeney, H. L. (2000) J. Biol. Chem.
275, 2541825426
205. Sase, I., Miyata, H., Ishiwata, S., and Kinosita,
K., Jr. (1997) Proc. Natl. Acad. Sci. U.S.A. 94,
56465650
206. Kitamura, K., Tokunaga, M., Iwane, A. H.,
and Yanagida, T. (1999) Nature (London) 397,
129134
207. Smith, D. A. (1998) Biophys. J. 75, 29963007
207a. Yanagida, T., and Iwane, A. H. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 93579359
208. Veigel, C., Coluccio, L. M., Jontes, J. D.,
Sparrow, J. C., Milligan, R. A., and Molloy, J.
E. (1999) Nature (London) 398, 530533
208a. Piazzesi, G., Reconditi, M., Linari, M., Lucii,
L., Sun, Y.-B., Narayanan, T., Boesecke, P.,
Lombardi, V., and Irving, M. (2002) Nature
(London) 415, 659662
208b. Tanaka, H., Homma, K., Iwane, A. H.,
Katayama, E., Ikebe, R., Saito, J., Yanagida, T.,
and Ikebe, M. (2002) Nature (London) 415,
192195
209. Berliner, E., Mahtani, H. K., Karki, S., Chu, L.
F., Cronan, J. E., Jr., and Gelles, J. (1994) J. Biol.
Chem. 269, 86108615
210. Schnitzer, M. J., and Block, S. M. (1997) Nature
(London) 388, 386390
211. Coppin, C. M., Finer, J. T., Spudich, J. A., and
Vale, R. D. (1996) Proc. Natl. Acad. Sci. U.S.A.
93, 19131917
212. Irving, M., and Goldman, Y. E. (1999) Nature
(London) 398, 463465
212a. Shimizu, T., Thorn, K. S., Ruby, A., and Vale,
R. D. (2000) Biochemistry 39, 52655273
212b. Ryu, W. S., Berry, R. M., and Berg, H. C. (2000)
Nature (London) 403, 444447
213. Young, E. C., Mahtani, H. K., and Gelles, J.
(1998) Biochemistry 37, 34673479
214. Foster, K. A., Correia, J. J., and Gilbert, S. P.
(1998) J. Biol. Chem. 273, 3530735318
215. deCastro, M. J., Ho, C.-H., and Stewart, R. J.
(1999) Biochemistry 38, 50765081
216. Platts, J. A., Howard, S. T., and Bracke, B. R. F.
(1996) J. Am. Chem. Soc. 118, 27262733
217. Sosa, H., and Milligan, R. A. (1996) J. Mol.
Biol. 260, 743755
218. Hirose, K., Cross, R. A., and Amos, L. A.
(1998) J. Mol. Biol. 278, 389400
219. Sablin, E. P., Case, R. B., Dai, S. C., Hart, C. L.,
Ruby, A., Vale, R. D., and Fletterick, R. J.
(1998) Nature (London) 395, 813816
220. Hirose, K., Lockhart, A., Cross, R. A., and
Amos, L. A. (1996) Proc. Natl. Acad. Sci. U.S.A.
93, 95399544
220a. Hua, W., Chung, J., and Gelles, J. (2002)
Science 295, 844848
221. Rice, S., Lin, A. W., Safer, D., Hart, C. L.,
Naber, N., Carragher, B. O., Cain, S. M.,
Pechatnikova, E., Wilson-Kubalek, E. M.,
Whittaker, M., Pate, E., Cooke, R., Taylor, E.
W., Milligan, R. A., and Vale, R. D. (1999)
Nature (London) 402, 778784
222. Hancock, W. O., and Howard, J. (1999) Proc.
Natl. Acad. Sci. U.S.A. 96, 1314713152
223. Crevel, I., Carter, N., Schliwa, M., and Cross,
R. (1999) EMBO J. 18, 58635872
224. Brendza, K. M., Rose, D. J., Gilbert, S. P., and
Saxton, W. M. (1999) J. Biol. Chem. 274, 31506
31514
225. Pate, E., Naber, N., Matuska, M., Franks-
Skiba, K., and Cooke, R. (1997) Biochemistry
36, 1215512166
226. Moschcovich, L., Peyser, Y. M., Salomon, C.,
Burghardt, T. P., and Muhlrad, A. (1998)
Biochemistry 37, 1513715143
227. Gopal, D., Pavlov, D. I., Levitsky, D. I., Ikebe,
M., and Burke, M. (1996) Biochemistry 35,
1014910157
227a. Kliche, W., Pfannstiel, J., Tiepold, M., Stoeva,
S., and Faulstich, H. (1999) Biochemistry 38,
1030710317
227b. Furch, M., Fujita-Becker, S., Geeves, M. A.,
Holmes, K. C., and Manstein, D. J. (1999) J.
Mol. Biol. 290, 797809
227c. Allin, C., and Gerwert, K. (2001) Biochemistry
40, 30373046
227d. Nyitrai, M., Hild, G., Lukcs, A., Bdis, E.,
and Somogyi, B. (2000) J. Biol. Chem. 275,
24042409
References
Ch 19IND page -E- 2/14/03, 11:24 AM 1125
1126 Chapter 19. The Chemistry of Movement
References
228. Xiao, M., Li, H., Snyder, G. E., Cooke, R.,
Yount, R. G., and Selvin, P. R. (1998) Proc.
Natl. Acad. Sci. U.S.A. 95, 1530915314
229. Palm, T., Sale, K., Brown, L., Li, H., Hambly,
B., and Fajer, P. G. (1999) Biochemistry 38,
1302613034
230. Corrie, J. E. T., Brandmeier, B. D., Ferguson, R.
E., Trentham, D. R., Kendrick-Jones, J.,
Hopkins, S. C., van der Heide, U. A., Gold-
man, Y. E., Sabido-David, C., Dale, R. E.,
Criddle, S., and Irving, M. (1999) Nature
(London) 400, 425430
231. Suzuki, Y., Yasunaga, T., Ohkura, R., Wakaba-
yashi, T., and Sutoh, K. (1998) Nature (London)
396, 380383
232. Ashley, R. (1972) J. Theor. Biol. 36, 339354
233. McClare, C. W. F. (1972) J. Theor. Biol. 35, 569
595
234. Davydov, A. S. (1973) J. Theor. Biol. 38, 559569
235. Porter, K. R., and Franzini-Armstrong, C.
(1965) Sci. Am. 212(Mar), 7380
236. Hoyle, G. (1970) Sci. Am. 222(Apr), 8593
237. Alberts, B., Bray, D., Lewis, J., Raff, M.,
Roberts, K., and Watson, J. D. (1994) Molecular
Biology of the Cell, 3rd ed., Garland, New York
238. Macrez-Leprtre, N., Kalkbrenner, F., Schultz,
G., and Mironneau, J. (1997) J. Biol. Chem. 272,
52615268
238a. Toyoshima, C., and Nomura, H. (2002) Nature
(London) 418, 605611
239. Sharma, M. R., Penczek, P., Grassucci, R., Xin,
H.-B., Fleischer, S., and Wagenknecht, T.
(1998) J. Biol. Chem. 273, 1842918434
240. Bers, D. M., and Fill, M. (1998) Science 281,
790791
241. Sonnleitner, A., Conti, A., Bertocchini, F.,
Schindler, H., and Sorrentino, V. (1998) EMBO
J. 17, 27902798
242. Du, G. G., and MacLennan, D. H. (1998) J.
Biol. Chem. 273, 3186731872
243. Bidasee, K. R., and Besch, H. R., Jr. (1998) J.
Biol. Chem. 273, 1217612186
243a. Sun, J., Xin, C., Eu, J. P., Stamler, J. S., and
Meissner, G. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 1115811162
243b. Feng, W., Liu, G., Allen, P. D., and Pessah, I.
N. (2000) J. Biol. Chem. 275, 3590235907
243c. Patel, S., Churchill, G. C., and Galione, A.
(2001) Trends Biochem. Sci. 26, 482489
243d. Baker, M. L., Serysheva, I. I., Sencer, S., Wu, Y.,
Ludtke, S. J., Jiang, W., Hamilton, S. L., and
Chiu, W. (2002) Proc. Natl. Acad. Sci. U.S.A. 99,
1215512160
244. Takeshima, H., Komazaki, S., Hirose, K.,
Nishi, M., Noda, T., and Lino, M. (1998)
EMBO J. 17, 33093316
245. Zhou, J., Cribbs, L., Yi, J., Shirokov, R., Perez-
Reyes, E., and Ros, E. (1998) J. Biol. Chem. 273,
2550325509
245a. Serysheva, I. I., Ludtke, S. J., Baker, M. R.,
Chiu, W., and Hamilton, S. L. (2002) Proc.
Natl. Acad. Sci. U.S.A. 99, 10370 10375
246. Knudson, C. M., Chaudhari, N., Sharp, A. H.,
Powell, J. A., Beam, K. G., and Campbell, K. P.
(1989) J. Biol. Chem. 264, 13451348
247. Smith, G. D., Keizer, J. E., Stern, M. D.,
Lederer, W. J., and Cheng, H. (1998) Biophys. J.
75, 1532
248. Fay, F. S. (1995) Science 270, 588589
248a. Kamm, K. E., and Stull, J. T. (2001) J. Biol.
Chem. 276, 45274530
249. Xie, X., Rao, S., Walian, P., Hatch, V., Phillips,
G. N., Jr., and Cohen, C. (1994) J. Mol. Biol.
236, 12121226
250. Dufour, C., Weinberger, R. P., Schevzov, G.,
Jeffrey, P. L., and Gunning, P. (1998) J. Biol.
Chem. 273, 1854718555
251. Kagawa, H., Sugimoto, K., Matsumoto, H.,
Inoue, T., Imadzu, H., Takuwa, K., and
Sakube, Y. (1995) J. Mol. Biol. 251, 603613
252. Amos, L. A. (1985) Ann. Rev. Biophys. Biophys.
Chem. 14, 291313
253. Zot, A. S., and Potter, J. D. (1987) Ann. Rev.
Biophys. Biophys. Chem. 16, 535559
254. Saeki, K., Sutoh, K., and Wakabayashi, T.
(1996) Biochemistry 35, 1446514472
255. Squire, J. M., and Morris, E. P. (1998) FASEB J.
12, 761771
256. Tripet, B., Van Eyk, J. E., and Hodges, R. S.
(1997) J. Mol. Biol. 271, 728750
257. Malnic, B., Farah, C. S., and Reinach, F. C.
(1998) J. Biol. Chem. 273, 1059410601
258. Hernndez, G., Blumenthal, D. K., Kennedy,
M. A., Unkefer, C. J., and Trewhella, J. (1999)
Biochemistry 38, 69116917
259. Vassylyev, D. G., Takeda, S., Wakatsuki, S.,
Maeda, K., and Meda, Y. (1998) Proc. Natl.
Acad. Sci. U.S.A. 95, 48474852
260. McKay, R. T., Tripet, B. P., Pearlstone, J. R.,
Smillie, L. B., and Sykes, B. D. (1999) Biochem-
istry 38, 54785489
260a. Mercier, P., Spyracopoulos, L., and Sykes, B.
D. (2001) Biochemistry 40, 1006310077
261. Kobayashi, T., Kobayashi, M., Gryczynski, Z.,
Lakowicz, J. R., and Collins, J. H. (2000)
Biochemistry 39, 8691
262. Strynadka, N. C. J., Cherney, M., Sielecki, A.
R., Li, M. X., Smillie, L. B., and James, M. N.
G. (1997) J. Mol. Biol. 273, 238255
263. da Silva, A. C. R., and Reinach, F. C. (1991)
Trends Biochem. Sci. 16, 5357
264. Gagn, S. M., Li, M. X., and Sykes, B. D. (1997)
Biochemistry 36, 43864392
265. Li, H.-C., and Fajer, P. G. (1998) Biochemistry
37, 66286635
265a. Lehman, W., Rosol, M., Tobacman, L. S., and
Craig, R. (2001) J. Mol. Biol. 307, 739744
266. Reiffert, S. U., Jaquet, K., Heilmeyer, L. M. G.,
Jr., and Herberg, F. W. (1998) Biochemistry 37,
1351613525
267. Simmerman, H. K. B., Kobayashi, Y. M.,
Autry, J. M., and Jones, L. R. (1996) J. Biol.
Chem. 271, 59415946
268. Reddy, L. G., Jones, L. R., and Thomas, D. D.
(1999) Biochemistry 38, 39543962
268a. Asahi, M., Green, N. M., Kurzydlowski, K.,
Tada, M., and MacLennan, D. H. (2001) Proc.
Natl. Acad. Sci. U.S.A. 98, 1006110066
269. Holmes, K. C. (1995) Biophys. J. 68, 2s 7s
270. Vibert, P., Craig, R., and Lehman, W. (1997) J.
Mol. Biol. 266, 814
270a. Moraczewska, J., and Hitchcock-DeGregori, S.
E. (2000) Biochemistry 39, 68916897
270b. Narita, A., Yasunaga, T., Ishikawa, T.,
Mayanagi, K., and Wakabayashi, T. (2001) J.
Mol. Biol. 308, 241261
270c. Craig, R., and Lehman, W. (2001) J. Mol. Biol.
311, 10271036
271. Hnath, E. J., Wang, C.-L. A., Huber, P. A. J.,
Marston, S. B., and Phillips, G. N., Jr. (1996)
Biophys. J. 71, 19201933
271a. Gerson, J. H., Kim, E., Muhlrad, A., and
Reisler, E. (2001) J. Biol. Chem. 276, 18442
18449
272. Allen, B. G., and Walsh, M. P. (1994) Trends
Biochem. Sci. 19, 362368
273. Ikura, M., Clore, G. M., Gronenborn, A. M.,
Zhu, G., Klee, C. B., and Bax, A. (1992) Science
256, 632638
274. Wu, X., Clack, B. A., Zhi, G., Stull, J. T., and
Cremo, C. R. (1999) J. Biol. Chem. 274, 20328
20335
275. Lee, M. R., Li, L., and Kitazawa, T. (1997) J.
Biol. Chem. 272, 50635068
276. Ye, L.-H., Kishi, H., Nakamura, A., Okagaki,
T., Tanaka, T., Oiwa, K., and Kohama, K.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 6666
6671
277. Lehman, W., Vibert, P., and Craig, R. (1997) J.
Mol. Biol. 274, 310317
278. Graceffa, P. (1997) Biochemistry 36, 37923801
278a. Wang, Z., and Yang, Z.-Q. (2000) Biochemistry
39, 1111411120
279. Gollub, J., Cremo, C. R., and Cooke, R. (1999)
Biochemistry 38, 1010710118
279a. Stafford, W. F., Jacobsen, M. P., Woodhead, J.,
Craig, R., ONeall-Hennessey, E. O., and
Szent-Gyrgyi, A. G. (2001) J. Mol. Biol. 307,
137147
280. Yamada, A., Yoshio, M., Oiwa, K., and
Nyitray, L. (2000) J. Mol. Biol. 295, 169178
280a. Yamada, A., Yoshio, M., Kojima, H., and
Oiwa, K. (2001) Proc. Natl. Acad. Sci. U.S.A. 98,
66356640
281. Brzeska, H., and Korn, E. D. (1996) J. Biol.
Chem. 271, 1698316986
282. Benian, G. M., Kiff, J. E., Neckelmann, N.,
Moerman, D. G., and Waterston, R. H. (1989)
Nature (London) 342, 4550
283. Heierhorst, J., Kobe, B., Feil, S. C., Parker, M.
W., Benian, G. M., Weiss, K. R., and Kemp, B.
E. (1996) Nature (London) 380, 636639
284. Lei, J., Tang, X., Chambers, T. C., Pohl, J., and
Benian, G. M. (1994) J. Biol. Chem. 269, 21078
21085
285. Johnson, K. A., and Quiocho, F. A. (1996)
Nature (London) 380, 585587
286. Bessman, S. P., and Carpenter, C. L. (1985)
Ann. Rev. Biochem. 54, 831862
287. Balaban, R. S., Kantor, H. L., and Ferretti, J. A.
(1983) J. Biol. Chem. 258, 1278712789
288. Park, J. H., Brown, R. L., Park, C. R., McCully,
K., Cohn, M., Haselgrove, J., and Chance, B.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 8976
8980
289. Savabi, F. (1988) Proc. Natl. Acad. Sci. U.S.A.
85, 74767480
289a. Kashiyama, T., Ito, K., and Yamamoto, K.
(2001) J. Mol. Biol. 311, 461466
289b. Pantaloni, D., Le Clainche, C., and Carlier, M.-
F. (2001) Science 292, 15021506
289c. Pollard, T. D. (2000) Trends Biochem. Sci. 25,
607611
290. Machesky, L. M., and Way, M. (1998) Nature
(London) 394, 125126
291. Carlier, M.-F., and Pantaloni, D. (1997) J. Mol.
Biol. 269, 459467
291a. Pruyne, D., Evangelista, M., Yang, C., Bi, E.,
Zigmond, S., Bretscher, A., and Boone, C.
(2002) Science 297, 612615
292. Yin, H. L., and Stull, J. T. (1999) J. Biol. Chem.
274, 3252932530
292a. Robinson, R. C., Turbedsky, K., Kaiser, D. A.,
Marchand, J.-B., Higgs, H. N., Choe, S., and
Pollard, T. D. (2001) Science 294, 16791684
292b. Dayel, M. J., Holleran, E. A., and Mullins, R.
D. (2001) Proc. Natl. Acad. Sci. U.S.A. 98,
1487114876
293. Svitkina, T. M., and Borisy, G. G. (1999) Trends
Biochem. Sci. 24, 432436
293a. Boldogh, I. R., Yang, H.-C., Nowakowski, W.
D., Karmon, S. L., Hays, L. G., Yates, J. R., III,
and Pon, L. A. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 31623167
293b. Kim, A. S., Kakalis, L. T., Abdul-Manan, N.,
Liu, G. A., and Rosen, M. K. (2000) Nature
(London) 404, 151158
293c. Higgs, H. (2001) Trends Biochem. Sci. 26, 219
293d. Kaibuchi, K., Kuroda, S., and Amano, M.
(1999) Ann. Rev. Biochem. 68, 459486
293e. Castellano, F., Le Clainche, C., Patin, D.,
Carlier, M.-F., and Chavrier, P. (2001) EMBO J.
20, 56035614
Ch 19IND page -E- 2/14/03, 11:25 AM 1126
1127
Study Questions
1. Describe briefly major aspects of the structure,
properties, locations, and functions of each of the
following proteins of skeletal muscle.
Actin Tropomyosin
Myosin Troponin
Titin Myomesin
Nebulin Desmin
-Actinin Vimentin
C-protein
2. Describe the generally accepted sliding filament
model of muscle contraction. List some uncertain-
ties in this description.
3. Compare mechanisms that regulate contraction in
skeletal muscle and in smooth muscle.
4. Compare myosin with kinesins and dyneins.
What features do they have in common? What
differences can you describe?
5. Compare the properties of actin in skeletal muscle
and in nonmuscle cells. What is meant by
treadmilling? What is actin-based motility?
6. The human genome contains more than 100 genes
for proteins of the kinesin superfamily. Why?
7. Describe some of the major diseases that involve
muscle proteins.
References
293f. Reinhard, M., Jarchau, T., and Walter, U.
(2001) Trends Biochem. Sci. 26, 243249
294. Gutsche-Perelroizen, I., Lepault, J., Ott, A.,
and Carlier, M.-F. (1999) J. Biol. Chem. 274,
62346243
295. Kang, F., Purich, D. L., and Southwick, F. S.
(1999) J. Biol. Chem. 274, 3696336972
296. Chen, H., Bernstein, B. W., and Bamburg, J. R.
(2000) Trends Biochem. Sci. 25, 1923
297. Carlier, M.-F., Ressad, F., and Pantaloni, D.
(1999) J. Biol. Chem. 274, 3382733830
297a. Ono, S., McGough, A., Pope, B. J., Tolbert, V.
T., Bui, A., Pohl, J., Benian, G. M., Gernert, K.
M., and Weeds, A. G. (2001) J. Biol. Chem. 276,
59525958
298. Theriot, J. A., and Mitchison, T. J. (1991)
Nature (London) 352, 126131
299. Stossel, T. P. (1993) Science 260, 10861094
300. Tsukita, S., and Yonemura, S. (1999) J. Biol.
Chem. 274, 3450734510
301. Hanakam, F., Gerisch, G., Lotz, S., Alt, T., and
Seelig, A. (1996) Biochemistry 35, 1103611044
301a. Niggli, V. (2001) Trends Biochem. Sci. 26, 604
611
301b. Caroni, P. (2001) EMBO J. 20, 43324336
301c. van den Ent, F., Amos, L. A., and Lwe, J.
(2001) Nature (London) 413, 3944
302. Bullock, T. L., Roberts, R. M., and Stewart, M.
(1996) J. Mol. Biol. 263, 284296
302a. Villeneuve, A. M. (2001) Science 291, 2099
2101
303. Spudich, J. D., and Lord, K. (1974) J. Biol.
Chem. 249, 60136020
304. Allen, R. D. (1987) Sci. Am. 256(Feb), 4247
305. Gibbons, I. R. (1988) J. Biol. Chem. 263, 15837
15840
305a. Setou, M., Nakagawa, T., Seog, D.-H., and
Hirokawa, N. (2000) Science 288, 17961802
305b. Goldstein, L. S. B. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 69997003
305c. Fan, J.-S., Zhang, Q., Tochio, H., Li, M., and
Zhang, M. (2001) J. Mol. Biol. 306, 97108
306. Rodionov, V., Nadezhdina, E., and Borisy, G.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 115120
307. Burgess, S. A. (1995) J. Mol. Biol. 250, 5263
308. Caplow, M., and Shanks, J. (1998) Biochemistry
37, 1299413002
309. Dougherty, C. A., Himes, R. H., Wilson, L.,
and Farrell, K. W. (1998) Biochemistry 37,
1086110865
310. Hyams, J. S., and Lloyd, C. W., eds. (1994)
Microtubules, Wiley-Liss, New York
311. Drewes, G., Ebneth, A., and Mandelkow, E.-
M. (1998) Trends Biochem. Sci. 23, 307 311
312. Vinh, J., Langridge, J. I., Br, M.-H., Levilliers,
N., Redeker, V., Loyaux, D., and Rossier, J.
(1999) Biochemistry 38, 31333139
313. Redeker, V., Levilliers, N., Schmitter, J.-M., Le
Caer, J.-P., Rossier, J., Adoutte, A., and Br,
M.-H. (1994) Science 266, 16881691
314. Regnard, C., Audebert, S., Desbruyres, E.,
Denoulet, P., and Edd, B. (1998) Biochemistry
37, 83958404
314a. Sharp, D. J., Rogers, G. C., and Scholey, J. M.
(2000) Nature (London) 407, 4147
315. Hoyt, M. A., Hyman, A. A., and Bhler, M.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 12747
12748
316. Barton, N. R., and Goldstein, L. S. B. (1996)
Proc. Natl. Acad. Sci. U.S.A. 93, 17351742
317. Habura, A., Tikhonenko, I., Chisholm, R. L.,
and Koonce, M. P. (1999) J. Biol. Chem. 274,
1544715453
318. Sams, M., Radermacher, M., Frank, J., and
Koonce, M. P. (1998) J. Mol. Biol. 276, 927937
319. Shimizu, T., Toyoshima, Y. Y., Edamatsu, M.,
and Vale, R. D. (1995) Biochemistry 34, 1575
1582
320. King, S. M., Barbarese, E., Dillman, J. F., III,
Patel-King, R. S., Carson, J. H., and Pfister, K.
K. (1996) J. Biol. Chem. 271, 1935819366
321. Koonce, M. P., Khler, J., Neujahr, R.,
Schwartz, J.-M., Tikhonenko, I., and Gerisch,
G. (1999) EMBO J. 18, 67866792
322. Sleigh, M. A., ed. (1974) Cilia and Flagella,
Academic Press, New York
323. Smith, E. F., and Sale, W. S. (1994) in Microtu-
bules (Hyams, J. S., and Lloyd, C. W., eds), pp.
381392, Wiley-Liss, New York
324. Tanaka-Takiguchi, Y., Itoh, T. J., and Hotani,
H. (1998) J. Mol. Biol. 280, 365373
325. Satir, P. (1999) FASEB J. 13, S235S237
325a. Fang, Y.-I., Yokota, E., Mabuchi, I., Nakamura,
H., and Obizumi, Y. (1997) Biochemistry 36,
1556115567
325b. Sakakibara, H., Kojima, H., Sakai, Y.,
Katayama, E., and Oiwa, K. (1999) Nature
(London) 400, 586590
326. Shingyoji, C., Higuchi, H., Yoshimura, M.,
Katayama, E., and Yanagida, T. (1998) Nature
(London) 393, 711714
327. Hunt, A. J. (1998) Nature (London) 393, 624
625
328. Benashski, S. E., Patel-King, R. S., and King, S.
M. (1999) Biochemistry 38, 72537264
329. Sleigh, M. A., ed. (1974) Cilia and Flagella, p. 14
Academic Press, New York
329a. Smith, E. F., and Sale, W. S. (1994) in
Microtubules (Hyams, J. S., and Lloyd, C. W.,
eds), pp. 381392, Wiley-Liss, New York
330. Klein, P. S., Sun, T. J., Saxe, C. L., III, Kimmel,
A. R., Johnson, R. L., and Devreotes, P. N.
(1988) Science 241, 14671472
331. Caterina, M. J., and Devreotes, P. N. (1991)
FASEB J. 5, 30783085
332. Snyderman, R., and Goetzl, E. J. (1981) Science
213, 830837
333. Gerard, N. P., Bao, L., Xiao-Ping, H., Eddy, R.
L., Jr., Shows, T. B., and Gerard, C. (1993)
Biochemistry 32, 12431250
334. Thelen, M., Peveri, P., Kernen, P., von
Tscharner, V., Walz, A., and Baggiolini, M.
(1988) FASEB J. 2, 27022706
335. Schwenk, U., and Schrder, J.-M. (1995) J. Biol.
Chem. 270, 1502915036
336. Hsu, M. H., Chiang, S. C., Ye, R. D., and
Prossnitz, E. R. (1997) J. Biol. Chem. 272,
2942629429
337. Jin, T., Zhang, N., Long, Y., Parent, C. A., and
Devreotes, P. N. (2000) Science 287, 10341036
338. Chung, C. Y., Funamoto, S., and Firtel, R. A.
(2001) Trends Biochem. Sci. 26, 557566
339. Mahadevan, L., and Matsudaira, P. (2000)
Science 288, 9599
340. Merz, A. J., So, M., and Sheetz, M. P. (2000)
Nature (London) 407, 98102
341. Kirchhausen, T. (1999) Nature (London) 398,
470471
342. McNiven, M. A., Cao, H., Pitts, K. R., and
Yoon, Y. (2000) Trends Biochem. Sci. 25, 115
120
Ch 19IND page -E- 2/14/03, 11:25 AM 1127
1128 Chapter 20. Some Pathways of Carbohydrate Metabolism
N
HO
OH
HO
H
HO
Castanospermine
1
2
3
5
6
7
8

Many steps
G
G
G
M
M
M
M
M
Gn
Gn
Asn
M M
M M
M
M
M
Gn
Gn
Asn
Gn Gn
Gn
Gal Gal
SA SA
Gal
SA
Fuc
Trimming of
glycoproteins is
one function of
the Golgi
1129
1130
1131
1132
1135
1136
1137
1140
1143
1143
1146
1147
1148
1148
1149
1150
1152
1152
1153
1153
1155
1155
1156
1157
1157
1159
1160
1160
1161
1161
1168
1168
1168
1169
1169
1170
1170
1172
1173
1179
A. Interconversions of Monosaccharides
1. The Metabolism of Galactose
2. Inositol
3. d-Glucuronic Acid, Ascorbic Acid, and Xylitol
4. Transformations of Fructose 6-Phosphate
5. Extending a Sugar Chain with
Phosphoenolpyruvate (PEP)
6. Synthesis of Deoxy Sugars
B. Synthesis and Utilization of Oligosaccharides
C. Synthesis and Degradation of Polysaccharides
1. Glycogen and Starch
2. Cellulose, Chitin, and Related Glycans
An insertion mechanism for synthesis of cellulose
Callose and other -1,3-linked glycans
Chitin
Cell walls of plants
3. Patterns in Polysaccharide Structures
Dextrans
Lipid-dependent synthesis of polysaccharides
D. Proteoglycans and Glycoproteins
1. Glycosaminoglycans
2. O-Linked Oligosaccharides
3. Assembly of N-Linked Oligosaccharides on
Glycosyl Carrier Lipids
Trimming of glycoprotein oligosaccharides
Extensions and terminal elaborations
The perplexing Golgi apparatus
Lysosomal enzymes
The hepatic asialoglycoprotein (Gal) receptor
E. Biosynthesis of Bacterial Cell Walls
The O-antigens and lipid A
Gram-positive bacteria
F. Biosynthesis of Eukaryotic Glycolipids
1. Glycophosphatidylinositol (GPI) Anchors
2. Cerebrosides and Gangliosides
G. The Intracellular Breakdown of Polysaccharides
and Glycolipids
1. Mucopolysaccharidoses
2. Sphingolipidoses
3. Causes of Lysosomal Diseases
4. Can Lysosomal Diseases Be Treated?
References
Study Questions
1131
1139
1142
1145
1149
1151
1164
1166
1171
Boxes
Box 20-A Fructose for Sperm Cells via the
Polyol Pathway
Box 20-B The Biosynthesis of Streptomycin
Box 20-C Osmotic Adaptation
Box 20-D Genetic Diseases of Glycogen
Metabolism
Box 20-E Oligosaccharides in Defensive and
Other Responses of Plants
Box 20-F What Does Boron Do?
Box 20-G Penicillins and Related Antibiotics
Box 20-H Antibiotic Resistance and Vancomycin
Tables
Table 20-1 Lysosomal Storage Diseases:
Sphingolipidoses and
Mucopolysaccharidoses
Contents
The branched oligosaccharides of glycoprotein surfaces are
formed on asparagine side chains of selected cell surface
proteins. The oligosaccharide at the left is formed in the ER
and is transferred intact (Fig. 20-6) to an acceptor asparagine.
It is then trimmed by removal of glucose and mannose units
and residues of glucosamine, galactose, and fucose are added
as in Fig. 20-7. These reactions begin in the ER and continue in
the Golgi apparatus (right). See also Fig. 20-8.
Ch 20 IND page-E- 2/14/03, 11:27 AM 1128
1129
Some Pathways of
Carbohydrate Metabolism 20
The general principles of biosynthesis, as well
as the pathways of formation of major carbohydrate
and lipid precursors, are considered in Chapter 17.
Also described are the processes of gluconeogenesis,
the synthesis of glucose 6-phosphate and fructose
6-phosphate from free glucose, and typical polymer-
ization pathways for formation of polysaccharides. In
this chapter, additional aspects of the metabolism of
monosaccharides, oligosaccharides, polysaccharides,
glycoproteins, and glycolipids are considered. These
are metabolic transformations that affect the physical
properties of cell surfaces and body fluids. They are
essential to signaling between cells, to establishment
of the immunological identity of individuals, and to
the development of strong cell wall materials. Some
of the differences in carbohydrates found in bacteria,
fungi, green plants, and mammals are considered.
A. Interconversions of Monosaccharides
Chemical interconversions between compounds
are easiest at the level of oxidation of carbohydrates.
Consequently, many reactions by which one sugar can
be changed into another are known. Most of the trans-
formations take place in the sugar nucleotide deriva-
tives (see also Eq. 17-56). The first of this group of
compounds to be recognized was uridine diphosphate
glucose (UDPG), which was discovered around 1950
by L. F. Leloir
1,2
during his investigation of the metab-
olism of galactose 1-P. The fact that interconversions
of hexoses take place largely at the sugar nucleotide
level was unknown at the time. Leloirs studies led
to the characterization of both UDP-glucose and UDP-
galactose.
Figure 20-1 summarizes pathways by which glucose
6-phosphate or fructose 6-phosphate can be converted
into many of the other sugars found in living things.
Galactose and mannose can also be interconverted
with the other sugars. A kinase forms mannose 6-
phosphate which equilibrates with fructose 6-phosphate.
Galactokinase converts free galactose to galactose 1-
phosphate, which can be isomerized to glucose 1-
phosphate by the reactions of Eq. 20-1. Fructose, an
important human dietary constituent
3
derived largely
by hydrolysis of sucrose, can also be formed in tissues
via the sorbitol pathway
4
(Box 20-A). Fructose can
be phosphorylated to fructose 1-phosphate by liver
fructokinase. We have no mutase able to convert
fructose 1-P to fructose 6-P, but a special aldolase
cleaves fructose 1-P to dihydroxyacetone phosphate
and free glyceraldehyde. Lack of this aldolase leads to
occasionally observed cases of fructose intolerance.
5,6
The glyceraldehyde formed from fructose can be
metabolized by reduction to glycerol followed by
phosphorylation (glycerol kinase) and reoxidation to
dihydroxyacetone phosphate. Some phosphorylation of
fructose 1-P to fructose 1,6-P
2
apparently also occurs.
7
Interconversion of ribose 5-P and other sugar phosphates
O
OH
HO
CH
2
HO
O
P
O
O O
P
O
O
O CH
2
O
N
N
O
O
H
OH
OH OH

Uridine diphosphate d-glucose


N
HO
OH
HO
H
HO
Castanospermine
1
2
3
5
6
7
8

Many steps
G
G
G
M
M
M
M
M
Gn
Gn
Asn
M M
M M
M
M
M
Gn
Gn
Asn
Gn Gn
Gn
Gal Gal
SA SA
Gal
SA
Fuc
Trimming of
glycoproteins is
one function of
the Golgi
Ch 20 IND page-E- 2/14/03, 11:28 AM 1129
1130 Chapter 20. Some Pathways of Carbohydrate Metabolism
is a central part of the pentose phosphate pathway
(Fig. 17-8). Free ribose can be phosphorylated by a
ribokinase.
8
Oxidation of UDP-glucose in two steps
9,9a
by NAD
+
yields UDP-glucuronic acid, which can be epimerized
to UDP-galacturonic acid. Likewise (see bottom of
Fig. 20-1), guanosine disphosphate-mannose (GDP-
mannose) is oxidized to GDP-mannuronic acid, which
undergoes 4-epimerization to GDP-guluronic acid.
Looking again at the top of the
scheme, notice that UDP-D-gluc-
uronic acid may be epimerized at
the 5 position to UDP-L-iduronic
acid. However, the iduronic acid
residues in dermatan sulfate arise
by inversion at C-5 of D-glucuronic
acid residues in the polymer.
10
The mechanism of these reactions,
like that of the decarboxylation
of UDP-glucuronic acid to UDP-
xylose (near the top of Fig. 20-1),
apparently have not been well
investigated.
Notice that glucuronic acid is
abbreviated GlcA, in accord with
IUB recommendations. However,
many authors use GlcUA, ManUA,
etc., for the uronic acids.
1. The Metabolism of
Galactose
The reactions of galactose have
attracted biochemists interest be-
cause of the occurrence of the rare
(30 cases / million births) hereditary
disorder galactosemia. When this
defect is present, the body cannot
transform galactose into glucose
metabolites but reduces it to the
sugar alcohol galactitol or oxidizes
it to galactonate, both products
being excreted in the urine. Unfor-
tunately, severe gastrointestinal
troubles often appear within a few
days or weeks of birth. Growth is
slow and cataracts develop in the
eyes, probably as a result of the
accumulating galactitol. Death may
come quickly from liver damage.
Fortunately, galactose-free diets can
be prepared for young infants, and
if the disease is diagnosed promptly
the most serious damage can be
avoided. However, it has not been
possible to prevent long-term effects
that include speech difficulties, learning disabilities,
and ovarian dysfunction.
1,11
In some less seriously affected galactosemic patients
galactokinase (Eq. 20-1, step a) is absent, but it is more
often galactose-1-phosphate uridyltransferase
(Eq. 20-1, step b) that is missing or inactive.
1215a
This
enzyme transforms galactose 1-P to UDP-galactose by
displacing glucose 1-P from UDP-glucose. The UDP-
galactose is then isomerized by the NAD
+
-dependent
Figure 20-1 Some routes of interconversion of monosaccharides and of
polymerization of the activated glycosyl units.
Typical end products
Pectin
Dermatan, heparin, heparan sulfate
Chondroitin, hyaluronan, glucuronides
Plant gums
Glycogen
Lactose (milk)
Cellulose
Starch
O Antigens
Inulin (polyfructose),
bacterial dextrans, fructans
Bacterial peptidoglycans
Chitin, hyaluronan, keratan,
heparan sulfate
Chondroitin, pectin, dermatan
Colominic acid (E. coli polyNeuNAc),
sialic acids, mucoproteins, glycolipids
UDP-l-iduronic
acid
UDP-GalA
UDP-GlcA
4-Epimerase
5-Epimerase
CO
2
4-Epimerase
UDP-l-Ara
UDP-Glc
NAD
+
4-Epimerase
UDP-Gal
Glc 1-P
UTP
PP
i
ADP-Glc
dTDP-Glc
dTDP-Rha
Inositol-P
Glc 6-P
Sucrose-P Sucrose
UDP-MurNAc
UDP-GlcNAc
UDP-GalNAc
CMP-NeurNAc
Fru 6-P
GlcNAc 6-P
Mannose 6-P
Man 1-P
Xylan (wood) UDP-Xyl
CO
2
UDP-apiose Rhamnogalacturonan
GDP-l-Gal Ascorbic acid (plants)
Mannans
Alginic acid
Alginic acid (minor component)
GDP-Man
GDP-ManA
GDP-guluronic acid
(GDP-GulA)
Blood group substances GDP-l-Fuc
Ch 20 IND page-E- 2/14/03, 11:28 AM 1130
1131 A. Interconversions of Monosaccharides
BOX 20-A FRUCTOSE FOR SPERM CELLS VIA THE POLYOL PATHWAY
a
McGilvery, R. W. (1970) Biochemistry, A Functional Approach,
Saunders, Philadelphia, Pennsylvania (pp. 631 632)
b
Hers, H. G. (1960) Biochim. Biophys. Acta. 37, 127
c
Gitzelmann, R., Steinmann, B., and Van den Berghe, G. (1995)
in The Metabolic and Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S., and Valle, D.,
eds), pp. 905 935, McGraw-Hill, New York
d
Prendergast, F. G., Veneziale, C. M., and Deering, N. G. (1975)
J. Biol. Chem. 250, 1282 1289
e
Luque, T., Hjelmqvist, L., Marfany, G., Danielsson, O., El-
Ahmad, M., Persson, B., Jrnvall, H., and Gonzlez-Duarte, R.
(1998) J. Biol. Chem. 273, 34293 34301
f
Podlasek, C. A., and Serianni, A. S. (1994) J. Biol. Chem. 269,
2521 2528
g
Lee, A. Y. W., and Chung, S. S. M. (1999) FASEB J. 13, 23 30
h
Srivastava, S., Watowich, S. J., Petrash, J. M., Srivastava, S. K.,
and Bhatnagar, A. (1999) Biochemistry 38, 42 54
An interesting example of the way in which the
high [NADPH]/ [NADP
+
] and [NAD
+
]/ [NADH]
ratios in cells can be used to advantage is found in
the metabolism of sperm cells. Whereas D-glucose
is the commonest sugar used as an energy source
by mammalian cells, spermatozoa use principally
D-fructose, a sugar that is not readily metabolized
by cells of surrounding tissues.
a c
Fructose, which
is present in human semen at a concentration of 12
mM, is made from glucose by cells of the seminal
vesicle by reduction with NADPH to the sugar
alcohol D-sorbitol, which in turn is oxidized in the
2 position by NAD
+
. The combination of high
[NADPH]/ [NADP
+
] and high [NAD
+
]/ [NADH]
ratio is sufficient to shift the equilibrium far toward
fructose formation.
d
The polyol pathway is an active bypass of the
dominant glycolysis pathway in many organisms.
e
Sorbitol and other polyols such as glycerol, erythritol,
C
C OH
R
H
O
H
d-Glucose
CH
2
OH
C OH
R
H
d-Sorbitol
(d-Glucitol)
CH
2
OH
C O
R
d-Fructose
NADPH + H
+
NADP
+
NAD
+
NADH + H
+
threitol, and ribitol serve as cryoprotectants in
plants, insects, and other organisms.
f
Sorbitol is
also an important osmolyte in some organisms
(see Box 20-C). On the other hand, accumulation
of sorbitol in lenses of diabetic individuals has often
been blamed for development of cataract. However,
doubts have been raised about this conclusion. The
polyol pathway is more active than normal in dia-
betes, and there is evidence that the increased flow
in this pathway may lead to an increase in oxidative
damage to the lens. This may result, in part, from
the depletion of NADPH needed for reduction of
oxidized glutathione in the antioxidant system.
g
Aldose reductase inhibitors, which reduce the rate
of sorbitol formation, decrease cataract formation.
However, the reason for this is not yet clear.
h
UDP-Gal 4-epimerase
1616b
(Eq. 20-1, step c; see also
Eq. 15-13 and accompanying discussion). Absence of
this enzyme also causes galactosemia.
11
The overall
effect of the reactions of Eq. 20-1 is to transform galac-
tose into glucose 1-P. At the same time, the 4-epimer-
ase can operate in the reverse direction to convert
UDP-glucose to UDP-galactose, when the latter is
needed for biosynthesis (Fig. 20-1).
Another enzyme important to galactose metabo-
lism, at least in E. coli, is galactose mutarotase.
17
Cleavage of lactose by -galactosidase produces -D-
galactose which must be converted to the -anomer by
the mutarotase before it can be acted upon by galac-
tokinase. Galactose is present in most glycoproteins
and glycolipids in the pyranose ring form. However,
in bacterial O-antigens, in cell walls of mycobacteria
and fungi, and in some protozoa glactose occurs in the
furanose form. The precursor is UDP-Galf, which is
formed from UDP-Galp by UDP-Galp mutase.
17a
2. Inositol
Related to the monosaccharides is the hexahydroxy-
cyclohexane myo-inositol (Eq. 20-2). This cyclitol,
which is apparently present universally within cells
(Fig. 11-9), can be formed from glucose-6-P according
to Eq. 20-2 using a synthase that contains bound
a
b c
Gal-1-P
UDP-Glc
UDP-Gal
4-epimerase
Glc-1-P
Further metabolism
Gal
ATP
Galactokinase
Gal-1-P-
uridyl-
transferase
UDP-Gal
(20-1)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1131
1132 Chapter 20. Some Pathways of Carbohydrate Metabolism
NAD
+
. In addition to the two redox steps (Eq. 20-2, b
and d), this enzyme catalyzes both the conversion of
the anomer of glucose 6-P to the open-chain aldehyde
form and the internal aldol condensation of Eq. 20-2,
step c.
18-19b
The pro-R hydrogen at C-6 of glucose 1-P
is lost in step b while the pro-S hydrogen is retained.
20
The ring numbering system is different for glucose
and for the inositols, C-5 of glucose 1-P becoming C-2
of L-myo-inositol. Since myo-inositol contains a plane
of symmetry D- and L- forms are identical. However,
they are numbered differently (Eq. 20-2). The phospho-
inositides and inositol polyphosphates are customarily
numbered as derivatives of D-myo-inositol.
Synthesis of inositol by animals is limited and
myo-inositol is sometimes classified as a vitamin. Mice
grow poorly and lose some of their hair if deprived of
dietary inositol. Various phosphate esters of inositol
occur in nature. For example, large amounts of the
hexaphosphate (phytic acid) are present in grains,
usually as the calcium or mixed Ca
2+
Mg
2+
salts known
as phytin. The two apical cells of the 28-cell larvae
of mesozoa (Fig. 1-12A) contain enough magnesium
phytate in granular form to account for up to half of
the weight of the larvae.
21
Inositol pentaphosphate
is an allosteric activator for hemoglobin in birds and
turtles (p. 358). Di-myo-inositol-1,1'-phosphate is an
osmolyte in some hypothermophilic archaea.
19a
Ino-
sitol is a component of galactinol, the glycoside of
D-galactose with inositol (Eq. 20-15). Galactinol, as
well as free inositol, circulates in human blood and in
plants and may be a precursor of cell wall polysaccha-
rides. However, in our own bodies the greatest impor-
tance for inositol doubtless lies in the inositol-containing
phospholipids known as phosphoinositides (Figs.
8-2, 11-9, 21-5). Their function in
generation of second messengers
for various hormones is dealt with
in Chapters 11 and 21.
A person typically ingests daily
about one gram of inositol, some
in the free form, some as phospho-
inositides, and some as phytin. As
much as four grams of inositol per
day may be synthesized in the kid-
neys.
22
Breast milk is rich in inositol
and dietary supplementation with
inositol has increased survival of
premature infants with respiratory
distress syndrome.
22
The action of
insulin is reported to be improved
by administration of D-chiro-inositol
(p. 998) to women with polycystic
ovary syndrome.
22a
3. D-Glucuronic Acid,
Ascorbic Acid, and Xylitol
In bacteria, as well as in animal
kidneys,
23
inositol may be converted
to D-glucuronic acid (Fig. 20-1) with
the aid of an oxygenase. Free glu-
curonic acid may also be formed
by animals from glucose or from
UDP-glucose (Fig. 20-2). Within
the animal body glucuronic acid
can be reduced with NADH (Fig.
20-2, step a) to yield L-gulonic acid,
an aldonic acid that could also be
formed by oxidation at the alde-
hyde end of the sugar gulose.
Because C-6 of the glucuronic acid
has become C-1 of gulonic acid,
the latter belongs to the L family of
HO
HO
OH
OH
O-P
OH
1
2
5
6
H
OH
HO
HO
HO
1
2
5
6
O
3
4
OH
P
OH
HO
HO
HO
1
2
5
6
O
3
4
OH
P
-d-Glucopyranose 6-P b
a
d
e
c
Aldol
condensation
NAD
+
NADH
+
H
+

l-myo-Inositol 1-phosphate
d-myo-Inositol 3-phosphate
myo-Inositol
P
i
H
2
O
HO
C
HO
H
OH
H
O
H
R
H
O-P
OH
5
1
HO
O
HO
OH
OH
O-P
HO
HO
OH
O
2
O-P
OH
H
1 3
4
5
6
HO
C
CH
2
HO
OH
H
O
O
O-P
(20-2)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1132
1133 A. Interconversions of Monosaccharides
Figure 20-2 Some pathways of metabolism of D-glucuronic acid and of ascorbic acid, vitamin C.
HO
O
HO
COO

OH
O P P-Uridine
O
O
OH
H
O
OH H
HOH
2
C
UDPglucose
NAD
+
(2-step oxidation)
Incorporation into
polysaccharides
Glucose (animals)
O
2
(bacteria)
myo-Inositol
Glucose 6-P
NADH
d-Glucuronic acid
l-Gulonic acid
Idiopathic
pentosuria
NADPH
l-Xylulose
Xylitol
NAD
+
d-Xylulose
ATP
d-Xylulose 5-phosphate
pentose
phosphate
pathway
l-Bisoxogulonic acid
CO
2
H
2
O
CO
2 2 H
O
O O
H
O
OH H
HOH
2
C
Dehydroascorbic acid
O
OH OH
H
O
OH H
HOH
2
C
l-Ascorbic acid
C
O
OH OH
H
O
OH H
HOH
2
C
l-Gulonolactone
block in
human
2 H
?
Enolization
a
b
c
d
e
CH
2
OH
C
CH
C
CH
2
OH
OH H
HO
O
CH
2
OH
C
C
C
CH
2
OH
OH H
HO
OH
H
H
CH
2
OH
C
CH
C
CH
2
OH
O
HO
OH H
CH
2
OH
C
C
C
C
OH
HO
OH
H
H
COOH
OH H
H
CH
2
OH
C
CH
C
CHO
OH H
HO
O
CHO
C
C
C
C
OH H
HO
OH
H
H
COOH
H OH
CH
2
OH
C
CH
C
C
OH H
HO
O
COOH
O
f
g
i
j
k
h
NADP
+
UDP-glucuronate acid
CO
2
UDP-xylose
Plants
Ch 20 IND page-E- 2/14/03, 11:28 AM 1133
1134 Chapter 20. Some Pathways of Carbohydrate Metabolism
sugars. Gulonic acid can be converted to a cyclic
lactone (step b) which, in a two-step process involving
dehydrogenation and enolization (steps c and d), is
converted to L-ascorbic acid. This occurs in most
higher animals.
24
However, the dehydrogenation step
is lacking in human beings and other primates, in the
guinea pig, and in a few other species. One might say
that we and the guinea pig have a genetic defect at
this point which obliges us to eat relatively large quan-
tities of plant materials to satisfy our bodily needs for
ascorbic acid (see Box 18-D). Gulonolactone oxidase
is one of the enzymes containing covalently bound
8-(N
1
-histidyl)riboflavin.
25
The defective human
gene for this enzyme has been identified, isolated,
and sequenced. It is found to have accumulated a
large number of mutations, which have rendered it
inactive and now only a pseudogene.
26
Mice with
an inactivated gulonolactone oxidase have a dietary
requirement for vitamin C similar to that of humans.
They suffer severe vascular damage on diets marginal
in ascorbic acid.
26a
Even in rodents Na
+
-dependent
ascorbic acid transporters are present in metabolically
active tissues to bring the vitamin from the blood into
cells.
26b
A clever bit of genetic engineering has permitted
the conversion of D-glucose to 2-oxo-L-gulonate in the
enzymatic sequence of Eq. 20-3, a, b.
The bacterium Erwinia herbicola naturally has the ability
to oxidize glucose to 2,5-bisoxo-D-gluconate (Eq. 20-3,
step a) but cannot carry out the next step, the stereo-
specific reduction to 2-oxo-L-gulonate. However, a
gene encoding a suitable reductase was isolated from
a genomic library from Corynebacterium. The cloned
gene was fused to an E. coli trp promoter (see Chapter
28) and was introduced in a multicopy plasmid into
E. herbicola. The resultant organism can carry out
both steps a and b of Eq. 20-3 leaving only step c, a
nonenzymatic acid-catalyzed reaction, to complete
an efficient synthesis of vitamin C from glucose.
27
Higher plants make large amounts of L-ascorbate,
which in leaves may account for 10% of the soluble
carbohydrate content.
28
However, the pathway
of synthesis differs from that in Fig. 20-2. Both
D-mannose and L-galactose are efficient precursors.
The pathway in Eq. 20-4, which starts with GDP-D-
mannose and utilizes known enzymatic processes,
has been suggested.
28,29
The GDP-D-mannose-3,
5-epimerase is a well documented but poorly under-
stood enzyme. Multistep mechanisms related to that
of UDP-glucose 4-epimerase (Eqs. 20-1, 15-14) can be
envisioned.
Ascorbic acid is readily oxidized to dehydro-
ascorbic acid (Box 18-D; Fig. 20-2, step e), which may
be hydrolyzed to L-bisoxogulonic acid (step f ). The
latter, after decarboxylation and reduction, is converted
to L-xylulose (steps g and h), a compound that can also
be formed by a standard oxidation and decarboxylation
sequence on L-gulonic acid (step i). Reduction of
xylulose to xylitol and oxidation of the latter with
NAD
+
(steps j and k) produces D-xylulose, which can
C
C H OH
C HO H
C OH H
C H OH
CH
2
OH
H O
C
C O
C HO H
C OH H
C O
CH
2
OH
O O
C
C
C HO H
HC OH
C H
CH
2
OH
O O
O
HO
C
O
C
O
HO
HO
C
CH
2
OH
OH
H
H
l-Ascorbic acid
d-Glucose

2,5-Bisoxo-d-gluconate

2-Oxo-l-gulonate
a
b
c
(20-3)
O
HOCH
2
O
HO
HO
HO
GDP
O
O
OH
HO
OH
GDP
CH
2
5
3
GDP-d-Mannose
GDP-d-Mannose-3,5-epimerase
O
OH
HO
O
HO
H
HOCH
2
l-Gal-1-P
GDP-l-Galactose
H
2
O
P
i
l-Gal
NAD
+
l-Ascorbic acid
2H
+
+ 2e

H
2
O
GMP
HO
(20-4)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1134
1135
be phosphorylated with ATP and enter the pentose
phosphate pathways. A metabolic variation produces
a condition called idiopathic pentosuria. Affected
individuals cannot reduce xylulose to xylitol and,
hence, excrete large amounts of the pentose into the
urine, especially if the diet is rich in glucuronic acid.
The defect seems to be harmless, but the sugar in
the urine can cause the condition to be mistaken for
diabetes mellitus.
30
Xylitol is as sweet as sucrose and has been used as
a food additive. Because it does not induce formation
of dental plaque, it is used as a replacement for sucrose
in chewing gum. It appeared to be an ideal sugar
substitute for diabetics. However, despite the fact that
it is already naturally present in the body, ingestion of
large amounts of xylitol causes bladder tumors as well
as oxalate stones in rats and mice. Its use has, there-
fore, been largely discontinued. A possible source of
the problem may lie in the conversion by fructokinase
of some of the xylitol to D-xylulose 1-P, which can be
cleaved by the xylulose 1-P aldolase to dihydroxyace-
tone P and glycolaldehyde.
The latter can be oxidized to oxalate and may also be
carcinogenic. As indicated in the upper left corner of
Fig. 20-2, UDP-glucuronate can be decarboxylated to
UDP-xylose.
4. Transformations of Fructose 6-Phosphate
Biosynthesis of D-glucosamine 6-phosphate is
accomplished by reaction of fructose 6-P with gluta-
mine (Eq. 20-5):
Glutamine is one of the principal combined forms of
ammonia that is transported throughout the body
(Chapter 24). Glucosamine 6-phosphate synthase,
which catalyzes the reaction of Eq. 20-5, is an amido-
transferase of the N-terminal nucleophile hydrolase
superfamily (Chapter 12).
31
It hydrolyzes the amide
A. Interconversions of Monosaccharides
C CH
2
OH
O
H
Glycolaldehyde
CH
2
OH
C O
CH
COH
COH
CH
2
O
CHO
C NH
2
CH
COH
COH
CH
2
O
HO
H
H
Fructose 6-P

HO
H
H
d-Glucosamine 6-P

H
Gln Glu
(20-5)
linkage of glutamine. The released ammonia pre-
sumably reacts with the carbonyl group of fructose
6-P to form an imine,
32 34a
which then undergoes
a reaction analogous to that catalyzed by sugar
isomerases.
35
The resulting D-glucosamine 6-P is
acetylated on its amino group by transfer of an acetyl
group,
36
and a mutase moves the phospho group to
formN-acetylglucosamine 1-P. In E. coli acetylation
occurs on GlcN 1-P and is catalyzed by a bifunctional
enzyme that also has mutase activity.
37-37b
The result-
ing N-acetylglucosamine 1-P is converted to UDP-N-
acetylglucosamine (UDP-GlcNAc) with cleavage of
UTP to inorganic pyrophosphate as in the synthesis of
UDP-glucose (Eq. 17-56). Cells of E. coli are also able
to catabolize glucosamine 6-phosphate. A deaminase,
with many properties similar to those of GlcN 6-P
synthase, catalyzes a reaction resembling the reverse
of Eq. 20-5 but releasing NH
3
.
38,39
One of the compounds formed from UDP-GlcNAc
is UDP-N-acetylmuramic acid. The initial step in its
synthesis is an unusual type of displacement reaction
on the -carbon of PEP by the 3-hydroxyl group of the
sugar (Eq. 20-6, step a).
40-41c
Inorganic phosphate is
a
b
O
HO
CH
2
NH
OH
HO
O P P
C
CH
3
O
Uridine
O
O
CH
2
NH
OH
HO
O P P
C
CH
3
O
Uridine
C H
2
C
COO

O
O
NH
O P P
C
CH
3
O
Uridine
C
COO

H
3
C
H
UDP-GlcNAc
P
i
NADPH + H
+
UDP-N-acetylmuramic acid
(UDP-MurNAc)
NADP
+
Flavoprotein
PEP
O C
CH
2
COO P

(20-6)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1135
1136 Chapter 20. Some Pathways of Carbohydrate Metabolism
displaced with formation of an enolpyruvyl derivative
of UDP-GlcNAc. This derivative is then reduced by
NADPH (Eq. 20-6, step b).
42-43a
A second sugar nucle-
otide formed from UDP-GlcNAc is UDP-N-acetyl-
galactosamine (UDP-GalNAc), which may be created
by the same 4-epimerase that generates UDP-Gal (Eq.
20-1).
44
Some animal tissues such as kidney and liver
also have a GalNAc kinase that may salvage, for
reuse, GalNAc that arises from the degradation of
complex polysaccharides.
44
Bacteria may dehydroge-
nate UDP-GalNAc to UDP-N-acetylgalactosaminuric
acid (UDP-GalNAcA).
44a
UDP-GlcNAc can be converted to UDP-N-acetyl-
mannosamine (UDP-ManNAc) with concurrent elimi-
nation of UDP (Eq. 20-7).
45 47b
This unusual epimeri-
zation occurs without creation of an adjacent carbonyl
group that would activate the 2-H for removal as a
proton. As indicated by the small arrows in Eq. 20-7,
step a, the UDP is evidently eliminated. In a bacterial
enzyme it remains in the ES complex and is returned
after a conformational change involving the acetamido
group. This allows the transient C1C2 double bond
to be protonated from the opposite side (Eq. 20-7,
step a).
47
In bacteria the UDP-ManNAc may be dehy-
drogenated to UDP-N-acetylmannos-aminuronic acid
(ManNAcA). Both ManNAc and ManNAcA are com-
ponents of bacterial capsules.
47
In mammals the epimerase (Eq. 20-7, step a)
probably utilizes a similar chemical mechanism but
eliminates UDP and replaces it with HO

to give
free N-acetylmannosamine, which is then phosphor-
ylated on the 6-hydroxyl (Eq. 20-7, step b). ManNAc
may also be formed from free GlcNAc by another
2-epimerase (step a).
47c,d
5. Extending a Sugar Chain with
Phosphoenolpyruvate (PEP)
The six-carbon chain of ManNAc 6-P can be ex-
tended by three carbon atoms using an aldol-type
condensation with a three-carbon fragment from PEP
(Eq. 20-7, step c) to give N-acetylneuraminic acid
(sialic acid).
48
The nine-carbon chain of this molecule
can cyclize to form a pair of anomers with 6-membered
rings as shown in Eq. 20-7. In a similar manner, arabi-
nose 5-P is converted to the 8-carbon 3-deoxy-D-
manno-octulosonic acid (KDO) (Fig. 4-15), a com-
ponent of the lipopolysaccharide of gram-negative
bacteria (Fig. 8-30), and D-Erythrose 4-P is converted
to 3-deoxy-D-arabino-heptulosonate 7-P, the first meta-
bolite in the shikimate pathway of aromatic synthesis
(Fig. 25-1).
48a
The arabinose-P used for KDO synthesis
is formed by isomerization of D-ribulose 5-P from the
pentose phosphate pathway, and erythrose 4-P arises
from the same pathway.
The mechanism of the aldol condenstion that
O
HO
H
HO
O
P P Uridine
HN
C
O
CH
3
O
HO
CH
2
NH
HO
OH
H
O
P P
C O
CH
3
+
Uridine
H
C
C H N
C H HO
C OH H
C OH H
CH
2
O P
C
H
3
C
O
OH H
CH
2
C
COO
O
H

O
OH
OH
C
COO
H
N
C
H
HO

H
3
C
O P
C
H
3
C
O
H
H
OH
O
C
C
H
H N
C H HO
C OH H
C OH H
CH
2
O P
C
H
3
C
O
H
N-Acetylmannosamine 6-P
(ManNAc 6-P)
PEP
O C
CH
2
COO P

c
P
i
N-Acetylneur-
aminic acid 9-P
(Neu5Ac 9-P)
b
ADP
ATP
O
HO
O
C
COO
N
C
H
HO

H
3
C
HO
C
H
3
C
O
H
H
OH P
O
O O
Cytidine

N-acetyl-
glucosamine
(GlcNAc)
a
a
UDP-GlcNAC
N-Acetylmannosamine
(ManNAc)
H
2
O
P
i
N-Acetylneuraminic acid (Neu5Ac)
d
PP
i
CTP
e
CMP-Neu5Ac
NAD
+
UDP-ManNAc
Polysaccharides
N-Acetylmannos-
aminuronic acid
(ManNAcA)
H
2
O
UDP
a
H
2
O
UDP
(20-7)
forms these sugars is somewhat unexpected. A reac-
tive enolate anion can be formed from PEP by hydro-
lytic attack on the phospho group with cleavage of
the O P bond. However, in reactions such as step a of
Ch 20 IND page-E- 2/14/03, 11:28 AM 1136
1137
Eq. 20-6, step c of Eq. 20-7,and also in EPSP synthase
(Eq. 25-4) the initial condensation does not involve
O P cleavage. NMR studies of the action of KDO
synthase reveal that the C O bond of PEP is cleaved
as is indicated in Eq. 20-8.
49-52b
The si face of PEP faces
the re face of the carbonyl group of the sugar phosphate.
A carbanionic center is generated at C-3 of PEP with
possible participation of the phosphate oxygen as well
as electrostatic stabilization of the carbocation formed
in step a. Ring closure (step b) occurs with loss of P
i
.
The immediate product of the aldol condensation, in
Eq. 20-7, is N-acetylneuraminic acid 9-phosphate,
which is cleaved through phosphatase action (step d)
and is activated to the CMP derivative by reaction
with CTP (Eq. 20-7,step e).
52c
Further alterations may
occur. For example, CMP-Neu5Ac is hydroxylated to
form CMP-N-glycolylneuraminic acid.
53
Furthermore,
an additional type of sialic acid, 2-oxo-3-deoxy-D-
O
HO
H
b
OH
OH
HO O
COO
H
a
P
H
C
OH
O
HO
COO
HO O P
O
P
C
H
a
H
b

C
OH
HO
COO
HO O P
OP C

HO
HO

H
H
+
a
b
P
i
Arabinose 5-P
PEP
KDO-8-P (20-8)
glycero-D-galacto-nononic acid (KDN), has been found
in human developmentally regulated glycoproteins
and also in many other organisms.
54-55a
It has an OH
group in the 5-position rather than the acetamido
group of the other sialic acids. Like NeuNAc it is
activated by reaction with CTP forming CMP-KDN.
These activated monosaccharides differ from most
others in being derivatives of a CMP rather than of
CDP. More than 40 different naturally occuring varia-
tions of sialic acid have been identified.
55b
In a similar fashion, KDO is converted to the
-linked CMP-KDO,
56-56b
which is incorporated into
lipid A as shown in Fig. 20-10. The ADP derivative
of the L-glycero-D-manno-heptose (Fig. 4-15), which
is also present in the lipopolysaccharide of gram-
negative bacteria, is formed from sedoheptulose 7-P
in a five-step process (Eq. 20-9).
57-58b
6. Synthesis of Deoxy Sugars
Metabolism of sugars often involves dehydration
to ,-unsaturated carbonyl compounds. An example
is the formation of 2-oxo-3-deoxy derivatives of sugar
acids (Eq. 14-36). Sometimes a carbonyl group is
O
HO
HO
O
HO
O
OH
P P Adenosine
H
6
CH
2
OH
C O
C
C
H HO
C
C
OH H
OH H
OH H
CH
2
O P
Sedoheptulose 7-P
Isomerization,
cylization
O
HO
HO
OP
HO
OH
OH
D-glycero-D-mannoheptose 7-P
Mutase
ATP
PP
i
6-epimerase (NAD
+
-dependent)
(20-9)
A. Interconversions of Monosaccharides
O
OH
HO
HO
OH
O dTDP
O
OH
CH
2
HO
OH
O dTDP
O
H
O
OH
CH
3
HO
O dTDP
O
H
3
5
O
OH
CH
2
HO
O dTDP
O
O
OH
H
O dTDP
CH
3
OH
HO
O
HO
OH
H
3
C O
HO
dTDP
b
H
2
O
l-Rhamnose
c
NADH
d
a
NAD
+
e
4
6
(20-10)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1137
1138 Chapter 20. Some Pathways of Carbohydrate Metabolism
created by oxidation of an OH group, apparently for
the sole purpose of promoting dehydration. For exam-
ple, the biosynthesis of L-rhamnose from D-glucose is a
multistep process (Eq. 20-10) that takes place while the
sugars are attached to deoxythymidine diphosphate.
59,59a,b
Introduction of the carbonyl group by dehydrogena-
tion with tightly bound NAD
+
(Eq. 20-10, step a) is
followed by dehydration (step b).
59c,d
To complete the
sequence, the double bond formed by dehydration is
reduced (step c) by the NADH produced in step a.
A separate enzyme, a 3,5-epimerase catalyzes inver-
sion at both C-3 and C-5 (step d).
59e
Finally, a third en-
zyme is needed for a second reduction (step e) using
NADPH.
59f
The biosynthesis of GDP-L-fucose from
GDP-D-mannose occurs by a parallel sequence.
60-61b
The metabolism of free L-fucose (6-deoxy-L-galac-
tose), which is present in the diet and is also generated
by degradation of glycoproteins, resembles the Entner
Doudoroff pathway of glucose metabolism (Eq. 17-18).
Similar degradative pathways act on D-arabinose and
L-galactose.
60
Bacterial surface polysaccharides contain a vari-
ety of dideoxy sugars. The four 3,6-dideoxy sugars
D-paratose (3,6-dideoxy-D-glucose), D-abequose
(3,6-dideoxy-D-galactose), D-tyvelose (3,6-dideoxy-D-
mannose), and L-ascarylose (3,6-dideoxy-L-mannose),
whose structures are shown in Fig. 4-15, arise from
CDP-glucose.
60a
This substrate is first converted, in
reactions parallel to the first three steps of Eq. 20-10, to
4-oxo-6-deoxy-CDP-glucose which reacts in two steps
with pyridoxamine 5'-phosphate (PMP) and NADH
(Eq. 20-11). This unusual reaction
62 65
is catalyzed by a
two-enzyme complex. The first component, E1, cata-
lyzes the formation of a Schiff base of the substrate
with PMP and a transamination, which also accom-
plishes dehydration, to give an unsaturated sugar ring
(Eq. 20-11, step a). The protein also contains an Fe
2
S
2
center suggesting a possible one-electron transfer. The
second component, E
3,
contains both an Fe
2
S
2
plant
type ferredoxin center and bound FAD.
65
Observation
by EPR spectroscopy revealed accumu-
lation of an organic free radical
64
that
may be an intermediate in step b of Eq.
20-11. Hydrolysis, epimerization at C-
5, and reduction yields L-ascarylose.
A similar reaction sequence without
the last epimerization would yield
D-abequose. CDP-D-tyvelose arises by
C-2 epimerization of CDP-D-paratose.
65a
Other unusual sugars
66 68
are formed
from intermediates in Eq. 20-11. One
is a 3-amino-3,4,6-trideoxyhexose
in which the amino group has been
provided by transamination
67
(see also
Box 20-B).
The unusual sulfur-containing
sugar 6-sulfoquinovose is present in
O
HO
CH
3
OH
N
O CMP
HN C
H
3
C O
OP
H
R
H
S

H
+
+
O
CH
3
OH
N
O CMP
HN
H
3
C O

+
OP
H
+
H
H
O
HO
CH
3
OH
O
O CMP
O
H
CH
3
OH
N
O CMP
HN
H
3
C O

+
OP
H
+
H 3
a
b
PMP
CMP-d-glucose
H
2
O
E1
E3
2H
+
+ 2e

via (Fe
2
S
2
), FAD
H
2
O
Epimerase
Reduction
Reduction
l-Ascarylose d-Abequose
(20-11)
COO
SO
3
H
3
N
H
+

Cysteic acid
C
C
SO
3
H
HO

Sulfolactaldehyde
O
H
C
C OH
H O
C H HO
C H OH
C OH H
CH
2
SO
3
6-Sulfoquinovose
H

Transamination
Reduction
a
O
O
O
O
HO
UDP
S O
H
3
C
O O
H

NADH
b
H
2
O
UDP-Glc
NAD
+
P
i
H
2
O
Aldol
Dihydroxyacetone-P
(20-12)
Ch 20 IND page-E- 2/14/03, 11:28 AM 1138
1139
BOX 20-B THE BIOSYNTHESIS OF STREPTOMYCIN
Streptomycin, the kanamycins, neomycins,
and gentamycins form a family of medically impor-
tant aminoglycoside antibiotics.
a
They are all
water-soluble basic carbohydrates containing three
or four unusual sugar rings. D-Glucose is a precur-
sor of streptomycin, all three rings being derived
from it. While the route of biosynthesis of 2-deoxy-2-
methylamino-L-glucose is not entirely clear, the
pathways to L-streptose and streptidine, the other
two rings, have been characterized.
b d
The starting
material for streptidine synthesis is a nucleoside
diphosphate sugar, which is an intermediate in the
synthesis of L-rhamnose (Eq. 20-10). The carbon
carbon chain undergoes an aldol cleavage as shown
in step a of the following equation:
OH
O
HN
NH
HO
O
C
H
3
C
O
OH
HO
CH
2
OH
NH H
3
C
O
HO
O
H
C NH
2
H
2
N
C
NH
2
NH
2
+
+
Streptidine
Streptose
2-Deoxy-2-methylamino-l-glucose
Streptomycin
Reduced in
dihydro-
streptomycin
Site of enzymatic
adenylylation or
phosphorylation
O
OH
CH
3
HO
dTDP
O
O
C
dTDP
C
HC
HO
O
O
CH
3
O
OH OH
H
3
C
CHO
Intermediate from Eq. 20-10
H
Streptosyl-TDP
H
a
b
OH
HO
HO
O
OH
NH
2 P
c
d
OH
HO
HO
HO
OH
OH
myo-Inositol
OH
HO
HO
HO
OH
NH
2
2H
Transamination
Gln
Oxoglutaramate
ATP
e
f
Arg
Orn
P
i
OH
HO
HO
HO
OH
N
C
NH
2
NH
2
+
H
Oxidation here next
The ring-open product is written
here as an enediol, which is able to
recyclize in an aldol condensation
(step b) to form a five-membered
ring with a branch at C-3. The L-
streptosyl nucleoside diphosphate
formed in this way serves as the
donor of streptose to streptomycin.
The basic cyclitol streptidine is
derived from myo-inositol, which
has been formed from glucose 6-P
(Eq. 20-2). The guanidino groups
are introduced by oxidation of the
appropriate hydroxyl group to a
carbonyl group followed by tran-
samination from a specific amino
donor. In the first step, illustrated
by the following equation, gluta-
mine is the amino donor for the
transamination, the oxoacid prod-
uct being -oxoglutaramic acid.
A. Interconversions of Monosaccharides
Ch 20 IND page-E- 2/14/03, 11:28 AM 1139
1140 Chapter 20. Some Pathways of Carbohydrate Metabolism
the hemiacetal groups of both sugar rings are blocked.
However, sucrose is thermodynamically reactive, the
glucosyl group having a group transfer potential of
29.3 kJ mol
1
. It is extremely sensitive toward hydrol-
ysis catalyzed by acid. Transport of sugar in the form
of a disaccharide provides an advantage to plants in
that the disaccharide has a lower osmotic pressure
than would the same amount of sugar in monosaccha-
ride form.
Biosynthesis of sucrose
71,71a
utilizes both UDP-
glucose and fructose 6-P (Eq. 20-13). Reaction of UDP-
glucose with fructose can also occur to give sucrose
directly.
72
Because this reaction is reversible, sucrose
serves as a source of UDP-glucose for synthesis of
cellulose and other polysaccharides in plants. Metab-
olism of sucrose in the animal body begins with the
action of sucrase (invertase), which hydrolyzes the
disaccharide to fructose and glucose (Eq. 20-14, step a).
The same enzyme is also found in higher plants and
fungi. Mammalian sucrase is one of several carbohy-
drases that are anchored to the external surfaces of
the microvilli of the small intestines. Sucrose is bound
the sulfolipid of chloroplasts (p. 387).
69
A possible
biosynthetic sequence begins with transamination of
cysteic acid to 3-sulfopyruvate, reduction of the latter
to sulfolactaldehyde, and aldol condensation with
dihydroxyacetone-P as indicated in Eq. 20-12a.
70
See
also Eq. 24-47 and Fig. 4-4. However, biosynthesis
in chloroplasts appears to start with action of a 4,6-
dehydratase on UDP-glucose followed by addition of
sulfite and reduction (Eq. 20-12b).
70a,b
The sulfite is
formed by reduction of sulfate via adenylyl sulfate
(Fig. 24-25). However, biosynthesis in chloroplasts
appears to start with action of a 4,6-dehydratase on
UDP-glucose followed by addition of sulfite and
reduction (Eq. 20-12b).
70a,b
The sulfite is formed by
reduction of sulfate via adenylyl sulfate (Fig. 24-25).
B. Synthesis and Utilization of
Oligosaccharides
Our most common food sugar sucrose is formed
in all green plants and nowhere else. It is made both
in the chloroplasts and in the vicinity of other starch
deposits. It serves both as a transport sugar and,
dissolved within vacuoles, as an energy store. Sucrose
is very soluble in water and is chemically inert because
UDPGlc Sucrose 6P Sucrose
H
2
O
P
i
Fru-6P (20-13)
BOX 20-B THE BIOSYNTHESIS OF STREPTOMYCIN (continued)
The amino group on the ring now receives an
amidine group, which is transferred from arginine
by nucleophilic displacement
e
in a reaction resem-
bling that in the synthesis of urea (see Fig. 24-10,
step h). However, there is first a phosphorylation
at the 2 position. After the amidine transfer has
occurred to form the guanidino group, the phos-
pho group is hydrolyzed off by a phosphatase.
This is another phosphorylationdephosphorylation
sequence (p. 977) designed to drive the reaction to
completion in the desired direction. The second
guanidino group is introduced in an analogous
way by oxidation at the 3 position followed by
transamination, this time with the amino group
being donated by alanine. Again, a phosphoryla-
tion is followed by transfer of an amidine group
from arginine. The final hydrolytic removal of the
phospho group (which this time is added at C-6)
does not occur until the two other sugar rings have
been transferred on from nucleoside diphosphate
precursors to form streptomycin phosphate.
As with other antibiotics,
f i
streptomycin is
subject to inactivation by enzymes encoded by
genetic resistance factors (Chapter 26). Among
these are enzymes that transfer phospho groups
a
Benveniste, R., and Davies, J. (1973) Ann. Rev. Biochem. 42, 471
506
b
Luckner, M. (1972) Secondary Metabolism in Plants and Animals,
Academic Press, New York (pp.7880)
c
Walker, J. B., and Skorvaga, M. (1973) J. Biol. Chem. 248, 2441
2446
d
Marquet, A., Frappier, F., Guillerm, G., Azoulay, M., Florentin,
D., and Tabet, J.-C. (1993) J. Am. Chem. Soc. 115, 21392145
e
Fritsche, E., Bergner, A., Humm, A., Piepersberg, W., and
Huber, R. (1998) Biochemistry 37, 1766417672
f
Cox, J. R., and Serpersu, E. H. (1997) Biochemistry 36, 23532359
g
McKay, G. A., and Wright, G. D. (1996) Biochemistry 35, 8680
8685
h
Thompson, P. R., Hughes, D. W., Cianciotto, N. P., and Wright,
G. D. (1998) J. Biol. Chem. 273, 1478814795
i
Gerratana, G., Cleleand, W. W., and Reinhardt, L. A. (2001)
Biochemistry 40, 29642971
j
Roestamadji, J., Grapsas, I., and Mobashery, S. (1995) J. Am.
Chem. Soc. 117, 8084
k
Thompson, P. R., Hughes, D. W., and Wright, G. D. (1996)
Biochemistry 35, 86868695
or adenylyl groups onto streptomycin at the site
indicated by the arrow in the structure.
j,k
Thus,
dephosphorylation at one site generates the active
antibiotic as the final step in the biosynthesis, while
phosphorylation at another site inactives the anti-
biotic.
Ch 20 IND page-E- 2/14/03, 11:29 AM 1140
1141 B. Synthesis and Utilization of Oligosaccharides
tightly but noncovalently to isomaltase, which hydro-
lyzes the -1,6-linked isomaltose and related oligo-
saccharides. A nonpolar N-terminal segment of the
isomaltase anchors the pair of enzymes to the micro-
villus membrane. The two-protein complex arises
naturally because the two enzymes are synthesized as
a single polypeptide, which is cleaved by intestinal
proteases.
73,74
Because of the relatively high group transfer
potential of either the glucosyl or fructosyl parts,
sucrose is a substrate for glucosyltransferases such as
sucrose phosphorylase (Eq. 20-14, step b; see also Eq.
12-7 and associated discussion). In certain bacteria
this reaction makes available the activated glucose
1-P which may enter catabolic pathways directly.
Cleavage of sucrose for biosynthetic purposes can
occur by reaction 20-14, step c, which yields UDP-
glucose in a single step.
A disaccharide with many of the same proper-
ties as sucrose is trehalose, which consists of two
-glucopyranose units in 1,1 linkage (p. 168). The
biosynthetic pathway from UDP-glucose and glu-
cose 6-P parallels that for synthesis of sucrose (Eq.
20-13).
75,76
In E. coli the genes for the needed glucosyl-
transferase and phosphatase are part of a single oper-
on. Its transcription is controlled in part by glucose-
mediated catabolite repression (Chapter 28) and also
by a repressor of the Lac family.
76,76a,77
The repressor
is allosterically activated by trehalose 6-P, the inter-
mediate in the synthesis. Trehalose formation in bacte-
ria, fungi, plants, and microscopic animals is strongly
induced during conditions of high osmolality (see Box
20-C).
77
Both trehalose and maltose can also be taken
up via an ABC type transporter (p. 417).
77a,b
Lactose, the characteristic sugar of milk, is formed
by transfer of a galactosyl unit from UDP-galactose
directly to glucose (Eq. 20-15, reaction a). The similar
transfer of a galactosyl unit to N-acetylglucosamine to
formN-acetyllactosamine (Eq. 20-15, reaction b) occurs
in many animal tissues. An interesting regulatory
mechanism is involved. The transferase catalyzing Eq.
20-15, reaction b, forms a complex with -lactalbumin
to become lactose synthase,
78 80b
the enzyme that
catalyzes reaction a. Lactalbumin was identified as a
milk constituent long before its role as a regulatory
protein was recognized.
A very common biochemical problem is intoler-
a
b
d
Lactose (milk)
N-Acetyllactosamine UDP-Gal
c
Raffinose
UDP Sucrose
UDP Glc
Glc-Nac UDP
myo-Inositol
OH
HO
OH
HO
O OH
HO
OH
OH
OH
Galactinol
UDP
(Gal (1-6) Glc (1-2) Fru)
(20-15)
ance to lactose.
81
This results from the inability of the
intestinal mucosa to make enough lactase to hydro-
lyze the sugar to its monosaccharide components
galactose and glucose. Among most of the peoples of
the earth only infants have a high lactase level, and the
use of milk as a food for adults often leads to a severe
diarrhea. The same is true for most animals. In fact,
baby seals and walruses, which drink lactose-free
milk, become very ill if fed cows milk.
The plant trisaccharide raffinose arises from
UDP-galactose by transfer of a galactosyl unit onto
the 6-hydroxyl of the glucose ring of sucrose (Eq. 20-
15, reaction c). Transfer of a galactosyl unit onto myo-
inositol (Eq. 20-15, reaction d) produces galactinol,
whose occurrence is widespread within the plant
kingdom. Galactinol, in turn, can serve as a donor of
activated galactosyl groups. Thus, many plants con-
tain stachyose and higher homologs, all of which are
formed by transfer of additional -D-galactosyl units
onto the 6-hydroxyl of the galactose unit of raffinose.
These sugars appear to serve as antifreeze agents in
the plants. The concentration of stachyose in soy
beans can be as high as that of sucrose. Some seeds,
e.g., those of maize, are coated with a glassy sugar
mixture of sucrose and raffinose in a ratio of ~ 3:1.
81a
Besides the oligosaccharides, living organisms form
a great variety of glycosides that contain nonsugar
components. Among these are the glucuronides
(glucosiduronides), excretion products found in urine
and derived by displacement of UDP from UDP-
glucuronic acid by such compounds as phenol, benzo-
ic acid, and sterols.
81b,c
Phenol is converted to phenyl
glucuronide (Eq. 20-16), while benzoic acid (also ex-
creted in part as hippuric acid, Box 10-A) yields an ester
by the same type of displacement reaction. Many other
aromatic or aliphatic compounds containing OH, SH,
NH
2
, or COO

groups also form glucuronides.


82
a
b
H
2
O
Fructose + glucose
Glucose 1-P
Fructose P
i
Sucrose
c
UDP-Glc
UDP Fructose
(20-14)
Ch 20 IND page-E- 2/14/03, 11:29 AM 1141
1142 Chapter 20. Some Pathways of Carbohydrate Metabolism
BOX 20-C OSMOTIC ADAPTATION
Bacteria, plants of many kinds, and a variety of
other organisms are forced to adapt to conditions of
variable osmotic pressure.
a,b
For example, plants must
resist drought, and some must adapt to increased
salinity. Some organisms live in saturated brine
~6 M in NaCl.
c
The osmotic pressure in dilute
aqueous solutions is proportional to the total molar
concentration of solute particles, c
s
, as follows.
= RTc
s
where R is the gas constant and
T the Kelvin temperature
At higher solute concentrations c
s
must be
replaced by the effective molar concentra-
tion, which is called osmolarity (OsM)
and has units of molarity (see Record et al. for
discussion).
b
An osmolarity difference across a
membrane of 0.04 OsM results in a turgor pressure
of ~ 1 atmosphere. To adapt to changes in the
environmental osmolarity organisms must alter
their internal solute concentrations.
Cells of E. coli can adapt to at least 100-fold
changes in osmolarity. Because of the porosity of
the bacterial outer membrane the osmolarity of the
periplasmic space is normally the same as that of
the external medium. However, the inner mem-
brane is freely permeable only to water and a few
solutes such as glycerol.
b
The bacterial cells avoid
loss of water when the external osmolarity is high
by accumulating K
+
together with anions such as
glutamate

and nonionic osmoprotectants such


as trehalose, sucrose, and oligosaccharides. E. coli
cells will also take up other osmoprotectants such
as glycine betaine, dimethylglycine, choline,
proline, and proline betaine. Some methanogens
accumulate N

-acetyl--lysine as well as glycine


betaine.
d
Functioning in a somewhat different way in
E. coli are 6- to 12-residue periplasmic membrane-
derived oligosaccharides. These are -1,2- and -
1,6-linked glucans covalently linked to sn-1-phospho-
glycerol, phosphoethanolamine, or succinate (see
Fig. 8-28).
b,e,f
They accumulate in the periplasm when
cells are placed in a medium of low osmolarity.
The resulting increased turgor in the periplasm is
thought to buffer the cytoplasm against the loss of
external osmolarity and to protect the periplasmic
space from being eliminated by expansion of the
plasma membrane. Related cyclic glucans, which
are attached to sn-1-phosphoglycerol or O-succinyl
ester residues, are accumulated by rhizobia.
g
N
C
C O
O CH
3
H H
H
3
C
H
3
C
N
C
CH
3
H
3
C
H
O
O
+

N
CH
3
H
3
C
H
3
C
Betaine (glycine betaine)
+

+
Proline betaine
O
Trimethylamine
N-oxide
C
Among these is bilirubin (Fig. 24-24). UDP-glucuron-
osyltransferases responsible for their synthesis are
present in liver microsomes.
Among the many glycosides and glycosylamines
made by plants are the anthocyanin and flavonoid
pigments of flowers (Box 21-E), cyanogenic glycosides
such as amygdalin (Box 25-B), and antibiotics (e.g.,
see Box 20-B).
83,84
Some are characteristic of certain
families of plants. For example, more than 100 -
thioglucosides known as glucosinolides are found
in the Cruciferae (cabbages, mustard, rapeseed). The
compounds impart the distinctive flavors and aromas
of the plants. However, some are toxic and may cause
goiter or liver damage. The enzyme myrosinase
hydrolyzes these compounds releasing isothiocyanates,
thiocyanates, and nitriles (Eq. 20-17).
85-86a
L-Ascorbate
acts as a cofactor for this enzyme, evidently providing
a catalytic base.
86a
O
OH
HO
S
HO
OH
R
N
OSO
3

N
O
R
HS SO
3

R N C S
H
2
O
d-Glucose
Sinigrin (R=allyl)
HSO
4

Glucosinolates
(20-17)
O
OH
HO
OOC
O HO

UDP-GlcA UDP
Phenol
Phenyl glucuronide
(20-16)
Ch 20 IND page-E- 2/14/03, 11:29 AM 1142
1143
BOX 20-C (continued)
a
Le Rudulier, D., Strom, A. R., Dandekar, A. M., Smith, L. T.,
and Valentine, R. C. (1984) Science 224, 1064 1068
Fungi, green algae, and higher plants more
often accumulate glycerol,
h,i
sorbitol, sucrose,
j
trehalose,
k
or proline.
a,l,m
These compounds are
all compatible solutes which tend not to disrupt
cellular structure.
n
Betaines and proline are espe-
cially widely used by a variety or organisms. How
is it then that some desert rodents, some fishes, and
other creatures accumulate urea, a well-known
protein denaturant? The answer is that they also
accumulate methylamine or trimethylamine N-oxide
in an approximately 2:1 ratio of urea to amine. The
mixture of compounds is compatible, the stabilizing
effects of the amines offsetting the destabilizing
effect of urea.
c,o
Adaptation to changes in osmotic pressure
involves sensing and signaling pathways that have
been partially elucidated for E. coli
p
and yeasts.
i,q
Major changes in structure and metabolism may
result. For example, in E. coli the outer membrane
porin OmpF (Fig. 8-20) is replaced by OmpC
(osmoporin), which has a smaller pore.
r
Aresurrection plant that normally contains an
unusual 2-octulose converts this sugar almost entirely
into sucrose when desiccated. This is one of a small
group of plants that are able to withstand severe
desiccation but can, within a few hours, reverse the
changes when rehydrated.
j
b
Record, M. T., Jr., Courtenay, E. S., Cayley, D. S., and Guttman,
H. J. (1998) Trends Biochem. Sci. 23, 143 148
c
Yancey, P. H., Clark, M. E., Hand, S. C., Bowlus, R. D., and
Somero, G. N. (1982) Science 217, 1214 1222
d
Sowers, K. R., Robertson, D. E., Noll, D., Gunsalus, R. P., and
Roberts, M. F. (1990) Proc. Natl. Acad. Sci. U.S.A. 87, 9083 9087
e
Kennedy, E. P. (1987) in Escherichia coli and Salmonella
typhimurium (Neidhardt, F. C., ed), pp. 672 679, Am. Soc. for
Microbiology, Washington,DC
f
Fiedler, W., and Rotering, H. (1988) J. Biol. Chem. 263, 14684
14689
g
Weissborn, A. C., Rumley, M. K., and Kennedy, E. P. (1991) J.
Biol. Chem. 266, 8062 8067
h
Ben-Amotz, A., and Avron, M. (1981) Trends Biochem. Sci. 6,
297 299
i
Davenport, K. R., Sohaskey, M., Kamada, Y., Levin, D. E., and
Gustin, M. C. (1995) J. Biol. Chem. 270, 30157 30161
j
Bernacchia, G., Schwall, G., Lottspeich, F., Salamini, F., and
Bartels, D. (1995) EMBO J. 14, 610 618
k
Dijkema, C., Kester, H. C. M., and Visser, J. (1985) Proc. Natl.
Acad. Sci. U.S.A. 82, 14 18
l
Garca-Ros, M., Fujita, T., LaRosa, P. C., Locy, R. D., Clithero, J.
M., Bressan, R. A., and Csonka, L. N. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 8249 8254
m
Verbruggen, N., Hua, X.-J., May, M., and Van Montagu, M.
(1996) Proc. Natl. Acad. Sci. U.S.A. 93, 8787 8791
n
Higgins, C. F., Cairney, J., Stirling, D. A., Sutherland, L., and
Booth, I. R. (1987) Trends Biochem. Sci. 12, 339 344
o
Lin, T.-Y., and Timasheff, S. N. (1994) Biochemistry 33, 12695
12701
p
Racher, K. I., Voegele, R. T., Marchall, E. V., Culham, D. E.,
Wood, J. M., Jung, H., Bacon, M., Cairns, M. T., Ferguson, S. M.,
Liang, W.-J., Henderson, P. J. F., White, G., and Hallett, F. R.
(1999) Biochemistry 38, 1676 1684
q
Shiozaki, K., and Russell, P. (1995) EMBO J. 14, 492 502
r
Kenney, L. J., Bauer, M. D., and Silhavy, T. J. (1995) Proc. Natl.
Acad. Sci. U.S.A. 92, 8866 8870
syl units from UDP-glucose (Eq. 17-56)
87,87a
or in bacte-
ria from ADP-glucose.
88 90
Utilization of glycogen by
the cell involves removal of glucose units as glucose
1-P by the action of glycogen phosphorylase. The
combination of growth and degradation from the same
chain ends provides a means of rapidly storing and
utilizing glucose units. The synthesis and breakdown
of glycogen in mammalian muscle (Fig. 11-4) involves
one of the first studied
91
and best known metabolic
control systems. Various aspects have been discussed
in Chapters 11, 12, and 17. The mechanism
92
and
regulatory features
93 96b
have been described. An
important recent development is the observation of
glycogen concentrations in human muscles in vivo
with
13
C NMR. This can be coupled with observation
of glucose 6-P by
31
P NMR. The concentration of the
latter is ~ 1 mM but increases after intense exercise.
94
Glycogen phosphorylase and glycogen synthase
alone are insufficient to synthesize and degrade glyco-
gen. Synthesis also requires the action of the branch-
ing enzyme amylo-(1,4 1,6-transglycosylase,
97
C. Synthesis and Degradation of Polysaccharides
C. Synthesis and Degradation of
Polysaccharides
Polysaccharides are all formed by transfer of
glycosyl groups onto initiating molecules or onto
growing polymer chains. The initiating molecule is
usually a glycoprotein. However, let us direct our
attention first to the growth of polysaccharide chains.
The glycosyl are transferred by the action of glycosyl-
transferases from substrates such as UDP-glucose,
other sugar nucleotides, and sometimes sucrose. The
glycosyltransferases act by mechanisms discussed in
Chapter 12 and are usually specific with respect both
to substrate structure and to the type of linkage
formed.
1. Glycogen and Starch
The bushlike glycogen molecules grow at their
numerous nonreducing ends by the transfer of gluco-
Ch 20 IND page-E- 2/14/03, 11:29 AM 1143
1144 Chapter 20. Some Pathways of Carbohydrate Metabolism
an enzyme with dual specificity. After the chain ends
attain a length of about ten glucose units, the branch-
ing enzyme attacks a 1,4-glycosidic linkage somewhere
in the chain. Acting much as does a hydrolase, it forms
a glycosyl enzyme or a stabilized carbocation interme-
diate. The enzyme does not release the severed chain
fragment but transfers it to another nearby site on
the glycogen molecule. There the enzyme rejoins the
bound oligosaccharide chain that it carries to a free
6-hydroxyl group of the glycogen creating a new
branch attached in -1,6-linkage. Degradation of
glycogen requires debranching after the long nonre-
ducing ends of the polysaccharide have been short-
ened until only four glycosyl residues remain at each
branch point. This is accomplished by amylo-1,6-
glucosidase / 4- -glucanotransferase. This 165-kDa
bifunctional enzyme transfers a trisaccharide unit
from each branch end to the main chain and also
removes hydrolytically the last glucosyl residue at
each branch point.
98-99a
How are new glycogen molecules made? There
is some evidence that a 37-kDa protein primer glyco-
genin is needed to initiate their formation.
100-101a
Thus, glycogen synthesis may be analogous to that
of the glycosaminoglycans considered in Section D,1.
Muscle glycogenin is a self-glycosylating protein,
which catalyzes attachment of ~ 7 to 11 glucose units
in 1,4 linkage to the hydroxyl group of Tyr 194.
The glucose units are added one at a time and when
the chain is long enough it becomes a substrate for
glycogen synthase.
100,102
The role of glycogenin in
liver has been harder to demonstrate,
103
but a second
glycogenin gene, which is expressed in liver, has
been identified.
104
Genes for several glycogenins or
glycogenin-like proteins have been identified in yeast,
Caenorhabditis elegans, and Arabidopsis.
101a,105
In contrast to animals, bacteria such as E. coli syn-
thesize glycogen via ADP-glucose rather than UDP-
glucose.
88
ADP-glucose is also the glucosyl donor
for synthesis of starch in plants. The first step in the
biosynthesis (Eq. 20-18) is catalyzed by the enzyme
ADP-glucose pyrophosphorylase (named for the
reverse reaction).
Glucose 1-P + ATP ADP-glucose + PP
i
In bacteria this enzyme is usually inhibited by AMP
and ADP and activated by glycolytic intermediates
such as fructose 1,6-P
2
, fructose 6-P, or pyruvate. In
higher plants, green algae, and cyanobacteria the en-
zyme is usually activated by 3-phosphoglycerate, a
product of photosynthetic CO
2
fixation, and is inhibited
by inorganic phosphate (P
i
).
106 108
.
In eukaryotic plants starch is deposited within
chloroplasts or in the cytoplasm as granules (Fig. 4-6)
in a specifically differentiated and physically fragile
plastid, the amyloplast.
108 110
Within the granules the
starch is deposited in layers ~ 9 nm in thickness. About
two-thirds of the thickness consists of nearly crystalline
arrays, probably of double helical amylopectin side
chains (Figs. 4-7, 4-8, 20-3) with amorphous segments
between the layers.
111 114
In maize there are at least
five starch synthases, one of which forms the straight
chain amylose.
115 117
There are also at least three
branching enzymes
118
and two or three debranching
enzymes.
119,120
As in the synthesis of glycogen the
molecules of amylopectin may grow at the many
nonreducing ends. A current model, which is related
to the broom-like cluster model of French (Fig. 4-7), is
shown in Fig. 20-3. The branches are thought to arise,
in part, by transglycosylation within the double helical
strands. After branching the two chains remain in a
double helix but the cut chain can now grow. Only
double helical parts of strands pack well in the crystal-
line layer. A recent suggestion is that debranching
enzymes then trim the molecule, removing single-
stranded regions.
112
The location (within the granule) of amylose, which
makes up 1530% by weight of many starches,
121
is
uncertain. It may fill in the amorphous layers. It may
be cut and provide primer pieces for new amylopectin
molecules.
122,122a
Another possibility is that it grows
by an insertion mechanism such as that portrayed for
cellulose in Fig. 20-5 and is extruded inward from the
membrane of the amyloplast. This mechanism might
explain a puzzling question about starch. The branched
amylopectin presumably grows in much the same way
as does glycogen. A branching enzyme transfers part
of the growing glycan chain to the CH
2
OH group of
(20-18)
Figure 20-3 Proposed structure of a molecule of amylopec-
tin in a starch granule. The highly branched molecule lies
within 9 nm thick layers, about 2 / 3 of which contains
parallel double helices of the kind shown in Fig. 4-8 in a
semicrystalline array. The branches are concentrated in the
amorphous region.
113,114,121
Some starch granules contain no
amylose, but it may constitute up to 30% by weight of the
starch. It may be found in part in the amorphous bands and
in part intertwined with the amylopectin.
122
Crystalline
layers
Amorphous
layers
Amylopectin
double helices
Reducing end
Amorphous
backbone
Ch 20 IND page-E- 2/14/03, 11:29 AM 1144
1145
BOX 20-D GENETIC DISEASES OF GLYCOGEN METABOLISM
In 1951, B. McArdle described a patient who
developed pain and stiffness in muscles after mod-
erate exericse.
a
Surprisingly, this person completely
lacked muscle glycogen phosphorylase. Since that
time several hundred others have been found with
the same defect. Glycogen accumulates in muscle
tissue in this disease, one of the several types of
glycogen storage disease.
b
Severe exercise is
damaging, but steady moderate exercise can be
tolerated. Until the time of McArdles discovery,
it was assumed that glycogen was synthesized by
reversal of the phosphorylase reaction. No hint of
the UDP-glucose pathway had appeared, and it
was, therefore, not obvious how glycogen could
accumulate in the muscles of these patients.
Leloirs discovery of UDP-glucose
at about the same time provided
the answer. Persons with McAr-
dle syndrome are greatly benefit-
ted by a high-protein diet, pre-
sumably because amino acids
such as alanine and glutamine are
converted efficiently to glucose
and because branched-chain
amino acids may serve as a direct
source of muscle energy.
c,d
Several other rare heritable
diseases also lead to accumula-
tion of glycogen because of some
block in its breakdown through
the glycolysis pathway. The en-
zyme deficiencies include those
of muscle phosphofructokinase,
e
liver phosphorylase kinase, liv-
er phosphorylase, and liver
glucose-6-phosphatase. In the
last case, glycogen accumulates
because the liver stores cannot be released to the
blood as free glucose.
b,f,g
This is a dangerous dis-
ease because blood glucose concentrations may fall
too low at night. The prognosis improved greatly
when methods were devised for providing the
body with a continuous supply of glucose. The
simplest treatment is ingestion of uncooked corn-
starch which is digested slowly.
b,h
In one of the
storage diseases the branching enzyme of glycogen
synthesis is lacking, and glycogen is formed with
unusually long outer branches. In another the
debranching enzyme is lacking, and only the outer
branches of glycogen can be removed readily.
i
The most serious of the storage diseases involve
none of the enzymes mentioned above. Pompe
disease is a fatal generalized glycogen storage disease
in which a lysosomal -1,4-glucosidase is lacking.
This observation suggested the existence of a new
and essential pathway of degradation of glycogen to
free glucose in the lysosomes. A few cases of glyco-
gen synthase deficiency have been reported. Little
or no glycogen is stored in muscle or liver, and
patients must eat at regular intervals to prevent
hypoglycemia. Severe diseases in which glycogen
synthesis is impaired include deficiencies of the
gluconeogenic enzymes pyruvate carboxylase and
PEP carboxykinase.
The following tabulation includes deficiencies
of glycogen metabolism, glycolysis, and gluconeo-
genesis.
a
Glycogen storage diseases are often desig-
nated as Types IV and these terms are included.
a
Huijing, F. (1979) Trends Biochem. Sci. 4, 192
b
Chen, Y.-T., and Burchell, A. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 935 965,
McGraw-Hill, New York
c
Slonim, A. E., and Goans, P. J. (1985) N. Engl. J. Med. 312, 355 359
d
Goldberg, A. L., and Chang, T. W. (1987) Fed. Proc. 37, 2301
2307
e
Raben, N., Sherman, J., Miller, F., Mena, H., and Plotz, P. (1993)
J. Biol. Chem. 268, 4963 4967
f
Nordlie, R. C., and Sukalski, K. A. (1986) Trends Biochem. Sci. 11,
85 88
g
Lei, K.-J., Shelly, L. L., Pan, C.-J., Sidbury, J. B., and Chou, J. Y.
(1993) Science 262, 580 583
h
Chen, Y.-T., Cornblath, M., and Sidbury, J. B. (1984) N. Engl. J. Med.
310, 171 175
i
Thon, V. J., Khalil, M., and Cannon, J. F. (1993) J. Biol. Chem. 268,
7509 7513
j
Shanske, S., Sakoda, S., Hermodson, M. A., DiMauro, S., and
Schon, E. A. (1987) J. Biol. Chem. 262, 14612 14617
Glycogen phosphorylase Muscle Moderate, late onset
(Type V), McArdle disease
Glycogen phosphorylase Liver Very mild
Phosphorylase kinase Liver Very mild
Debranching enzyme (Type III) Liver Mild
Lysosomal -glucosidase (Type II) Lethal, infant and adult form
Phosphofructokinase Muscle Moderate, late onset
Phosphoglycerate mutase
j
Muscle Moderate
Pyruvate carboxylase Lethal
PEP carboxykinase Lethal
Fructose-1,6-bisphosphatase Muscle Severe
Glycogen synthase Liver Mild
Branching enzyme (Type IV) Lethal, liver transplantation
Glucose-6-phosphatase (Type I) Severe if untreated
Severity Deficiency Organ
C. Synthesis and Degradation of Polysaccharides
Ch 20 IND page-E- 2/14/03, 11:29 AM 1145
1146 Chapter 20. Some Pathways of Carbohydrate Metabolism
a glucose unit in an adjacent polysaccharide chain
that lies parallel to the first, possibly in a double helix.
Since amylose and amylopectin are intimately inter-
mixed in the starch granules, it seems strange that the
branching enzyme never transfers a branch to mole-
cules of the straight-chain amylose. However, if the
linear amylose chains are oriented in the opposite
direction from the amylopectin chains, the nonreduc-
ing ends of the amylose molecules would be located
toward the center of the starch granule. Growth could
occur by an insertion mechanism at the reducing ends
and the ends could move out continually with the
amyloplast membrane as the granule grows.
123
Recent
evidence from
14
C labeling indicates that both amylose
and amylopectin too may grow by insertion at the
reducing end of glucose units from ADP-glucose.
123a,b
Branching could occur to give the structure of Fig. 20-3.
Starch synthesis in leaves occurs by day but at night
the starch is degraded by amylases, -glucosidases,
and starch phosphorylase. Both the starch synthases
and catabolic enzymes are present within the amylo-
plasts where they may be associated with regulatory
proteins of the 14-3-3 class.
122a
Digestion of dietary glycogen and starch in the
human body begins with the salivary and pancreatic
amylases, which cleave -1,4 linkages at random. It
continues with a glucoamylase found in the brush
border membranes of the small intestine where it
occurs as a complex with maltase.
74
Carbohydrases
are discussed in Chapter 12, Section B.
2. Cellulose, Chitin, and Related Glycans
Cellulose synthases transfer glucosyl units from
UDP-glucose, while chitin synthases utilize UDP-N-
acetylglucosamine. Not only green plants but some
fungi and a few bacteria form cellulose. The ameba
Dictyostelium discoideum also coats its spores with
cellulose.
124
Electron microscopic investigations suggest
that both in bacteria
125
and in plants
126
multienzyme
aggregates located at the plasma membrane synthesize
many polymer chains side by side to generate hydrogen-
bonded microfibrils which are extruded through the
membrane. Both green plants and fungi also form
important -1,3-linked glycans.
The bacterial cellulose synthase from Acetobacter
xylinum can be solubilized with detergents, and the
resulting enzyme generates characteristic 1.7 nm
cellulose fibrils (Fig. 20-4) from UDP-glucose.
125,127 129
These are similar, but not identical, to the fibrils of
cellulose I produced by intact bacteria.
125,130
Each
native fibril appears as a left-handed helix which may
contain about nine parallel chains in a crystalline
array. Three of these helices appear to coil together
(Fig. 20-4) to form a larger 3.7-nm left-handed helical
fibril. Similar fibrils are formed by plants. In both
Figure 20-4 Cellulose microfibrils being formed by Aceto-
bacter xylinum.
127
(A) Dark-field light micrograph after five
minutes of cellulose production (x 1250). (B) After 15 min-
utes a pellicle of cellulose fibers is forming (x 2000) (C)
Negatively stained cellulose ribbon. At the right the sub-
division into microfibrils is visible. Courtesy of R. Malcolm
Brown, Jr. (D) Cellulose microfibrils overlaying the plasma
membrane in the secondary cell wall of a tracheary element
of Zinnia elegans. Bar = 100 nm. (E) Rosettes in the plasma
membrane underlying the cellulose-rich secondary cell wall
thickening in A. elegans. Bar = 30 nm. (D) and (E) from Haigler
and Blanton.
132
Courtesy of Candace H. Haigler. (F) Chitin
microfibrils purified from protective tubes of the tube-worm
Lamellibrachia satsuma.
137
Courtesy of Junji Sugiyama.
A B
C
D E
F
Ch 20 IND page-E- 2/14/03, 11:29 AM 1146
1147
bacteria and plants the cellulose I fibrils that are formed
are highly crystalline, contain parallel polysaccharide
chains (Fig. 4-5), and have the tensile strength of steel.
Electron micrographs show that the cell envelope of
A. xylinum contains 5 80 pores, through which the
cellulose is extruded, lying along the long axis of the
cell.
129
The biosynthetic enzymes are probably bound
to the plasma membrane. Similar, but more labile,
cellulose synthases are present in green plants.
131
In
Arabidopsis there are ten genes. The encoded cellulose
synthases appear to be organized as rosettes on some
cell surfaces (Fig. 20-4E).
131a-133a
The rosettes may be
assembled to provide parallel synthesis of ~ 36 indi-
vidual cellulose chains needed to form a fibril.
131a
Because of the insolubility of cellulose fibrils it has
been difficult to determine whether they grow from
the reducing ends or the nonreducing ends of the
chains. From silver staining of reducing ends and
micro electron diffraction of cellulose fibrils attached to
bacteria,
134
Koyama et al. concluded that the reducing
ends are extruded from cells. New glucosyl rings
would be added at the nonreducing ends, which remain
attached noncovalently to the cells.
134
From amino
acid sequence similarities it was also concluded that
the same is true for Arabidopsis.
131,133
A single cellulose
chain has a twofold screw axis, each residue being
rotated 180

from the preceding residue (Fig. 4-5). It


was postulated that two synthases act cooperatively
to add cellobiose units. Another suggestion is that
sitosterol -glucoside acts in some fashion as a primer
for cellulose synthesis in plants.
133b
An insertion mechanism for synthesis of cellu-
lose. Using
14
C pulse and chase labeling Han and
Robyt found that new glucosyl units are added at the
reducing ends of cellulose chains formed by cell mem-
brane preparations from A. xylinum.
135
This conclusion
is in accord with the generalization that extracellular
polysaccharides made by bacteria usually grow from
the reducing end by an insertion mechanism that
depends upon a polyprenyl alcohol present in the
cell membrane.
136
This lipid alcohol, often the C
55
Figure 20-5 Proposed
insertion mechanism for
biosynthesis of cellulose.
Three enzymatic steps are
involved: a nucleophilic
displacement reaction of a
lipid phosphate on UDP-
glucose yields a glucosyl
diphosphate lipid in which the
-glycosyl linkage is retained. After
passage through the membrane the glycosyl
group is inserted into the reducing end of a cellulose
chain, which is covalently attached by a pyrophosphate link-
age to another lipid. The first lipid diphosphate is released and is hydrolyzed (step c) to the monophosphate, which crosses
the membrane to complete the cycle. After Han and Robyt.
135
As throughout this book P represents the phospho group
PO
3
H. The H may be replaced by groups which may contain oxygen atoms. This explains why an O is included in Lipid-O-P
but no O is shown between the Ps in -O-P-P.
a
c
Lipid-O-P
(Cytosolic surface)
Lipid-O-P
(Periplasmic surface)
O
OH
O
HO
OH
O
O
OH
OH
HO O
O
OH
HO
OH
Cellulose (n + 1 residues)

O
O
OH
OH
HO
O
P P
lipid
O
O
OH
OH
HO
O
P P lipid
Glucose-P-P-lipid
(Cytosolic surface)
O
OH
HO
OH
O
UDP-glucose

UMP
H
H

O
OH
O
HO
OH
O
O
OH
OH
HO O
O
OH
HO
OH
O
O
OH
H
OH
HO
O
P P
lipid
(Periplasmic surface)
Cellulose (n residues)
B
b
Lipid-O-P-P P
i
H
2
O
O
P P
lipid
O
P P
lipid
C. Synthesis and Degradation of Polysaccharides
Ch 20 IND page-E- 2/14/03, 11:29 AM 1147
1148 Chapter 20. Some Pathways of Carbohydrate Metabolism
bactoprenol, reacts with UDP-glucose (or other glyco-
syl donor) to give a lipopyrophospho-glucose (step a,
Fig. 20-5). The linkage of the UDP-glucose is retained
in this compound. The growing cellulose chain is
attached at the reducing end by a similar linkage to a
second lipid molecule. Then, in a displacement on the
anomeric carbon of the first glucosyl residue of the
cellulose chain, the new glucosyl unit is inserted with
inversion of the linkage to . In step c the pyrophos-
phate linkage of the lipid diphosphate is hydrolyzed
to regenerate the lipid monophosphate and to drive
the reaction toward completion. Two of the steps in
the cycle involve transport across the bacterial mem-
brane. The first involves the lipid O-P-P-glucose and
the second the lipid monophosphate. This type of
insertion mechanism is a common feature of poly-
prenol phosphate-dependent synthetic cycles for
extracellular polysaccharides (Figs. 20-6, 20-9 and Eq.
20-20). However, further verification is needed for
cellulose synthesis.
Regulation of cellulose synthesis in bacteria depends
on allosteric activator of cellulose synthase, cyclic
diguanylate (c-di-GMP), and a Ca
2+
-activated phos-
phodiesterase that degrades the activator.
129,138139a
Sucrose is the major transport form of glucose in plants.
Synthesis of both cellulose and starch is reduced in
mutant forms of maize deficient in sucrose synthase
(Eq. 20-13). This synthase, acting in the reverse direc-
tion, forms UDP-Glc from sucrose.
140,141
The enzymatic
degradation of cellulose is an important biological
reaction, which is limited to certain bacteria, to fungi,
and to organisms such as termites that obtain cellulases
from symbiotic bacteria or by ingesting fungi.
142
These enzymes are discussed in Chapter 12, Section
B,6. Genetic engineering methods now offer the
prospect of designing efficient cellulose-digesting
yeasts
143
that may be used to produce useful fermen-
tation products from cellulose wastes.
Callose and other -1,3-linked glycans.
Attempts to produce cellulose from UDP-Glc using
enzymes of isolated plasma membranes from higher
plants have usually yielded the -1,3-linked glucan
(callose) instead. This is a characteristic polysaccha-
ride of plant wounds which, as healing occurs, is
degraded and replaced by cellulose.
140,144
Callose
O
HO O
P
O CH
2
O O
G CH
2
O
P
O
O
O
O
G
OH

Cyclic diguanylate
formation is induced by a specific activator -furfuryl-
-glucoside, and callose synthase is virtually inactive
unless both the activator and Ca
2+
are present.
144
Beta-1,3-linked glycans are major components of
the complex layered cell wall of yeasts and other fungi.
In the fission yeast Saccharomyces pombe ~ 55% of the
cell wall carbohydrate consists of -1,3-linked glucan
with some -1,4-linked branches, ~ 28% is -1,3-linked
glucan, ~ 6% is -1,6-linked glucan, and ~ 0.5% is chitin.
There are two carbohydrate layers, the outer one
appearing amorphous. The inner layer contains inter-
woven fibrils of both -1,3-linked and -1,4-linked
glucans and holds the shape of the cell. The -1,3
glucan synthase is localized on the inner side of the
cell membrane and is activated by GTP and a small
subunit of the Rho family of G proteins.
145
Plants synthesize 1,3--glucanases that hydrolyze
the glycans of fungal cell walls. Synthesis is induced
by wounding as a defense reaction (see Box 20-E). These
glycanases also function in the removal of callose.
146
Chitin. Like cellulose synthase, fungal chitin
synthases are present in the plasma membrane and
extrude microfibrils of chitin to the outside.
147 150
In the
fungus Mucor the majority of the chitin synthesized
later has its N-acetyl groups removed hydrolytically to
form the deacetylated polymer chitosan.
151,152
Chitin
is also a major component of insect exoskeletons. For
this reason, chitin synthase is an appropriate target
enzyme for design of synthetic insecticides.
153
Chitin hydrolyzing enzymes are formed by fungi
and in marine bacteria.
154
Chitinases are also present
in plant vacuoles, where they participate in defense
against fungi and other pathogens
155
(Box 20-E). More
recently a chitinase has been identified in human
activated macrophages.
156
Another unanticipated
discovery was that a developmental gene designated
DG42, from Xenopus, has a sequence similar to that
of the NodC gene. The latter encodes a synthase for
chitin oligosaccharides (Nod factors) that serve as
nodulation factors in Rhizobia (Chapter 24). The enzyme
is synthesized for only a short time during early em-
bryonic development.
157
The significance of this
discovery is not yet clear. Synthesis of both the bac-
terial Nod factors and chitin oligosaccharides in zebra-
fish embryos occurs by transfer of GlcNAc residues
from UDP-GlcNAc at the nonreducing ends of the
O
OH
HO
O
HO
O
OH
-Furfuryl--glucoside
Ch 20 IND page-E- 2/14/03, 11:29 AM 1148
1149
chains.
158
Whether the same is true of chitin in fungi
or arthropods remains uncertain.
Cell walls of plants. The thick walls of higher
plant cells (Figs. 1-7, 4-14, and 20-4D) provide strength
and rigidity to plants and, at the same time, allow
rapid elongation during periods of growth.
159 163a
Northcote
164
likened the wall structure to glass fiber-
reinforced plastic (fiber glass). Thus, the cell wall
contains microfibrils of cellulose and other polysaccha-
rides embedded in a matrix, also largely polysaccha-
ride. The primary cell wall laid down in green plants
during early stages of growth contains loosely inter-
woven cellulose fibrils ~ 10 nm in diameter and with
an ~ 4 nm crystalline center. The cellulose in these
fibrils has a degree of polymerization of 8000 12,000
glucose units. As the plant cell matures, a secondary
cell wall is laid down on the inside of the primary
wall. This contains many layers of closely packed
microfibrils, alternate layers often being laid down
at different angles to one another (Fig. 20-4D). The
microfibrils in green plants are most often cellulose
but may contain other polysaccharides as well. Some
algae are rich in fibrils of xylan and mannan.
The materials present in the matrix phase vary
with the growth period of the plant. During initial
phases pectin (polygalacturonic acid derivatives)
predominate but later xylans and a variety of other
polysaccharides known as crosslinking glycans (or
hemicelluloses) appear. Primary cell wall constituents
of dicotyledons include xyloglucans (linear glucan
chains with xylose, galactose, and fucose units in
C. Synthesis and Degradation of Polysaccharides
BOX 20-E OLIGOSACCHARIDES IN DEFENSIVE AND OTHER RESPONSES OF PLANTS
Plants that are attacked by bacteria, fungi, or
arthropods respond by synthesizing broad-spectrum
antibiotics called phytoalexins,
a,b
by strengthening
their cell walls with lignin and hydroxyproline-rich
proteins called extensins,
c
and by making protease
inhibitors and other proteins that help to block the
chemical attack.
d
These plant responses seem to be
initiated by the release from an invading organism
of elicitors, which are often small oligosaccharide
fragments, sometimes called oligosaccharins.
e
These include -1,6-linked glucans that carry -1,3-
linked branches as well as chitin and chitosan oligo-
mers, derived from fungal cell walls.
f
Other elicitors
include galacturonic acid oligomers released from
damaged plant cell walls,
g
metabolites such as
arachidonic acid and glutathione,
h
and bacterial
toxins.
i
Any of these may serve as signals to plants
to take defensive measures.
Phytoalexins are often isoflavonoid derivatives
(Box 21-E). Their synthesis, like that of lignin, occur
via 4-coumarate (4-hydroxycinnamate, Fig. 25-8).
The ligase which forms the thioester of 4-coumarate
with coenzyme A is one of the pathogenesis-related
proteins whose synthesis is induced.
j
A second
induced enzyme is chalcone synthase, which con-
denses three acetyl units onto 4-coumaroyl-CoA as
shown in Box 21-E. Its induction by elicitors acting
on bean cells requires only five minutes.
h
Another
rapidly induced gene is that of cinnamoyl alcohol
dehydrogenase,
k
essential to lignin synthesis. Other
proteins formed in response to infections include
chitinases that are able to attack invading fungi
l,m
as well as the protease inhibitors. Their synthesis is
induced via derivatives of jasmonate, a product of
the octadecenoic acid pathway (Eq. 21-18).
a
As yet,
little is known about the mechanism by which
a
Blechert, S., Brodschelm, W., Hlder, S., Kammerer, L., Kutch-
an, T. M., Mueller, M. J., Xia, Z.-Q., and Zenk, M. H. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 40994105
b
Ebel, J., and Grisebach, H. (1988) Trends Biochem. Sci. 13, 2327
c
Kieliszewski, M. J., ONeill, M., Leykam, J., and Orlando, R.
(1995) J. Biol. Chem. 270, 25412549
d
Ryan, C. A. (1988) Biochemistry 27, 88798883
e
Ryan, C. A. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 12
f
Baureithel, K., Felix, G., and Boller, T. (1994) J. Biol. Chem. 269,
1793117938
g
Reymond, P., Grnberger, S., Paul, K., Mller, M., and Farmer,
E. E. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 41454149
h
Dron, M., Clouse, S. D., Dixon, R. A., Lawton, M. A., and Lamb,
C. J. (1988) Proc. Natl. Acad. Sci. U.S.A. 85, 67386742
i
Bidwai, A. P., and Takemoto, J. Y. (1987) Proc. Natl. Acad. Sci.
U.S.A. 84, 67556759
j
Douglas, C., Hoffmann, H., Schulz, W., and Hahlbrock, K.
(1987) EMBO J. 6, 11891195
k
Walter, M. H., Grima-Pettenati, J., Grand, C., Boudet, A. M., and
Lamb, C. J. (1988) Proc. Natl. Acad. Sci. U.S.A. 85, 55465550
l
Legrand, M., Kauffmann, S., Geoffroy, P., and Fritig, B. (1987)
Proc. Natl. Acad. Sci. U.S.A. 84, 67506754
m
Payne, G., Ahl, P., Moyer, M., Harper, A., Beck, J., Meins, F., Jr.,
and Ryals, J. (1990) Proc. Natl. Acad. Sci. U.S.A. 87, 98102
n
Cedergren, R. A., Lee, J., Ross, K. L., and Hollingsworth, R. I.
(1995) Biochemistry 34, 44674477
o
Jabbouri, S., Relic, B., Hanin, M., Kamalaprija, P., Burger, U.,
Prom, D., Prom, J. C., and Broughton, W. J. (1998) J. Biol.
Chem. 273, 1204712055
p
Dnari, J., Debell, F., and Prom, J.-C. (1996) Ann. Rev.
Biochem. 65, 503535
elicitors induce the defensive responses, but the
presence of receptors, of phosphorylation, and of
release of second messengers have been suggested.
d
Lipooligosaccharides known as Nod factors
(p. 1365) are another group of signaling mole-
cules. These chitin-related N-acylated oligomers
of N-acetylglucosamine (GlcNAc) do not defend
against infection but invite infection of roots of
legumes by appropriate species of Rhizobia
np
lead-
ing to formation of nitrogen-fixing root nodules.
Ch 20 IND page-E- 2/14/03, 11:29 AM 1149
1150 Chapter 20. Some Pathways of Carbohydrate Metabolism
branches),
164a
other crosslinking glycans, and galactur-
onic acid-rich pectic materials.
163a
The xyloglycans,
which comprise 20% of the cell wall in some plants,
have a backbone of -1,4-linked glucose units with
numerous -1,6-linked xylose rings, some of which
carry attached L-arabinose, galactose, or fucose. The
structures, which vary from species to species, are
organized as repeating blocks with a continuous
glucan backbone. Another crosslinking glycan is
glucuronoarabinoxylan. The backbone is -1,4-linked
xylose. Less abundant glucomannans, galactomannans,
and galactoglucomannans, with -1,4-linked mannan
backbone structures, are also present in most angio-
sperms.
163a
Pectins form a porous gel on the inside surface
of plant cell walls.
163a,164a
A major component is a
homogalacturonan, which consists of -1,4-linked
galacturonic acid (GalA). A second is rhamnogalac-
turonan I, an alternating polymer of (2-L-Rha1
4GalA ) units. The most interesting pectin compo-
nent is rhamnogalacturonan II, one of the less abun-
dant constuents of pectin. It is obtained by hydrolytic
cleavage of pectin by a polygalacturonidase. Before
such release it forms parts (hairy regions) of pectin
molecules that are largely homogalacturonans (in
smooth regions). A rhamnogalacturonan II segment
consists of 11 different monomer units.
164b f
Attached
to the polygalacturonic acid backbone are four oligo-
saccharides, consisting of rhamnose, galactose and
fucose as well as some unusual sugars (see structure in
Box 20-E). This polysaccharide is apparently present
in all higher plants and is unusually stable, accumu-
lating, for example, in red wine.
164e
It contains two
residues of the branched chain sugar apiose, one of
which is a site of crosslinking by boron (Box 20-F). A
borate diol ester linkage binds two molecules of the
pectin together as a dimer, perhaps controlling the
porosity of the pectin gel. All of the complex cell wall
polysaccharides bind, probably through multiple
hydrogen bonds, to the cellulose microfibrils (Fig. 4-14).
The resulting structures are illustrated in drawings of
Carpita and McCann,
163a
which are more current than
is Fig. 4-14. The cellulose plus crosslinking glycans
form one network in the cell wall. The pectic sub-
stances form a second independent network. Some
covalent crosslinking occurs, but most interactions are
noncovalent.
163a
The site of biosynthesis of pectins
and hemicelluloses is probably Golgi vesicles which
pass to the outside via exocytosis. However, the cellu-
lose fibrils as well as the chitin in fungi are apparently
extruded from the plasma membrane.
Although the principal cell wall components of
plants are carbohydrates, proteins account for 5 10%
of the mass.
165
Predominant among these are glyco-
protein extensins. Like collagen, they are rich in
4-hydroxyproline which is glycosylated with arabi-
nose oligosaccharides and galactose (p. 181). Other
hydroxyproline-containing proteins with the charac-
teristic sequence (hydroxyproline)
4
-Ser are also found,
e.g., in soybean cell walls.
166
Some plant cell walls
contain glycine-rich structural proteins. One in the
petunia consists of 67% glycine residues.
167
During
advanced stages of formation, as the walls harden into
wood, large amounts of lignins are laid down in some
plant cells. These chemically resistant phenylpropanoid
polymers contain many crosslinked aromatic rings (Fig.
25-9).
163a
A remarkable aspect of primary plant cell walls is
their ability to be elongated extremely rapidly during
growth. While the driving force for cell expansion is
thought to be the development of pressure within the
cell, the manner in which the wall expands is closely
regulated. After a certain point in development, elon-
gation occurs in one direction only and under the
influence of plant hormones. Most striking is the
effect of the gibberellins (Eq. 22-5), which cause very
rapid elongation. Elongation of plant cell walls may
depend to some extent upon chemical cleavage and
reforming of crosslinking polysaccharides. However,
the cellulose fibrils probably remain intact and slide
past each other.
161
A curious effect, which is mediated
by proteins called expansins,
133a,168
is the ability of
plant tissues to extend rapidly when incubated in a
mildly acidic buffer of pH < 5.5. Expansins are also
involved in ripening of fruit. They may disrupt non-
covalent bonding between cellulose fibrils and the
hemicelluloses.
169,170
The -expansins of grasses are
allergens found in grass pollens.
133a,168
The borate
diol ester linkages in the pectin may also facilitate
expansion.
3. Patterns in Polysaccharide Structures
How can the many complex polysaccharides
found in nature be synthesized? Are there genetically
determined patterns? How are these controlled? The
answer can be found in the specificities of the hundreds
of known glycosyltransferases
171,172
and in the patterns of
expression of the genes for transferases and other proteins.
As a consequence, a great variety of structurally varied
polysaccharide structures arise, especially on cell
surfaces. The structures are not random but depend
upon the assortment of glycosyltransferases available
at the particular stage of development in a tissue.
The numerous possibilities can account for much of
the variation observed between species, between
tissues, and also among individuals.
The simplest pattern is the growth of straight-
chain homopolysaccharides such as amylose, cellulose,
and chitin. The glycosyltransferases must recognize
both the glycosyl donor, e.g., ADP-glucose, UDP-
glucose, and also the correct end of the growing
polymer, always adding the same monomer unit.
Ch 20 IND page-E- 2/14/03, 11:29 AM 1150
1151
BOX 20-F WHAT DOES BORON DO?
For 75 years or more it has been known that
boron is essential for growth of green plants.
a,b
In
its absence root tips fail to elongate normally, and
synthesis of DNA and RNA is inhibited. Boron in
the form of boric acid, B(OH)
3
, is absorbed from soil.
Although deficiency is rare it causes disintegration
of tissues in such diseases as heart rot of beets and
drought spot of apples. The biochemical role has
been obscure, but is usually thought to involve
formation of borate esters with sugar rings or other
molecules with adjacent pairs of OH groups ( as
in the accompanying structures). A regulatory role
involving the plant hormones auxin, gibberelic acid,
and cytokinin has also been suggested.
Diatoms also require boron, which is incorpo-
rated into the silicon-rich cell walls.
a
Some strains
of Streptomyces griseus produce boron-containing
macrolide antibiotics such as aplasmomycin (right).
c
Recently a function in plant cell walls has been
identified (see also main text) as crosslinking of
rhamnogalacturonan portions of pectin chains by
borate diol ester linkages as illustrated.
It was long thought that boron was not required
by human beings, but more recent studies suggest
that we may need ~ 30 g / day.
d
The possible func-
tions are uncertain. Animals deprived of boron
show effects on bone, kidney, and brain as well as a
relationship to the metabolism of calcium, copper,
and nitrogen. Nielson proposed a signaling func-
tion, perhaps via phosphoinositides, in animals.
b
O
H
CH
3
CH
3
OH
H
O
O
O O
O
H
H
CH
3
H
CH
3
H
O
H
CH
3
CH
3
HO
H
O
B
O
O O
O
H
H
CH
3
H
CH
3
H
H
H

Aplasmomycin, a boron-containing antibiotic


a
Salisbury, F. B., and Ross, C. W. (1992) Plant Physiology, 4th ed.,
Wadsworth, Belmont, California
b
Nielsen, F. H. (1991) FASEB J. 5, 2661 2667
c
Lee, J. J., Dewick, P. M., Gorst-Allman, C. P., Spreafico, F.,
Kowal, C., Chang, C.-J., McInnes, A. G., Walter, J. A., Keller, P.
J., and Floss, H. G. (1987) J. Am. Chem. Soc. 109, 5426 5432
d
Nielsen, F. H. (1999) in Modern Nutrition in Health and Disease,
9th ed. (Shils, M. E., Olson, J. A., Shike, M., and Ross, A. C.,
eds), pp. 283 303, Williams & Wilkins, Baltimore, Maryland
O O
O
O
O
Chain 1
O
B
O
O
O
O
Chain 2

Borate diol ester linkage


between apiose rings in
oligosaccharide clusters
of rhamnogalacturonan II
in two pectin chains.
Rhamnogalacturonan II monomer showing position of the boron crosslinks.
Most sugars are of the D-configuration with the exception of L-rhamnose and
L-fucose. Except when designated f the sugar rings are pyranose. Unusual
sugars include apiose (Api), aceric acid (Ace, 3-C-carboxy-5-deoxyxylose),
and 3-deoxy-D-lyxo-2-heptulosaric acid (Dha).
C. Synthesis and Degradation of Polysaccharides
4GalA1 4GalA1 4GalA1 4GalA1 4GalA1 4GalA1 4GalA1
2
Apif1

3
DHA1

2
Apif1

3
KDO1

5
Rha1

3
Rha1

5
Araf1

Glc1 4Fuc1 4Rha1


2
Gal1

2
2 O-Me-Xyl1
3
GalA1
2
AcefA1

2
Ara1 4Gal1

2
2-O-Me-Fuc1

2
Rha1
2
Araf1
2
GalA1
O
3
O
2
O
B
O

Ch 20 IND page-E- 2/14/03, 11:29 AM 1151


1152 Chapter 20. Some Pathways of Carbohydrate Metabolism
a second dextran chain enters the catalytic site, serving
as the glycosyl acceptor. See Robyt for a detailed dis-
cussion of synthesis of dextrans and related polysac-
charides such as alternan and the -1,3-linked mutan
(p. 175).
121,176
Some bacteria form -2,6-linked fructans
by a similar mechanism, with glucose being released
by displacement on C2 of sucrose.
181
Fructans are also
formed in green plants, apparently from reaction of two
molecules of sucrose with release of glucose to form the
trisaccharide Fruf2 1Fruf21-Glcp, which then
transfers a fructosyl group to the growing chain.
Lipid-dependent synthesis of polysaccharides.
Insertion of monomer units at the base of a chain is a
major mechanism of polymerization that is utilized
for synthesis not only of polysaccharides but also of
proteins (Chapter 29). For most carbohydrates the
synthesis is dependent upon a polyprenyl lipid alcohol.
In bacteria this is often the 55-carbon undecaprenol or
bactoprenol,
136
which functions as a phosphate ester:
It serves as a membrane anchor for the growing poly-
saccharide. We have already discussed one example
in the hypothetical cellulose synthase mechanism of
Fig. 20-5. For some polysaccharides the mechanism is
better established. The synthetic cycles all resemble
that of Fig. 20-5 and can be generalized as in Eq. 20-20.
Here NDP-Glx is a suitable nucleotide disphosphate
derivative of sugar Glx, and Z-Glx is the repeating unit
of the polysaccharide formed by the action of glycosyl-
transferases and other enzymes.
For example, the biosynthesis of alginate involves
GDP-mannuronic acid (GDP-ManA) as NDP-Glx,
bactoprenol as the lipid, and a glycosyltransferase
that inserts a second mannuronate residue (as Z).
In contrast, hyaluronan and the polysaccharide chains
of glycosaminoglycans (Fig. 4-11) have an alternating
pattern. For a hyaluronan chain growing at the reduc-
ing end, one active site of hyaluronan synthase must
be specific for UDP-GlcNAc and transfer the sugar
unit only to the end of a glucuronic acid ring. A second
active site must be specific for UDP-glucuronic acid
but attach it only to the end of an acetylglucosamine
unit.
172a,172b
There is still uncertainty about the direc-
tion of growth of hyaluronan.
173 175
Some hyaluronan
synthases are lipid-dependent and their mechanism
may resemble that proposed for cellulose synthesis
(Fig. 20-5).
Dextrans. Some polysaccharides, such as the
bacterial dextrans, are synthesized outside of cells by
the action of secreted enzymes. An enzyme of this
type, dextran sucrase of Leuconostoc and Streptococcus,
adds glucosyl units at the reducing ends of the dextran
chains (p. 174). Sucrose is the direct donor of the
glucosyl groups, which are added by an insertion
mechanism.
121,176 178
However, it is not dependent
upon a membrane lipid as is that of Fig. 20-5. The
glucosyl groups are transferred from sucrose to one
of a pair of carboxylate groups of aspartate side chains
in the active site.
179,180
If both carboxylates are gluco-
sylated, a dextran chain can be initiated by insertion
of one glucosyl group into the second (Eq. 20-19). The
dextran grows alternating binding sites between the
two carboxylates. Chain growth can be terminated by
reaction with a sugar or oligosaccharide that fits into
the active site and acts in place of the glucosyl group
attached to Asp 1 as pictured in Eq. 20-19. The -1,3-
linked branches can be formed when a 3-OH group of
H
O
PO
3
2
CH
3
CH
3

10
Undecaprenol phosphate
O
OH
HO
HO
OH
O
Fructose
Asp 1
O
O
O
OH
O
HO
OH
Asp 2
O
O
R
O
OH
HO
HO
O
Asp 1
O

O
H

6
Base
Sucrose
Fructose
R = H or growing 1,6-linked chain
(20-19)
(20-20)
NDPGlx NMP
LipidP LipidPPGlx
LipidPP LipidPPGlxZ
Polysaccharide PolysaccharideZGlx
P
i
H
2
O
Action of various glycosyl
transferases and other
enzymes to build a modified
sugar or oligosaccharide
O Y O Y
Ch 20 IND page-E- 2/14/03, 11:29 AM 1152
1153
An additional transferase that uses acetyl-CoA as a
substrate sometimes acetylates one mannuronate
unit. The disaccharide units are then inserted into
the growing chain. An additional modification, which
occurs after polymerization, is random C5 epimeriza-
tion of unacetylated D-mannuronate residues to L-
guluronate.
136,182
Formation of alginate is of medical
interest because infections by alginate-forming bacte-
ria are a major cause of respiratory problems in cystic
fibrosis.
182
Sometimes an oligosaccharide assembled on the
polyprenol phosphate represents a substantial block
in assembly of a repeating polymer. For example,
the xanthan gum (p. 179) produced by the bacterium
Xanthomonas campestris is formed by several successive
glycosyl transfers to bactoprenol-P-P-Glc. A second
glucose is transferred onto the first from UDP-Glc,
forming a pair of glucosyl groups in -1,4 linkage.
Mannose is then transferred from GDP-Man and
joined in an -1,3 linkage to the first GDP-Man to
form a branch point. A glucuronate residue is then
transferred from UDP-GlcA and another mannose
from GDP-Man. The last mannose is modified by
reaction with PEP to form a ketal (Eq. 4-9). The prod-
uct of this assembly is the following lipid-bound
oligosaccharide block.
This is inserted into the growing polysaccharide using
the free 4-OH on the second glucose to link the units
in a cellulose type chain. The twelve separate genes
needed for synthesis of xanthan gum are contained in
a 16-kb segment of the X. campestris genome.
136
Lipid-
bound intermediates are also involved in synthesis
of peptidoglycans (Fig. 20-9) and in the assembly of
bacterial O-antigens (Fig. 8-30). Both of these also
yield block polymers.
D. Proteoglycans and Glycoproteins
The glycoproteins contain oligosaccharides attached
to the protein either through O-glycosidic linkages
with hydroxyl groups of side chains of serine, threo-
nine, hydroxyproline, or hydroxylysine (O-linked) or
via glycosylaminyl linkages to asparagine side chains
(N-linked). The core proteins of the proteoglycans
carry long polysaccharide chains, which are usually
O-linked and are usually described as glycosamino-
glycans.
1. Glycosaminoglycans
Synthesis of the alternating polysaccharide hyalu-
ronan has been discussed in Section C,3 and may
occur by an insertion mechanism. However, other
glycosaminoglycans (sulfate esters of chondroitin,
dermatan, keratan, heparan, and heparin) grow at
their nonreducing ends.
183,184
Their synthesis is usually
initiated by the hydroxyl group of serine or threonine
side chains at special locations within several secreted
proteins.
185
These proteins are synthesized in the rough
ER and then move to the Golgi. Addition of the first
sugar ring begins in the ER with transfer of single
xylosyl residues to the initiating OH groups.
186 190b
This reaction is catalyzed by the first of a group of
special glycosyltransferases of high specificity that
form the special terminal units (Chapter 4, Section
D,1), that anchor the alternating polysaccharide repre-
sented here as (XY)
n
:
(X Y)
n
4 GlcA1 3 Gal1
3 Gal1 4 Xyl1 O-Ser / Thr
After transfer of the xylosyl residue from UDP-xylose
to the OH group in the protein,
190a
a second enzyme
with proper specificity transfers a galactosyl group
from UDP-galactose, joining it in -1,4 linkage. A third
enzyme transfers another galactosyl group onto the
first one in -1,3 linkage. A fourth enzyme, with a
specificity different from that used in creating the
main chain, then transfers a glucuronosyl group from
UDP-glucuronic acid onto the chain terminus to com-
plete the terminal unit.
190c
Then two more enzymes
transfer the alternating units in sequence to form the
repeating polymer with lengths of up to 100 or more
monosaccharide residues. The sequence (XY)
n
in the
preceding formula is:
( 4 GlcA1 3 GalNAc1 )
n
for chondroitin
172a,191,192
( 4 GlcA1 4 GlcNAc1 )
n
for heparan sulfate and heparin
( 3 Gal1 4 GlcNAc1 )
n
for dermatan sulfate
Subsequent modifications of the polymers involve
extensive formation of O-sulfate esters,
190a,193 197
N-
deacetylation and N-sulfation,
198,199
and epimerization
at C5.
10
In some tissues almost all GluA is epimer-
ized.
200
The modifications are especially extensive in
dermatan, heparan sulfates, and heparin (see also p.
177).
196,201 203b
The modifications are not random and
follow a defined order. N-Deacetylation must precede
N-sulfation, and O-sulfation is initiated only after N-
sulfation of the entire chain is complete. The modifica-
tions occur within the Golgi (see Fig. 20-7) but not all
Acetyl Glc1
6 4
Man1 4 GlcA1 2 Man1 3 Glc1 O-P-P-lipid

Pyruvate
D. Proteoglycans and Glycoproteins
Ch 20 IND page-E- 2/14/03, 11:30 AM 1153
1154 Chapter 20. Some Pathways of Carbohydrate Metabolism
of the glycosyltransferases, PAPS (3'-phosphoadenosine
5'-phosphosulfate)-dependent sulfotransferases, and
epimerases are present within a single compartment.
Nevertheless, an entire glycosaminoglycan chain can
be synthesized within 1 3 min.
189
The completed polymers are modified uniformly.
There are clusters of sulfo groups with unusual structures
in chondroitin from squid and shark cartilages
204,205
and fucosylated chondroitin from echinoderms.
206
Similar modifications are present less extensively in
vertebrates. One of the best known modifications
forms the unique pentasaccharide sequence shown
in Fig. 4-13, which is essential to the anticoagulant
activity of heparin. This sequence has been synthe-
sized in the laboratory as have related longer heparin
chains. A sequence about 17 residues in length con-
taining an improved synthetic version of the unique
pentasaccharide binds tightly to both thrombin and
antithrombin (Chapter 12, Section C,9).
207,208
This
opens the door to the development of improved sub-
stitutes for the medically important heparin. Heparan
sulfate chains are found on proteoglycans throughout
the body, but the highly modified heparin does not
circulate in the blood. It is largely sequestered in cyto-
plasmic granules within mast cells and is released as
needed.
208a
Heparin binds to many different proteins
(p. 177). Among them is the glycoprotein selenoprotein
P (p. 824), which may impart antioxidant properties to
the extracellular matrix.
208b
Although glycosaminoglycans are most often
attached to O-linked terminal units, chondroitin sulfate
chains can also be synthesized with N-linked oligosac-
charides attached to various glycoproteins serving
as initiators.
209
At least one form of keratan sulfate,
found in the cornea, is linked to its initiator protein via
GlcNAc-Man to N-linked oligosaccharides of the type
present in many glycoproteins (Section D).
At least 25 different proteins that are secreted into
the extracellular spaces of the mammalian body carry
glycosaminoglycan chains.
183,210,211
Most of these
proteins can be described as (1) small leucine-rich
proteoglycans with 36- to 42-kDa protein cores and
(2) large modular proteoglycans whose protein
cores have molecular masses of 40 to 500 kDa.
210
The
most studied of the second group is aggrecan, a major
component of cartilage. This 220-kDa protein carries
~ 100 chondroitin chains, each averaging about 100
monosaccharide residues and ~ 100 negative charges
from the carboxylate and sulfate groups. Aggrecan
has three highly conserved globular domains near the
N and C termini.
212213a
The G1 domain near the N
terminus is a lectin (p. 186), which, together with a
small link protein that is structurally similar to the G1
domain, binds to a decasaccharide unit of hyaluronan.
One hyaluronan molecule of 500- to1000-kDa mass
(~ 2500 5000 residues) may bind 100 aggrecan and
link molecules to form an ~ 200,000-kDa particle such
as that shown in Fig. 4-16. These enormous highly
negatively charged molecules, together with associated
counterions, draw in water and preserve osmotic
balance. It is these molecules that keep our joints
mobile and which deteriorate by proteolytic degrada-
tion in the common osteoarthritis.
214,215
The keratan
sulfate content of cartilage varies with age, and the
level in serum and in synovial fluid is increased in
osteoarthritis.
215
Keratan sulfate is also found in
the cornea and the brain. Its content is dramatically
decreased in the cerebral cortex of patients with
Alzheimer disease.
216
Other modular core proteins
210
include versican
of blood vessels and skin,
210,213a,217,217a
neurocam and
brevican of brain, perlecan of basement membranes,
218
agrin of neuromuscular junctions, and testican of
seminal fluid. However, several of these have a broader
distribution than is indicated in the foregoing descrip-
tion. The sizes vary from 44 kDa for testican to greater
than 400 kDa for perlecan. The numbers of glycos-
aminoglycan chains are smaller than for aggrecan,
varying from 1 to 30. Another of the chondroitin
sulfate-bearing core protein is appican, a protein
found in brain and one of the splicing variants of the
amyloid precursor protein that gives rise to amyloid
deposits in Alzheimer disease (Chapter 30).
217a,b
The core proteins of the leucine-rich proteoglycans
have characteristic horseshoe shapes and are con-
structed from ~ 28-residue repeats, each containing a
turn and an helix. The three-dimensional struc-
tures are doubtless similar to that of a ribonuclease
inhibitor of known structure which contains 15 tandem
repeats.
219,220
A major function of these proteoglycans
seems to be to interact with collagen fibrils, which
have distinct proteoglyan-binding sites,
221
and also
with fibronectin.
222
The small leucine-rich proteo-
glycans have names such as biglycan, decorin,
222a
fibromodulin, lumican, keratoglycan, chondro-
adherin, osteoglycin, and osteoadherin.
219,223223c
The distribution varies with the tissue and the stage
of development. For example, biglycan may function
in early bone formation; decorin, which has a high
affinity for type I collagen, disappears from bone
tissue as mineralization takes place. Osteoadherin is
found in mature osteoblasts.
223
Phosphocan, another
brain proteoglycan, has an unusually high content
(about one residue per mole) of L-isoaspartyl residues
(see Box 12-A).
224
Proteoglycans bind to a variety of different proteins
and polysaccharides. For example, the large extracel-
lular matrix protein tenascin, which is important to
adhesion, cell migration, and proliferation, binds to
chondroitin sulfate proteoglycans such as neurocan.
225
Syndecan, a transmembrane proteoglycan, carries
both chondroitin and heparan sulfate chains, enabling
it to interact with a variety of proteins that mediate
cellmatrix adhesion.
185
Ch 20 IND page-E- 2/14/03, 11:30 AM 1154
1155
The ability of dissociated cells of sponges to aggre-
gate with cells only of a like type (p. 29) depends upon
large extracellular proteoglycans. That of Microciona
prolifera appears to be an aggregate of about three
hundred 35-kDa core protein molecules with equal
masses of attached carbohydrate. This aggregation
factor has a total mass of ~ 2 x 10
4
kDa.
226,227
It appar-
ently interacts specifically, in the presence of Ca
2+
ions,
with a 210-kDa cell matrix protein to hold cells of the
same species together.
227
2. O-Linked Oligosaccharides
A variety of different oligosaccharides are attached
to hydroxyl groups on appropriate residues of serine,
threonine, hydroxylysine, and hydroxyproline in many
different proteins (Chapter 4). Such oligosaccharides
are present on external cell surfaces, on secreted pro-
teins, and on some proteins of the cytosol and the
nucleus.
228 231b
The rules that determine which OH
groups are to become glycosylated are not yet clear.
232
Glycosylation occurs in the ER, and, just as during
synthesis of the long carbohydrate chains of proteo-
glycans, the sugar rings are added directly to an OH
group, either of the protein or of the growing oligosac-
charide. The first glycosyl group transferred is most
often GalNAc for external and secreted proteins
233
but more often GlcNAc for cytosolic and nuclear
proteins.
228,231,233ac
Glycosylation of protein OH
groups can occur on either the luminal or cytosolic
faces of the ER membranes.
234
The external O-linked
glycoproteins often have large clusters of oligosaccha-
rides attached to OH groups of serine or threonine,
but cytosolic proteins may carry only a small number
of small oligosaccharides.
Of great importance are the blood group deter-
minants which are discussed in Box 4-C. The ABO
determinants are found at the nonreducing ends of
O-linked oligosaccharides. Conserved Ser/Thr sites
in the epidermal growth factor domains (Table 7-3) of
various proteins carry O-linked fucose.
235
The secreted mucins are unique in having clus-
ters of large numbers of oligosaccharides linked by
N-acetylgalactosamine to serine or threonine of the
polypeptide.
236
The following core structures predomi-
nate.
237
These may be lengthened or further branched by
the particular variety of glycosyltransferases present in a
tissue and by their specificities.
233
The human genome
contains at least nine mucin genes.
238
The large apomu-
cins contain central domains with tandem repeats rich in
Ser, Thr, Gly, and Ala and flanked at the ends by cysteine-
rich domains.
239
For example, porcine submaxillary
mucins are encoded by a gene with at least three alleles
that encode 90, 125, or 133 repeats. The polypeptide may
contain as many as 13,288 residues. N-terminal cysteine-
rich regions are involved in dimer formation.
240
3. Assembly of N-Linked Oligosaccharides on
Glycosyl Carrier Lipids
In eukaryotes the biosynthesis of the N-linked
oligosaccharides of glycoproteins depends upon the
polyprenyl alcohols known as dolichols, which are
present in membranes of the endoplasmic reticulum.
They contain 16 20 prenyl units, of which the one
bearing the OH group is completely saturated as a
result of the action of an NADPH-dependent reductase
on the unsaturated precursor.
241
The predominant
dolichol in mammalian cells contains 19 prenyl units.
The structure of its mannosyl phosphate ester, one
of the intermediates in the oligosaccharide synthesis,
is illustrated below. The fully extended 95-carbon
dolichol has a length of almost 10 nm, four times
greater than that of oleic acid and twice the thickness
of the nonpolar membrane bilayer core. The need
for this great length is not clear nor is it clear why the
first prenyl unit must be saturated for good acceptor
activity.
The assembly of the oligosaccharides that will
become linked to Asn residues in proteins occurs on
the phosphate head of dolichol-P. The process begins
on the cytoplasmic face of the membrane and within
the lumen of the rough or smooth ER and continues
within cisternae of the Golgi apparatus.
234,242 245
The
initial transfer of GlcNAc-P to dolichol-P (Fig. 20-6,
step a) appears to occur on the cytoplasmic face of the
ER and is specifically inhibited by tunicamycin.
246,247
As the first committed reaction of N-glycosylation,
it is regulated by a variety of hormonal and other
factors.
248,249
The reaction takes place cotranslation-
ally as the still unfolded peptide chain leaves the
ribosome.
242
The oligosaccharide, still attached to the
dolichol, continues to grow on the cytosolic surface
of the ER membrane by transfer of GlcNAc and
five residues of mannose from their sugar nucleotide
forms (Fig. 20-6, steps b and c).
249a
The intermediate
Dol-P-P-GlcNAc
2
Man
5
crosses the membrane bilayer
(Fig. 20-6, step d), after which mannosyl and glucosyl
units are added (steps e and f). These sugars are car-
ried across the membrane while attached to dolichol.
D. Proteoglycans and Glycoproteins
core 1
core 2
Gal1 3 GalNAc Ser/Thr
Gal1 3 GalNAc Ser/Thr Ser/Thr
6
GalNAc1

core 3
core 4
GlcNAc1 3 GalNAc Ser/Thr
Gal1 4 GlcNAc1 6 GalNAc Ser/Thr
3
GlcNAc1

Ch 20 IND page-E- 2/14/03, 11:30 AM 1155


1156 Chapter 20. Some Pathways of Carbohydrate Metabolism
O
OH
HO
O
HO
P
OH
CH
3
CH
3
CH
3
CH
3
O O
12-17
Z E E
O

Man-P-dolichol
CH
3
H
O
N
HO
O
O
NH
HO
O
(CH
2
)
n
C
H
3
C
H
3
C
HO
C H
3
C
O
OH
OH
O
H
OH
HO OH
N
NH
O
O
H
H
Tunicamycins
n = 8, 9, 10, 11
The completed 14-residue branched oligosaccharide
Glc
3
Man
9
GlcNAc
2
, with the structure indicated in Fig.
20-6, is then transferred to a suitable asparagine side
chain (step g). This may be on a newly synthesized
protein or on a still-growing polypeptide chain that is
being extruded through the membrane into the lumenal
space of the rough ER (Eq. 20-21; Fig. 20-6). The glyco-
sylation site is often at the sequence Asn-X-Ser(Thr),
which is likely to be present at a beta bend in the folded
protein. Bends of the type illustrated in Eq. 20-21 and
stabilized by the asparagine side chain are apparently
favored.
250
In such a bend the OH of the serine or
threonine helps to polarize the amide group of the
Asn side chain, perhaps enolizing it and generating a
nucleophilic center that can participate in a displace-
ment reaction
250,251
as indicated in Eq. 20-21. The
oligosaccharyltransferase that catalyzes the reaction
is a multisubunit protein. As many as eight different
subunits have been reported for the
enzyme in yeast. Genes for at least
five of these are essential.
250252b
One subunit serves to recognize
suitable glycosylation sites.
Trimming of glycoprotein
oligosaccharides. After transfer to
glycoproteins the newly synthesized
oligosaccharides undergo trimming,
the hydrolytic removal of some of
the sugar units, followed by addi-
tion of new sugar units to create the
finished glycoproteins. The initial
glycosylation process ensures that
the glycoproteins remain in the
lumen of the ER or within vesicles
or cisternae separated from the cyto-
plasm. The subsequent processing
appears to allow the cell to sort the proteins. Some
remain attached to membranes and take up residence
within ER, Golgi, or plasma membrane. Others are
passed outward into transfer vesicles, Golgi, and
secretion vesicles. A third group enter lysosomes.
A series of specific inhibitors of trimming reactions,
some of whose structures are shown in Fig. 20-7, has
provided important insights.
253 255
Use of these inhi-
bitors, together with immunochemical methods and
study of yeast mutants,
250,252,256
is enabling us to learn
many details of glycoprotein biosynthesis.
Whereas the formation of dolichol-linked oligosac-
charides occurs in an identical manner in virtually all
eukaryotic cells, trimming is highly variable as is the
addition of new monosaccharide units.
257257b
The
major pathway for mammalian glycoproteins is shown
in Fig. 20-7. Specific hydrolases in the ER remove all
of the glucosyl units and one to three mannosyl
units.
258
Removal of additional
mannosyl residues occurs in the cis
Golgi, to give the pentasaccharide
core Man
3
GlcNAc
2
which is common
to all of the complex N-linked oligo-
saccharides. However, partial trim-
ming without additional glycosyl-
ation produces some high mannose
oligosaccharides.
258a
Removal of
glucose may be necessary to permit
some glycoproteins to leave the ER.
Sulfate groups and in some
cases fatty acyl groups
259
may also
be added. The exact composition of
the oligosaccharides may depend
upon the condition of the cell and
may be altered in response to exter-
nal influences.
257
Oligosaccharides
attached to proteins that remain in
the ER membranes may undergo
O
Glc Glc Glc Man Man Man
Man
Man
Man
Man Man
Man
GlcNAc O
O
NH
HO
OH
C
CH
3
O
O P P O Dolichol
O
N
H
H
H
N
H
R
O
N H O C
O
O
N
H
CH
3
N
H
H
H
O C
N
H
B

Enzyme
(20-21)
Ch 20 IND page-E- 2/14/03, 11:30 AM 1156
1157
Figure 20-6 Biosynthesis of the dolichol
diphosphate-linked oligosaccharide pre-
cursor to glycoproteins. The site of inhi-
bition by tunicamycin is indicated.
a
b
c
d
e
f
g
5GDP
5GDPMan
DolPPGlcNAcGlcNAcMan
5
Translocation into lumen of ER
DolPPGlcNAcGlcNAcMan
9
Glc
3
DolPP
H
2
O
P
i
DolP
Dolichol (Dol)
CDP
CTP
UDPGlcNAc
Tunicamycin
UMP
DolPPGlcNAc
DolPPGlcNAcGlcNAc
UDP
UDPGlcNAc
4DolPMan
4DolP
4GDP
4GDPMan
3DolPGlc
3DolP
3UDP
3UDPGlc

Man12Man1
Man12Man1
Man1
6
3
6
Man13(4)GlcNAc14GlcNAc
3
Glu12Glc13Glc13Man12Man12Man1
N C
O
O C
C
H
NH
H
H
2
N C
O
O C
C
H
N H
Protein
h
D. Proteoglycans and Glycoproteins
very little trimming. However, the three glucosyl
residues are usually removed by the glucosidases
present in the rough ER. Some plant glycoproteins of
the high-mannose type undergo no further processing.
Extensions and terminal elaborations. Even
before trimming is completed, addition of new resi-
dues begins within the medial cisternae.
260
In mam-
malian Golgi, N-acetylglucosamine is added first.
Galactose, sialic acid, and often fucose are then trans-
ferred from their activated forms to create such elabo-
rate oligosaccharides as that shown in Fig. 4-17. As
many as 500 glycosyltransferases, having different
specificities for glycosyl donor and glucosyl acceptor,
may be involved.
245
Extensions of the basic oligosac-
charide structure often contain polylactosamine
chains, branches with fucosyl residues, and sulfate
groups.
245,261
More than 14 sialyltransferases place
sialic acid residues, often in terminal positions, on
these cell surface oligosaccharides.
262,263
The cell wall of the yeast Saccharomyces is rich in
mannoproteins that contain 50 90% mannose.
264
The ~ 250-residue mannan chains consist of an -1,6-
linked backbone with mono-, di-, and tri-mannosyl
branches. These are attached to the same core structure
as that of mammalian oligosaccharides. All of the core
structures are formed in a similar way.
258,265
The man-
noproteins may serve as a filler to occupy spaces in
a cell wall constructed from -1,3- and -1,6-linked gly-
cans and chitin. All of the four components, including
the mannoproteins, are covalently linked together.
266
As was emphasized in Chapter 4 (pp. 186 188)
glycoproteins serve many needs in biological recogni-
tion. The N-linked oligosaccharides play a major role
in both animals and plants.
266a c
Use of mass spec-
trometry, new automated methods of oligosaccharide
synthesis,
266d
and development of new synthetic inhib-
itors
266c
are all contributing to current studies of what
is commonly called glycobiology.
The perplexing Golgi apparatus. First observed
by Camillo Golgi
267,268
in 1898, the stacked membranes,
now referred to simply as Golgi, remain somewhat
mysterious.
268 271
There are at least three functionally
distinct sets of Golgi cisternae, the cis (nearest the
nucleus), medial, and trans. An additional series of
tubules referred to as the trans Golgi network lies
between the Golgi and the cell surface and may be
the site at which lysosomal enzymes are sorted from
proteins to be secreted.
260,272
Immunochemical staining
directed toward specific glycosidases and glycosyl
transferases suggested that the trimming reactions of
glycoproteins start in the ER and continue as the pro-
teins pass outward successively from one compartment
of the Golgi to the next (Fig. 20-8). This has been the
conventional view since the 1970s. The movement of
the glycoproteins between compartments is thought
to take place in small vesicles using a rather elaborate
system of specialized proteins. Some of these coat the
vesicles
273,274
while others target the vesicles to specific
locations, e.g., the lysosomes
275
or the plasma mem-
branes where they may be secreted.
273,277
A host of
regulatory G-proteins assist these complex processes
and drive them via hydolysis of GTP.
271,278
Ch 20 IND page-E- 2/14/03, 11:30 AM 1157
1158 Chapter 20. Some Pathways of Carbohydrate Metabolism
Lysosome
Endosomes
Secretion
vesicles
Nucleus
M C T TGN CGN
Plasma
membrane
Addition of sialate
Removal of Gal
Addition of GlcNAc
Removal
of mannose
Phosphorylation of
lysosomal oligosaccharides
Peptide synthesis,
Addition of GlcNAc, Man
RER
Mannose-6-P
receptors
Figure 20-8 (A) Electron micrograph showing a transverse section through part of the Golgi apparatus of an early spermatid.
Cisternae of the ER, Golgi stacks (S), and vesicles (V) can be seen. Curved arrows point to associated tubules. Magnification
X45,000.
276
Courtesy of Y. Clermont. (B) Scheme showing functions of endoplasmic reticulum, transfer vesicles, Golgi
apparatus, and secretion vesicles in the metabolism of glycoproteins.
A B
G
G
G
M
M
M
M
M
Gn
Gn
Asn
M M
M M
G
M
M
M
M
M
Gn
Gn
Asn
M M
M M M
M
M
M
M
Gn
Gn
Asn
M M
M M
M
M
M
Gn
Gn
Asn
M M
M
M
M
Gn
Gn
Asn
M M Gn
M
M
M
Gn
Gn
Asn
Gn Gn
Gn
M
M
M
Gn
Gn
Asn
Gn
M
M
M
Gn
Gn
Asn
Gn Gn
Gn
Gal Gal
SA SA
Gal
SA
Fuc
N
HO
OH
HO
H
HO
Castanospermine
1
2
3
5
6
7
8
N
OH
HO
H
OH
Swainsonine
UDP
UDP-GlcNAc
UDP-Gal
CMP-Sia
GDP-Fuc

2 Man

Glc
HO
OH
HO
Br

Glc

UDP UDP-GlcNAc
N
HO
OH
HO
OH
H
Deoxynojirimycin
N
HO
OH
HO
OH
H
Deoxymannojirimycin
Synthesis of high
mannose glycoproteins
Bromoconduritol
Man
High mannose
oligosaccharides
Figure 20-7 Steps in trimming of the oligosaccharide precursor and synthesis of
complex N-linked oligosaccharides of mammalian glycoproteins. Sites of action of
several inhibitors are shown. Abbrevations: M, mannosyl, Gn, N-acetylglucosaminyl,
G, glucosyl residues. Structures of several inhibitors are shown.
Ch 20 IND page-E- 2/14/03, 11:30 AM 1158
1159
While most proteins synthesized in the ER follow
the exocytotic pathway through the Golgi, some are
retained in the ER and some that pass on through
the Golgi are returned to the ER.
279
In fact, such retro-
grade transport can carry some proteins taken up
by endocytosis through the plasma membrane and
through the Golgi to the ER where they undergo
N-glycosylation. Retrograde transport is essential for
recycling of plasma membrane proteins and lipids.
The forward flow of glycoproteins and membrane
components from the ER to the Golgi can be blocked
by the fungal macrocyclic lactone brefeldin A. In
cells treated with this drug, which inactivates a small
CTP-binding protein,
280
the Golgi apparatus is al-
most completely resorbed into the ER by retrograde
transport. Proteins remaining in the ER undergo
increased O-glycosylation as well as unusual types
of N-glycosylation.
Although the conventional view of flow through
the Golgi is generally accepted, it is difficult to distin-
guish it from an alternative explanation: The Golgi
compartments may move outward continuously while
retrograde transport occurs via the observed vesicles.
268,272
Some evidence for this cisternal maturation model has
been known for many years but was widely regarded
as reflecting unusual exceptions to the conventional
model. In fact, both views could be partially correct;
vesicular transport may function in both directions.
280a
High-resolution tomographic images are also altering
our view of the Golgi.
280b
The proteins of Golgi membranes are largely
integral membrane proteins and peripheral proteins
associated with the cytosolic face. Some of the integral
membrane proteins are the oligosaccharide-modifying
enzymes, which protrude into the Golgi lumen.
280c,280d
Many other proteins participate in transport,
280df
docking, membrane fusion,
280g,h
and acidification of
Golgi compartments.
280i
Many of the first studies of
vesicular transport were conducted with synaptic
vesicles and are considered in Chapter 30 (see Fig.
30-20). Other aspects of membrane fusion and trans-
port are discussed in Chapter 8. A group of special-
ized Golgi proteins, the golgins, are also present.
They are designated golgin-84, -95, -160, -245, and -376
(giantin or macrogolgin) and were identified initially
as human autoantigens (Chapter 31), appearing in
the blood of persons with autoimmune disorders
such as Sjgrens syndrome.
281,282
Another protein
of molecular mass ~130 kDa, and which appears to be
specific to the trans-Golgi network, has been found in
human serum of patients with renal vasculitis.
283
Lysosomal enzymes. Various sorting signals
are encoded within proteins. These include the pre-
viously mentioned C-terminal KDEL (mammals) or
HDEL (yeast) amino acid sequence, which serves as a
retrieval signal for return of proteins from the Golgi to
the ER (p. 521).
279,284
Other sorting signals are provided
by the varied structures of the oligosaccharides attached
to glycoproteins. These sugar clusters convey important
biological information, which is decoded in the
animal body by interaction with various lectins that
serve as receptors.
285
This often leads to endocytosis
of the glycoprotein. An example is provided by the
more than 50 proteins that are destined to become
lysosomal enzymes and which undergo phosphoryla-
tion of 6-OH groups of the mannosyl residue marked
by asterisks on the following structure. This is an
N-linked oligosaccharide that has been partially
trimmed. The phosphorylation is accomplished in
two steps by enzymes present in the cis Golgi com-
partment (Eq. 20-22) An N-acetylglucosaminyl-
phosphotransferase transfers phospho-GlcNAc units
from UDP-GlcNAc onto the 6-OH groups of mannosyl
residues. These must be recognized by the phospho-
transferase as appropriate.
286,287
Then a hydrolase
cleaves off GlcNAc.
The proteins carrying the mannose 6-phosphate
groups bind to one of two different mannose 6-P
receptors present in the Golgi membranes and are
subsequently transported in clathrin-coated vesicles
to endosomes where the low pH causes the proteins
to dissociate from the receptors, which may be recy-
cled.
288 290a
The hydrolytic enzymes are repackaged
in lysosomes. The same mannose 6-P receptors also
appear on the external surface of the plasma mem-
brane allowing many types of cells to take up lysoso-
mal enzymes that have escaped from the cell. These
proteins, too, are transported to the lysosomes. The
mannose 6-P receptors have a dual function, for they
also remove insulinlike growth factor from the circula-
tion, carrying it to the lysosomes for degradation.
287,290
Most Man 6-P groups are removed from proteins once
they reach the lysosomes but this may not always be
true.
291
Not all lysosomal proteins are recruited by the
mannose 6-P receptors. Some lysosomal membrane
proteins are sorted by other mechanisms.
292
O
OH H
3
C
OH
O
Brefeldin A
D. Proteoglycans and Glycoproteins
GlcNAc GlcNAc Man Asn
Man
Man Man
Man
Man Man Man
*
*
Ch 20 IND page-E- 2/14/03, 11:30 AM 1159
1160 Chapter 20. Some Pathways of Carbohydrate Metabolism
The hepatic asialoglycoprotein (Gal) receptor.
A variety of proteins are taken out of circulation in the
blood by the hepatocytes of the liver. Serum glycopro-
teins bearing sialic acid at the ends of their oligosac-
charides (see Fig. 20-7) have relatively long lives, but
if the sialic acid is removed by hydrolysis, the exposed
galactosyl residues are recognized by the multisubunit
asialoglycoprotein receptor.
293 295
The bound pro-
teins are then internalized rapidly via the coated pit
pathway and are degraded in the lysosomes. Other
receptors, including those that recognize transferrin,
low-density lipoprotein,
2
macroglobulin, and T
lymphocyte antigens, also depend upon interaction
with oligosaccharides.
296
E. Biosynthesis of Bacterial Cell Walls
The outer surfaces of bacteria are rich in special-
ized polysaccharides. These are often synthesized
while attached to lipid membrane anchors as indicated
in a general way in Eq. 20-20.
136,296a
One of the specific
biosynthetic cycles (Fig. 20-9) that depends upon undeca-
prenol phosphate is the formation of the peptidoglycan
(murein) layer (Fig. 8-29) of both gram-negative and
gram-positive bacterial cell walls. Synthesis begins
with attachment of L-alanine to the OH of the lactyl
O
O
HO
O
O
OH
H
GlcNAc O P O
O
O
P O
O
O
Uridine

UDP-GlcNAc
O
O
HO
O
O
OH
P
O
O O
GlcNAc
H
2
O

O
O
HO
O
O
OH
P
HO
HO O
H
2
O
GlcNAc
(20-22)
group of UDP-N-acetylmuramic acid in a typical ATP-
requiring process (Fig. 20-9, step a).
297
Next D-glutamic
acid, meso-diaminopimelate (Fig. 8-29), or L-lysine,
and D-alanyl-D-alanine are joined in sequence, each
in another ATP-requiring step.
298 301d
The entire unit
assembled in this way is transferred to undecaprenol
phosphate with creation of a pyrophosphate linkage
(step e). An N-acetylglucosamine unit is added by
action of another transferase (step f), and in an ATP-
requiring process ammonia is sometimes added to cap
the free -carboxyl group of the D-glutamyl residue
(step g). In Staphylococcus aureus and related gram-
positive bacteria five glycyl units are also added, each
from a molecule of glycyl-tRNA (green arrows in
Fig. 20-9). The completely assembled repeating unit,
together with the connecting peptide chain needed
in the crosslinking reaction, is transferred onto the
growing chain (step h). As in formation of dextrans,
growth is by insertion of the repeating unit at the
reducing end of the chain. The polyprenyl diphos-
phate is released, and the cycle is completed by the
action of a pyrophosphatase (step i). This step is
blocked by bacitracin, an antibiotic which forms an
unreactive complex with the polyprenyl diphosphate
carrier. Completion of the peptidoglycan requires
crosslinking. This is accomplished by displacement
of the terminal D-alanine of the pentapeptide by attack
by the NH
2
group of the diaminopimelate or lysine
or other diamino acid (see also Fig. 8-29).
301e
Because the peptidoglycan layer must resist swell-
ing of the bacteria in media of low osmolarity, it must
be strong and must enclose the entire bacterium. At
the same time the bacterium must be able to grow in
size and also to divide. For these reasons bacteria
must continuously not only synthesize peptidoglycan
but also degrade it.
302,303
The latter is accomplished
by hydrolytic cleavage using cell wall enzymes to the
N-acetylglucosamine-anhydro-N-acetylmuramate-
tripeptide (GlcNAc-1,6-anhydro-MurNAc-L-Ala-D-Glu-
A
2
pm) fragment.
304,305
A hydrolase cuts the peptide
bridge.
305a
This process is probably essential to forma-
tion of new growing points for expansion of the murein
layer. Most of the peptide fragments that are released
in the periplasm are transported back into the cytosol.
N
O
H
O
N
CH
3
H
H
H
+
NH
2
d-Alanyl-d-alanine group
CH
3
H COO

Amino group
from diamino acid
in another chain
Ch 20 IND page-E- 2/14/03, 11:30 AM 1160
1161
The anhydroMurNAc is removed, and new UDP-
MurNAc and D-Ala-D-Ala units are added salvaging
the tripeptide unit. The repaired UDP-MurNA-
pentapeptide can then reenter the biosynthetic
pathway (Fig. 20-9).
The O-antigens and lipid A. A cluster of sugar
units of specific structure makes up the repeating unit
of the O-antigen of Salmonella. The many structural
variations in this surface polysaccharide account for the
over two thousand serotypes of Salmonella (p. 180).
121,306
As is illustrated in Fig. 8-30, the O-antigen is a repeating
block polymer that is attached to a complex lipopoly-
saccharide core and a hydrophobic membrane anchor
known as lipid A (Figs. 8-28 and 8-30).
307308a
Lipid A
and the attached core and O-antigen are synthesized
inside the bacterial cell by enzymes found in the cyto-
plasmic membrane.
309
The complete lipopolysaccharide
units are then translocated from the inner membrane
to the outer membrane of the bacteria. The synthesis
of the O-antigen is understood best. Consider the
following group E3 antigen, where Abe is abequose
(Fig. 4-15) and Rha is rhamnose:
Abe1

4
( 6 Gal1 6 Man14 Rha1)
n
Assembly of this repeating unit begins with the transfer
of a phosphogalactosyl unit from UDP-Gal to the phospho
group of the lipid carrier undecaprenol phosphate.
The basic reaction cycle is much like that in Fig. 20-9
for assembly of a peptidoglycan.
The oligosaccharide repeating unit of the O-antigen
is constructed by the consecutive transferring action of
three more transferases. For the antigen shown above,
one enzyme transfers a rhamnosyl unit, another a
mannosyl unit, and another an abequosyl unit from
the appropriate sugar nucleotides. Then the entire
growing O-antigen chain, which is attached to a second
molecule of undecaprenol diphosphate, is transferred
onto the end of the newly assembled oligosaccharide
unit. In effect, the newly formed oligosaccharide is
inserted at the reducing end of the growing chain just
as in Fig. 20-9. Elongation continues by the transfer of
the entire chain onto yet another tetrasaccharide unit.
As each oligosaccharide unit is added, an undecaprenol
diphosphate unit is released and a phosphatase cleaves
off the terminal phospho group to regenerate the
original undecaprenol phosphate carrier. When the
O-antigen is long enough, it is attached to the rest of
the lipopolysaccharide.
The lipid A anchor is also based on a carbohy-
drate skeleton. Its assembly in E. coli,
307
which
requires nine enzymes, is depicted in Fig. 20-10. N-
Acetylglucosamine 6-P is acylated at the 3-position
310
and after deacetylation
311
at the 2-position. As
shown in this figure, acylation is accomplished by
transfer of hydroxymyristoyl groups from acyl car-
rier protein (ACP). Two molecules of the resulting
UDP-2,3-diacyl-GlcN are then joined via the reac-
tions shown to give the acylated disaccharide
precursor to lipid A. Stepwise transfer of KDO,
L-glycero-D-manno-heptose, and other monosaccharide
units from the appropriate sugar nucleotides and
further acylation follows (Fig. 20-10). The assembled
O-antigen chain is transferred from undecaprenol
diphosphate onto the lipopolysaccharide core. This
apparently occurs on the periplasmic surface of the
plasma membrane. If so, the core lipid domain must
be flipped across the plasma membrane before the
O-antigen chain is attached.
312
Less is known about the
transport of the completed lipopolysaccharide across
the periplasmic space and into the outer membrane.
The core structures of the lipopolysaccharides vary
from one species to another or even from one strain of
bacteria to another. All three domains (lipid A, core,
and O-antigen) contribute to the antigenic properties
of the bacterial surface
313
and to the virulence of the
organism.
313,314
Nitrogen-fixing strains of Rhizobium
require their own peculiar lipopolysaccharides for
successful symbiosis with a host plant.
315
However,
there are some features common to most lipopolysac-
charides. Two to three residues of KDO are usually
attached to the acylated diglucosamine anchor, and
these are often followed by 3 4 heptose rings.
316318
The structure of the inner core regions of a typical
lipopolysaccharide from E. coli is indicated in Fig. 8-30.
The complete structure of the lipopolysaccharide from
a strain of Klebsiella is shown at the top of the next
page.
319
Here L, D-Hepp is D-manno-heptopyranose
and D, D-Hepp is D-glycero-D-manno-heptose. As in this
case, the outer core often contains several different
hexoses. The lipopolysaccharide of Neisseria menin-
gititis has sialic acid at the outer end.
320
However, the
major virulence factor for this organism, which is a
leading cause of bacterial meningitis in young children,
is a capsule of poly(ribosyl)ribitol phosphate that
surrounds the cell.
321
Haemophilus influenzae, a common
cause of ear infections and meningitis in children, has
no O-antigen but a more highly branched core oligo-
saccharide than is present in E. coli.
321a
Legionella has
its own variations.
321b
Gram-positive bacteria. Although their outer
coatings are extremely varied, all gram-positive bacteria
have a peptidoglycan similar to that of gram-negative
bacteria but often containing the intercalated penta-
glycine bridge indicated in Fig. 8-29. However, the
peptidoglycan of gram-positive bacteria is 2050 nm
thick, as much as ten times thicker than that of E. coli.
Furthermore, the peptidoglycan is intertwined with
the anionic polymers known as teichoic acids and
E. Biosynthesis of Bacterial Cell Walls
Ch 20 IND page-E- 2/14/03, 11:30 AM 1161
1162 Chapter 20. Some Pathways of Carbohydrate Metabolism
b
c
d
e
f
g
MurNAc-P-P-undecaprenol
pentapeptide
UMP
UDP-GlcNAc
UDP
GlcNAc-MurNAc-P-P
pentapeptide
NH
4
+
ATP
Growing
peptidoglycan
Undecaprenol-P-P
(C
55
)
P
i
H
2
O
Undecaprenol-P
GlcNAc-MurNAc-P-P-undecaprenol
pentapeptide-amide
pentaglycine
This pentaglycine peptide
bridge is formed by
Staphylococcus aureus
(see Fig. 8-29)
UDP-MurNAc-l-Ala-d-Glu-meso-A
2
pm-d-Ala-d-Ala
(UDP-MurNAc-pentapeptide)
Completed repeating unit
GlcNAc-MurNAc-P-P-undecaprenol
pentapeptide-amide (see Fig. 8-29)
d-Ala
Crosslinked murein layer
Peptidoglycan
containing one more
repeating unit
h
5-glycyl-tRNA
5 tRNA
ATP
d-Glu
l-Glu
ATP
O
NH
HO
O
C O
CH
3
O UDP C
NH
COO
H
3
C
H
H
3
C
H
O
UDP-MurNAc

a
UDP-MurNAc
l-Ala
ATP
Also formed by salvage
pathway (see text)
ATP
meso-2,6-Diaminopimelate (mesoA
2
pm or l-Lys)
i
d-Ala-d-Ala
d-Ala
l-Ala
ATP c
d-Ala
Site of binding
of vancomycin
Bacitracin
Figure 20-9 Biosynthesis of bacterial
peptidoglycans. See Fig. 8-29 for details
of the peptidoglycan structures. Green
arrows show alternative route used by
gram-positive bacteria.
GalpA1 6Glcp1 4l,dHepp1
3

d,d-Hepp1 6GlcpN1 4GalpA1 3l,d-Hepp1


7

d,d-Hepp1
2

l,d-Hepp1
d,d-Hepp1
2

d,d-Hepp1
2

Inner core
Outer core
5DO2 6GlcpN1 6GlcpN1 P
4 4

DO2 P
4
DO2
Anchor after removal
of fatty acyl groups
Lipopolysaccharide from Klebsiella
319
Green shade indicates partial presence.
Ch 20 IND page-E- 2/14/03, 11:30 AM 1162
1163
O
O
OH
HO
HO
C O
O

O
HO
OH
HO
HO
C O
O

O
O
O
P HO
HO
O
O
O
O
O
NH
O
O
O
O
O
HO
O
HO
NH
O
HO
O P
O
OH
OH
O
Lauryl and
myristoyl-ACP
ACP
CMP
P
O
O
HO
OH
HO
HO
C O
O

O Cytidine
O O

ATP
CMP
O
O
HO
HO
O
HO
NH
O
HO
O P
O
O

O P
O
O Uridine
O

O
O
HO
HO
O
HO
NH
O
HO
O P
O

O
OH
O
O
O
O
O
HO
NH
O
P
O
P
O
O Uridine
O
O O

H
P HO
O
HO
H
2
O
UMP
UMP
R-3-Hydroxymyristoyl-ACP
Acetate
ACP
H
2
O
UDP-GlcNAc
ACP
O
O
HO
HO
O
HO
NH
O
HO
O
O
HO
O
HO
NH
O
HO
O P
O
OH
O
O

O
O
O
P O
HO
O
HO
O
O
NH
O
O
O
O
HO
O
HO
NH
O
HO
O P
O
OH
O
O

O
O
OH
HO
HO
C O
O

O
HO
OH
HO
HO
C O
O

O
O
O
P O
HO
O
O
O
O
NH
O
O
O
O
HO
O
HO
NH
O
HO
O P
O
OH
O
O

Lipid A
(Enterotoxin)
Outer core
sugars
O-Antigen
Mature lipopolysaccharide
CMP-KDO

O
Figure 20-10 Proposed biosynthetic route for synthesis of lipid A and the mature lipopolysaccharide of the E. coli cell wall.
After C. R. H. Raetz et al.
307
E. Biosynthesis of Bacterial Cell Walls
Ch 20 IND page-E- 2/14/03, 11:30 AM 1163
1164 Chapter 20. Some Pathways of Carbohydrate Metabolism
BOX 20-G PENICILLINS AND RELATED ANTIBIOTICS
O
N
H H
N
O
S
CH
3
CH
3
COO
H H
H
NH
2
in ampicillin
Penicillin G
(Benzylpenicillin)

BH
+
various
cations
Many organisms produce chemical substances
that are toxic to other organisms. Some plants
secrete from their roots or leaves compounds that
block the growth of other plants. More familiar to
us are the medicinal antibiotics produced by fungi
and bacteria. The growth inhibition of one kind of
organism upon another was well known in the last
century, e.g., as reported by Tyndall
a,b
in 1876. The
beginning of modern interest in the phenomenon is
usually attributed to Alexander Fleming, who, in
1928, noticed the inhibition of growth of staphylo-
cocci by Penicillium notatum. His observation led
directly to the isolation of penicillin, which was first
used on a human patient in 1930. The early history
as well as the subsequent purification, characteriza-
tion, synthesis, and development as the first major
antibiotic has been recorded in numerous books and
articles.
c i
During the same time period, Rene
Dubos isolated the peptide antibiotics gramicidin
and tyrocidine.
j
A few years later actinomycin
(Box 28-A) and streptomycin (Box 20-B) were
isolated from soil actinomycetes (streptomyces) by
Waksman, who coined the name antibiotic for
these compounds. Streptomycin was effective
against tuberculosis, a finding that helped to stimu-
late an intensive search for additional antibacterial
substances. Since that time, new antibiotics have
been discovered at the rate of more than 50 a year.
More than 100 are in commercial production.
Major classes of antibiotics include more than
200 peptides such as the gramicidins, bacitracin,
tyrocidines and valinomycin (Fig. 8-22)
k
; more than
150 penicillins, cephalosporins, and related com-
pounds; tetracyclines (Fig. 21-10); the macrolides,
large ring lactones such as the erythromycins (Fig.
21-11); and the polyene antibiotics (Fig. 21-10).
Penicillin was the first antibiotic to find practi-
cal use in medicine. Commercial production began
in the early 1940s and benzylpenicillin (penicillin
G), one of several natural penicillins that differ in
the R group boxed in the structure above, became
one of the most important of all drugs. Most effec-
tive against gram-positive bacteria, at higher con-
centrations it also attacks gram-negative bacteria
including E. coli. The widely used semisynthetic
penicillin ampicillin (R = D--aminobenzyl) attacks
both gram-negative and gram-positive organisms.
It shares with penicillin extremely low toxicity but
some danger of allergic reactions. Other semisyn-
thetic penicillins are resistant to -lactamases, en-
zymes produced by penicillin-resistant bacteria
which cleave the four-membered -lactam ring of
natural penicillins and inactivate them.
Closely related to penicillin is the antibiotic
cephalosporin C. It contains a D--aminoadipoyl
side chain, which can be replaced to form various
semisynthetic cephalosporins. Carbapenems have
similar structures but with CH
2
replacing S and
often a different chirality in the lactam ring.
These and other related -lactams are medically
important antibacterial drugs whose numbers are
increasing as a result of new isolations, synthetic
modifications, and utilization of purified biosyn-
thetic enzymes.
c,l,m
How do antibiotics act? Some, like penicillin,
block specific enzymes. Peptide antibiotics often
form complexes with metal ions (Fig. 8-22) and
disrupt the control of ion permeability in bacterial
membranes. Polyene antibiotics interfere with
proton and ion transport in fungal membranes.
Tetracyclines and many other antibiotics interfere
directly with protein synthesis (Box 29-B). Others
intercalate into DNA molecules (Fig. 5-23; Box
28-A). There is no single mode of action. The
search for suitable antibiotics for human use con-
sists in finding compounds highly toxic to infective
organisms but with low toxicity to human cells.
Penicillin kills only growing bacteria by pre-
venting proper crosslinking of the peptidoglycan
OOC N
O NH
3
N
O
S
O
O
CH
3
COOH
H H
Cephalosporin C
H

+
N
S
O
N NH
2
H H
HO
H
H
3
C
H
COO
Imipenem, a carbapenem antibiotic of last resort

+
Ch 20 IND page-E- 2/14/03, 11:30 AM 1164
1165
BOX 20-G (continued)
layer of their cell walls. An amino group from a
diamino acid in one peptide chain of the peptido-
glycan displaces a D-alanine group in a transpepti-
dation (acyltransferase) reaction. The transpeptidase
is also a hydrolase, a DD-carboxypeptidase. Penicil-
lins are structural analogs of D-alanyl-D-alanine and
bind to the active site of the transpeptidase.
l,n p
The -lactam ring of penicillins is unstable, making
penicillins powerful acylating agents. The trans-
peptidase apparently acts by a double displacement
mechanism, and the initial attack of a nucleophilic
serine hydroxyl group of the enzyme on penicillin
bound at the active site leads to formation of an
inactivated, penicillinoylated enzyme.
q,r
More than
one protein in a bacterium is derivatized by penicillin.
s
Therefore, more than one site of action may be
involved in the killing of bacterial cells.
Several classes of -lactamases, often encoded
in transmissible plasmids, have spread worldwide
rapidly among bacteria, seriously decreasing the
effectivenss of penicillins and other -lactam anti-
biotics.
t y
Most -lactamases (classes A and C) contain
an active site serine and are thought to have evolved
from the DD transpeptidases, but the B type
y
has a
catalytic Zn
2+
. The latter, as well as a recently
discovered type A enzyme,
z
hydrolyze imipenem,
currently one of the antibiotics of last resort used
to treat infections by penicillin-resistant bacteria.
Some -lactam antibiotics are also powerful inhibitors
of -lactamases.
u,aa,bb
These antibiotics may also
have uses in inhibition of serine proteases
cc,dd
such
as elastase. Some antibiotic-resistant staphylococci
produce an extra penicillin-binding protein that
protects them from beta lactams.
ee
Because of anti-
biotic resistance the isolation of antibiotics from
mixed populations of microbes from soil, swamps,
and lakes continues. Renewed efforts are being
a
Tyndall, J. (1876) Phil. Trans. Roy. Soc. London B 166, 27
b
Reese, K. M. (1980) Chem. Eng. News, Sept. 29, p64
c
Abraham, E. P. (1981) Sci. Am. 244 (Jun), 76 86
d
Sheehan, J. C. (1982) The Enchanted Ring: The Untold Story of
Penicillin, MIT Press, Cambridge, Massachusetts
e
Hobby, G. L. (1985) Penicillin: Meeting the Challenge, Yale Univ. Press,
f
Chain, B. (1991) Nature (London) 353, 492 494
g
Williams, T. I. (1984) Howard Florey: Penicillin and After, Oxford
Univ. Press, London
h
Moberg, C. L. (1991) Science 253, 734 735
i
Nayler, J. H. C. (1991) Trends Biochem. Sci. 16, 195 197
j
Crease, R. P. (1989) Science 246, 883 884
k
Gabay, J. E. (1994) Science 264, 373 374
l
Nayler, J. H. C. (1991) Trends Biochem. Sci. 16, 234 237
m
Mourey, L., Miyashita, K., Swarn, P., Bulychev, A., Samama,
J.-P., and Mobashery, S. (1998) J. Am. Chem. Soc. 120, 9382 9383
n
Yocum, R. R., Waxman, D. J., and Strominger, J. L. (1980) Trends
Biochem. Sci. 5, 97 101
o
Kelly, J. A., Moews, P. C., Knox, J. R., Frere, J.-M., and Ghuysen,
J.-M. (1982) Science 218, 479 480
p
Kelly, J. A., and Kuzin, A. P. (1995) J. Mol. Biol. 254, 223 236
q
Englebert, S., Charlier, P., Fonz, E., Toth, Y., Vermeire, M., Van
Beeumen, J., Grandchamps, J., Hoffmann, K., Leyh-Bouille, M.,
Nguyen-Distche, M., and Ghuysen, J.-M. (1994) J. Mol. Biol.
241, 295 297
r
Kuzin, A. P., Liu, H., Kelly, J. A., and Knox, J. R. (1995) Biochem-
istry 34, 9532 9540
s
Thunnissen, M. M. G. M., Fusetti, F., de Boer, B., and Dijkstra,
B. W. (1995) J. Mol. Biol. 247, 149 153
t
Siemers, N. O., Yelton, D. E., Bajorath, J., and Senter, P. D. (1996)
Biochemistry 35, 2104 2111
u
Swarn, P., Massova, I., Bellettini, J. R., Bulychev, A., Mavey-
raud, L., Kotra, L. P., Miller, M. J., Mobashery, S., and Samama,
J.-P. (1999) J. Am. Chem. Soc. 121, 5353 53595
v
Guillaume, G., Vanhove, M., Lamotte-Brasseur, J., Ledent, P.,
Jamin, M., Joris, B., and Frre, J.-M. (1997) J. Biol. Chem. 272,
5438 5444
w
Adediran, S. A., Deraniyagala, S. A., Xu, Y., and Pratt, R. F.
(1996) Biochemistry 35, 3604 3613
x
Brown, R. P. A., Aplin, R. T., and Schofield, C. J. (1996) Biochem-
istry 35, 12421 12432
y
Carfi, A., Pares, S., Due, E., Galleni, M., Duez, C., Frre, J. M.,
and Dideberg, O. (1995) EMBO J. 14, 4914 4921
z
Swarn, P., Maveyraud, L., Raquet, X., Cabantous, S., Duez, C.,
Pdelacq, J.-D., Mariotte-Boyer, S., Mourey, L., Labia, R.,
Nicolas-Chanoine, M.-H., Nordmann, P., Frre, J.-M., and
Samama, J.-P. (1998) J. Biol. Chem. 273, 26714 26721
aa
Trehan, I., Beadle, B. M., and Shoichet, B. K. (2001) Biochemistry
40, 7992 7999
bb
Swarn, P., Golemi, D., Cabantous, S., Bulychev, A.,
Maveyraud, L., Mobashery, S., and Samama, J.-P. (1999)
Biochemistry 38, 9570 9576
cc
Wilmouth, R. C., Kassamally, S., Westwood, N. J., Sheppard, R.
J., Claridge, T. D. W., Aplin, R. T., Wright, P. A., Pritchard, G. J.,
and Schofield, C. J. (1999) Biochemistry 38, 7989 7998
dd
Taylor, P., Anderson, V., Dowden, J., Flitsch, S. L., Turner, N. J.,
Loughran, K., and Walkinshaw, M. D. (1999) J. Biol. Chem. 274,
24901 24905
ee
Zhang, H. Z., Hackbarth, C. J., Chansky, K. M., and Chambers,
H. F. (2001) Science 291, 1962 1965
Serine OH group of transpeptidase.
N
O
COO H
H
N
O
H
N CH
3
H
H
N
H
3
C
O
O
H COO
H

-Lactam
Structure of penicillin
and related antibiotics

OH
Bonds in d-alanyl-d-alanine
group of peptidoglycan or in
penicillins are cleaved as
indicated by dashed lines
d-Alanyl-d-alanine group
made to find new targets for antibacterial drugs
and to synthesize new compounds in what will
evidently be a never-ending battle. We also need
better antibiotics against fungi and protozoa.
E. Biosynthesis of Bacterial Cell Walls
Ch 20 IND page-E- 2/14/03, 11:30 AM 1165
1166 Chapter 20. Some Pathways of Carbohydrate Metabolism
BOX 20-H ANTIBIOTIC RESISTANCE AND VANCOMYCIN
Cl
O O
O Cl
OH
H
H
N
O
O
N
H
N
CH
3
O
N
H
O
N
OH
OH HO
HN
O
HOOC
H
H
HO
N
H
O
H
H
NH
2
O
O
CH
3
H
3
C
O
OH
O
OH
OH
HO
NH
2
Vancomycin
CH
3
CH
3
H
H H
H
H
H
As antibiotics came into widespread use, an
unanticipated problem arose in the rapid develop-
ment of resistance by bacteria. The problem was
made acute by the fact that resistance genes are easily
transferred from one bacterium to another by the
infectious R-factor plasmids.
a d
Since resistance
genes for many different antibiotics may be carried
on the same plasmid, super bacteria, resistant to a
large variety of antibiotics, have developed, often in
hospitals.
The problem has reached the crisis stage, perhaps
most acutely for tuberculosis. Drug-resistant Myco-
bacteria tuberculosis have emerged, especially, in
patients being treated for HIV infection (see Box 21-C).
Mechanisms of resistance often involve inactivation
of the antibiotics. Aminoglycosides
such as streptomycin, spectinomycin,
and kanamycin (Box 20-B) are inacti-
vated by en-zymes catalyzing phos-
phorylation or adenylylation of
hydroxyl groups on the sugar rings.
e
g
Penicillin and related antibiotics are
inactivated by -lactamases (Box 20-
G). Chloramphenicol (Fig. 25-10) is
inactivated by acylation on one or
both of the hydroxyl groups.
What is the origin of the drug
resistance factors? Why do genes for
inactivation of such unusual mole-
cules as the antibiotics exist widely in
nature? Apparently the precursor to
drug resistance genes fulfill normal
biosynthetic roles in nature. An
antibiotic-containing environment,
such as is found naturally in soil,
leads to selection of mutants of such
genes with drug-inactivating proper-
ties. Never-theless, it is not entirely
clear why drug resistance factors have appeared so
promptly in the population. Overuse of antibiotics
in treating minor infections is one apparent cause.
h,i
Another is probably the widespread use on farms.
j
A nationwide effort to decrease the use of erythro-
mycin in Finland had a very favorable effect in
decreasing the incidence of erythromycin-resistant
group A streptocci.
h
Because of the rapid development of resistance,
continuous efforts are made to alter antibiotics by
semisynthesis (see Box 20-G) and to identify new
targets for antibiotics or for synthetic antibacterial
compounds.
d
An example is provided by the
discovery of vancomycin. Like the penicillins,
this antibiotic interferes with bacterial cell wall
teichuronic acids (p. 431). Both proteins and neutral
polysaccharides, sometimes covalently bound, may
also be present.
303
Like peptidoglycans, teichoic and
teichuronic acids are assembled on undecaprenyl
phosphates
303
or on molecules of diacylglycerol.
322
Either may serve as an anchor. A linkage unit may
be formed by transfer of several glycosyl rings onto an
anchor unit. For example, in synthesis of ribitol teichoic
acid sugar rings are transferred from UDP-GlcNAc,
UDP-ManNAC, and CDP-Gal to form the following
linkage unit:
LipidPP GlcNAc ManNAc (P-Gal)
2,3
Then many ribitol phosphate units are added by trans-
fer from CDP-ribitol. Finally, the chain is capped by
transfer of a glucose from UDP-Glc. Lipoteichoic acids
often carry covalently linked D-alanine in ester linkage,
altering the net electrical charge on the cell surface.
322a
The completed teichoic acid may then be transferred
to a peptidoglycan, releasing the lipid phosphate for
reuse.
303
Glycerol teichoic acid may be formed in a
similar fashion.
322
Teichuronic acids arise by alternate
transfers of P-GalNAc from UDP-GalNAc and of GlcA
from UDP-GlcA.
303
Gram-positive bacteria often carry surface proteins
that interact with host tissues in establishing human
infections. Protein A of Staphylococcus is a well-known
Ch 20 IND page-E- 2/14/03, 11:31 AM 1166
1167
BOX 20-H (continued)
synthesis but does so by binding tightly to the
D-alanyl-D-alanine termini of peptidoglycans that
are involved in crosslinking (Fig. 8-29, Fig. 20-9).
k,l
Like penicillin, vancomycin prevents crosslinking
but is unaffected by -lactamases. Initially bacteria
seemed unable to develop resistance to vancomycin,
and this antibiotic was for 25 years the drug of
choice for -lactam resistant streptococci or staphyl-
ococci. However, during this period bacteria carry-
ing a plasmid with nine genes on the transposon
Tn1546 (see Fig. 27-30) developed resistance to van-
comycin and were spread worldwide.
l
Vancomycin-
resistant bacteria are able to sense the presence
of the antibiotic and to synthesize an altered
D-alanine:D-alanine ligase, the enzyme that joins
two D-alanine molecules in an ATP-dependent
reaction to form the D-alanyl-D-alanine needed to
permit peptidoglycan crosslinking (Fig. 20-9, step c).
The altered enzyme adds D-lactate rather than
D-alanine providing an OH terminus in place of
NH
3
+
. This prevents the binding of vancomycin
l,m
and allows crosslinking of the peptidoglycan via
depsipeptide bonds. Another gene in the transpo-
son encodes an oxoacid reductase which supplies
the D-lactate.
l
A different resistant strain synthesizes
a D-Ala-D-Ser ligase.
n
High-level vancomycin resis-
tance is not attained unless the bacteria also synthe-
size a D-alanyl-D-alanine dipeptidase.
o
The D-Ala-D-Ala ligase does provide yet another
attractive target for drug design.
p,q
Still another is
the D-Ala-D-Ala adding enzyme (Fig. 20-8, step d;
encoded by the E. coli MurF gene).
r,s
Strategies for
combatting vancomycin resistance include synthesis
of new analogs of the antibiotic
t,u
and simultaneous
administration of small molecules that catalyze
cleavage of the D-Ala-D-lactate bond formed in cell
wall precursors of resistant bacteria.
v
Another possibility is to use bacteriophages
a
Davies, J. (1994) Science 264, 375 382
b
Benveniste, R., and Davies, J. (1973) Ann. Rev. Biochem. 42, 471
506
c
Clowes, R. C. (1973) Sci. Am. 228(Apr), 19 27
d
Neu, H. C. (1992) Science 257, 1064 1073
e
McKay, G. A., and Wright, G. D. (1996) Biochemistry 35, 8680
8685
f
Thompson, P. R., Hughes, D. W., Cianciotto, N. P., and Wright,
G. D. (1998) J. Biol. Chem. 273, 14788 14795
g
Cox, J. R., and Serpersu, E. H. (1997) Biochemistry 36, 2353 2359
h
Seppl, H., Klaukka, T., Vuopio-Varkila, J., Muotiala, A.,
Helenius, H., Lager, K., and Huovinen, P. (1997) N. Engl. J. Med.
337, 441 446
i
Gorbach, S. L. (2001) N. Engl. J. Med. 345, 1202 1203
j
Witte, W. (1998) Science 279, 996 997
k
Sheldrick, G. M., Jones, P. G., Kennard, O., Williams, D. H., and
Smith, G. A. (1978) Nature (London) 271, 223 225
l
Walsh, C. T. (1993) Science 261, 308 309
m
Sharman, G. J., Try, A. C., Dancer, R. J., Cho, Y. R., Staroske, T.,
Bardsley, B., Maguire, A. J., Cooper, M. A., OBrien, D. P., and
Williams, D. H. (1997) J. Am. Chem. Soc. 119, 12041 12047
n
Park, I.-S., Lin, C.-H., and Walsh, C. T. (1997) Proc. Natl. Acad.
Sci. U.S.A. 94, 10040 10044
o Aroz, R., Anhalt, E., Ren, L., Badet-Denisot, M.-A.,
Courvalin, P., and Badet, B. (2000) Biochemistry 39, 15971 15979
p
Fan, C., Moews, P. C., Walsh, C. T., and Knox, J. R. (1994)
Science 266, 439 443
q
Fan, C., Park, I.-S., Walsh, C. T., and Knox, J. R. (1997) Biochem-
istry 36, 2531 2538
r
Duncan, K., van Heijenoort, J., and Walsh, C. T. (1990) Biochem-
istry 29, 2379 2386
s
Anderson, M. S., Eveland, S. S., Onishi, H. R., and Pompliano,
D. L. (1996) Biochemistry 35, 16264 16269
t
Walsh, C. (1999) Science 284, 442 443
u
Ge, M., Chen, Z., Onishi, H. R., Kohler, J., Silver, L. L., Kerns,
R., Fukuzawa, S., Thompson, C., and Kahne, D. (1999) Science
284, 507 511
v
Chiosis, G., and Boneca, I. G. (2001) Science 293, 1484 1487
w
Stone, R. (2002) Science 298, 728731
example. After synthesis in the cytoplasm, it enters
the secretory pathway. An N-terminal hydrophobic
leader sequence and a 35-residue C-terminal sorting
signal guide it to the correct destination. There a free
amino group of an unlinked pentaglycyl group of the
peptidoglycan carries out a transamidation reaction
with an LPXTG sequence in the proteins, cutting the
chain between the threonine and glycine residues, and
anchoring the protein A to the peptidoglycan.
323
Group A streptococci, which are serious human
pathogens, form -helical coiled-coil threads whose
C termini are anchored in the cell membrane. They
protrude through the peptidoglycan layers and provide
a hairlike layer around the bacteria. A variable region
at the N termini provides many antigens, some of
which escape the hosts immune system allowing
infection to develop.
324
Group B streptococci form
carbohydrate antigens linked to teichoic acid.
325
Streptococci, which are normally present in the mouth,
utilize their carbohydrate surfaces as receptors for
adhesion, allowing them to participate in formation
of dental plaque.
326
Cell walls of mycobacteria are composed of a
peptidoglycan with covalently attached galactan
chains. Branched chains of arabinan, a polymer of
the furanose ring form of arabinose with covalently
attached mycolic acids, are glycosidically linked to the
galactan.
327
Shorter glycopeptidolipids, containing
E. Biosynthesis of Bacterial Cell Walls
directly as antibacterial medicines. This approach
was introduced as early as 1919 and has enjoyed
considerable success. It is now regarded as a prom-
ising alternative to the use of antibiotics in many
instances.
w
Ch 20 IND page-E- 2/14/03, 11:31 AM 1167
1168 Chapter 20. Some Pathways of Carbohydrate Metabolism
modified glucose and rhamnose rings as well as fatty
acids, contribute to the complexity of mycobacterial
surfaces.
328
These examples describe only a small sample of
the great diversity of cell coats found in the prokary-
otic world. Some bacteria also provide themselves
with additional protection in the form of external
sheaths of crystalline arrays of proteins known as
S-layers.
329
F. Biosynthesis of Eukaryotic Glycolipids
Glycolipids may be thought of as membrane lipids
bearing external oligosaccharides. In this sense, they
are similar to glycoproteins both in location and in
biological significance. Like the glycoproteins, glyco-
lipids are synthesized in the ER, then transported into
the Golgi apparatus and eventually outward to join the
outer surface of the plasma membrane. Some glyco-
lipids are attached to proteins by covalent linkage.
1. Glycophosphatidylinositol (GPI) Anchors
More than 100 different human proteins are
attached to phosphatidylinositol anchors of the type
shown in Fig. 8-13.
329a
Similar anchors are prevalent
in yeast and in protozoa including Leishmania and
Trypanosoma,
330 334b
and Plasmodium
334c
where they
often bind major surface proteins to the plasma
membrane.
335
They are also found in mycobacteria.
336
The structures of the hydrophobic anchor ends are
all similar.
337
Two or three fatty acyl groups hold the
molecule to the bilayer. Variations are found in the
attached glycan portion, both in the number of sugar
rings and in the structures of the covalently attached
phosphoethanol-amine groups.
337 339
A typical assem-
bly pathway is shown in Eq. 20-23. The first step (step
a in Eq. 20-23), the transfer of an N-acetylglucosamine
residue to phosphatidylinositol, is surprisingly com-
plex, requiring at least three proteins.
340
The hydro-
lytic deacetylation (step b) helps to drive the synthetic
process. Step c provides dolichol-P-mannose for the
GPI anchors as well as for glycoproteins. The phos-
phoethanolamine part of the structure is added from
phosphatidylethanolamine, apparently via direct
nucleophilic displacement.
333
In this way the C termi-
nus of the protein forms an amide linkage with the
NH
2
group of ethanolamine in the GPI anchor. An-
other unexpected finding was that this completed
anchor unit undergoes remodeling during which the
fatty acyl chains of the original phosphatidylinositol
are replaced by other fatty acids.
339,339a
2. Cerebrosides and Gangliosides
These two groups of glycolipids are derived from
the N-acylated sphingolipids known as ceramides.
Some biosynthetic pathways from sphingosine to
these substances are indicated in Fig. 20-11. Acyl,
glycosyl, and sulfo groups are transferred from appro-
priate derivatives of CoA, CDP, UDP, CMP, and from
PAPS to form more than 40 different gangliosides.
341,342
The biosynthesis of a sphingomyelin is also shown in
this scheme but is discussed in Chapter 21.
Each biosynthetic step in Fig. 20-11 is catalyzed
by a specific transferase. Most of these enzymes are
present in membranes of the ER and the Golgi.
343 346
Furthermore, the sequence by which the transferases
act may not always be fixed, and a complete biosyn-
thetic scheme would be far more complex than is
shown in the figure. For example, one alternative
a
b
d
e
f
PI (phosphatidylinositol)
GlcN-PI
H
2
O
Acetate
GlcNAc-PI
UDP-Glc NAc
UDP
Man
3
GlcN-PI
Dolichol-P-Man
Three steps
Dolichol-P
Ethanolamine-P-Man
3
GlcN-PI
Phosphatidylethanolamine
Diacylglycerol
Translocation across
ER membrane
Transamidase
Protein
Complete GPI-anchored protein
g
c
GDP-Man
GDP
Dolichol-P
(20-23)
H OH
CH
2
OH
NH H
C
O
R
Long-chain acyl group
Carbohydrate chain is added here in the
glycolipids; phosphocholine in sphingomyelin
A ceramide
Ch 20 IND page-E- 2/14/03, 11:31 AM 1168
1169
sequence is the synthesis of galactosyl ceramide by
transfer of galactose to sphingosine followed by acyla-
tion (Eq. 20-24). However, the pathway shown in Fig.
20-11 is probably more important.
G. The Intracellular Breakdown of
Polysaccharides and Glycolipids
The attention of biochemists has been
drawn to the importance of pathways of
degradation of complex polysaccharides
through the existence of at least 35 inherited
lysosomal storage diseases.
347 351
In
many of these diseases one of the 40-odd
lysosomal hydrolases is defective or absent.
1. Mucopolysaccharidoses
There are at least seven mucopolysac-
charidoses (Table 20-1) in which glyco-
saminoglycans such as hyaluronic acid
accumulate to abnormal levels in tissues
and may be excreted in the urine. The
diseases cause severe skeletal defects;
varying degrees of mental retardation;
and early death from liver, kidney, or
cardiovascular problems. As in other
lysosomal diseases, undegraded material
is stored in intracellular inclusions lined
by a single membrane. Various tissues
are affected to different degrees, and the
diseases tend to progress with time.
First described in 1919 by Hurler,
mucopolysaccharidosis I (MPS I, the
Hurler syndrome) leads to accumulation
of partially degraded dermatan and hepa-
ran sulfates (Fig. 4-11).
347,352,353
A standard
procedure in the study of diseases of this
type is to culture fibroblasts from a skin
biopsy. Such cells cultured from patients
with the Hurler syndrome accumulate the
polysaccharide, but when fibroblasts from
a normal person are cultured in the same
vessel the defect is corrected. It was
shown that a protein secreted by the nor-
mal fibroblasts is taken up by the defective
fibroblasts, permitting them to complete the
degradation of the stored polysaccharide.
G. The Intracellular Breakdown of Polysaccharides and Glycolipids
Figure 20-11 Biosynthesis and catabolism of glycosphingolipids. The
heavy bars indicate metabolic blocks in known diseases.
Sphingosine
UDP-Gal
UDP
O-Galactosylsphingosine (psychosine)
Acyl-CoA
Gal-Cer
(20-24)
This Hurler corrective factor was identified as an
-L-iduronidase. In the Hunter syndrome (MPS II)
dermatan sulfate and heparan sulfate accumulate.
The missing enzyme is a sulfatase for 2-sulfated
iduronate residues.
354,355
The diagram at the bottom
of the page illustrates the need for both of these en-
zymes as well as three others in the degradation of
dermatan.
352,356,357
The Sanfilippo disease type A (MPS III) corrective
factor is a heparan N-sulfatase. However, as is true for
many other metabolic diseases, the same symptoms
Sphingomyelin
Phosphatidylcholine
POCholine
1
Ceramide (Cer)
2
Sphingosine
Acyl-CoA
9
UDP-Gal
GalCer
PAPS
12
10

SO
3
-Gal-Cer
11
UDP-Glc
3
Glc-Cer
UDP-Gal
4
Gal14Glc-Cer
(Lactosyl ceramide)
CMP-NeuNAc
NeuNAc-Gal-Glc-Cer (GM
3
)
UDP-GalNAC
5
13
NeuNAc-Gal-Glc-Cer (GM
2
or
Tay-Sachs ganglioside)
Gal-GalNAc
4
6
UDP-Gal
NeuNAc-Gal-Glc-Cer (GM
1
)
Gal-GalNAc
CMP-NeuNAc
NeuNAc-Gal-Glc-Cer
NeuNAc-Gal-GalNAc
UDP-Gal
7
Gal14Gal-Glc-Cer
GalNAc13Gal-Gal-Glc-Cer
(Globoside or cytolipin K)
UDP-GalNAc
GalNAc-Gal-Glc-Cer
NeuNAc-NeuNAc-Gal-Glc-Cer (GD
3
)
NeuNAc-NeuNAc-Gal-Glc-Cer (GD
3
)
GalNAc
NeuNAc-NeuNAc-Gal-Glc-Cer (GD
3
)
Gal-GalNAc
H OH
CH
2
NH H
C
R
O
O
P
O
O O
Choline

H OH
CH
2
OH
NH H
C
R
O
Diacylglycerol
Ch 20 IND page-E- 2/14/03, 11:31 AM 1169
1170 Chapter 20. Some Pathways of Carbohydrate Metabolism
may arise from several causes. Thus, Sanfilippo diseases
B and D arise from lack of an N-acetylglucosaminidase
and of a sulfatase for GlcNac-6-sulfate, respectively.
354
In Sanfilippo disease C the missing or defective enzyme
is an acetyl transferase that transfers an acetyl group
from acetyl-CoA onto the amino groups of glucosamine
residues in heparan sulfate fragments. All four of
these enzymes are needed to degrade the glucosamine-
uronic acid pairs of heparan. The N-sulfate groups
must be removed by the N-sulfatase. The free amino
groups formed must then be acetylated before the
N-acetylglucosaminidase can cut off the GlcNac groups.
Remov-al of the 6-sulfate groups requires the fourth
enzyme. Completion of the degradation also requires
both -glucuronidase and -L-iduronidase. Another
lysosomal enzyme deficiency, which is most prevalent
in Finland, is the absence of aspartylglucosamini-
dase, an N-terminal nucleophile hydrolase (Chapter
12, Section C,3) that cleaves glucosamine from aspar-
tate side chains to which oligosaccharides were at-
tached in glycoproteins.
358 360
Some hereditary diseases are characterized by
lack of two or more lysosomal enzymes. In I-cell
disease (mucolipidosis II), which resembles the
Hurler syndrome, at least ten enzymes are absent or are
present at much reduced levels.
350,361
The biochemical
defect is the absence from the Golgi cisternae of the
N-acetylglucosaminyl phosphotransferase that trans-
fers P-GlcNAc units from UDP-GlcNAc onto mannose
residues (Eq. 20-22) of glycoproteins marked for use
in lysosomes.
2. Sphingolipidoses
There are at least ten lysosomal storage diseases,
known as sphingolipidoses, that involve the metabo-
lism of the glycolipids. Their biochemical bases are
indicated in Fig. 20-11 and in Table 20-1. Gaucher
disease
362 365
is a result of an autosomal recessive trait
that permits glucosyl ceramide to accumulate in macro-
phages. The liver and spleen are seriously damaged,
the latter becoming enlarged to four or five times
normal size in the adult form of the disease. In the
more severe juvenile form mental retardation occurs.
By 1965, it was established that cerebroside is synthe-
sized at a normal rate in the individuals affected, but
that a lysosomal hydrolase was missing. This blocked
the catabolic pathway indicated by dashed arrows in
Fig. 20-11 (block No. 3 in the figure). In many patients
a single base change causing a Leu Pro substitution
accounts for the defect. In Fabry disease an X-linked
gene that provides for removal of galactosyl residues
from cerebrosides is defective.
350
This leads to accu-
mulation of the triglycosylceramide whose degrada-
tion is blocked at point 7 in Fig. 20-11.
The best known and the commonest sphingolipi-
dosis is TaySachs disease.
366 368
Several hundred
cases have been reported since it was first described in
1881. A terrible disease, it is accompanied by mental
deterioration, blindness, paralysis, dementia, and
death by the age of three. About 15 children a year
are born in North America with this condition, and
the world figure must be 5 7 times this. The defect is
in the subunit of the -hexosaminidase A (point 7
in Fig. 20-10)
366,366a
with accumulation of ganglioside
G
M2
. Somewhat less severe forms of the disease are
caused by different mutations in the same gene
369
or in
a protein activator. Sandhoff disease, which resembles
TaySachs disease, is caused by a defect in the sub-
unit, which is present in both -hexosaminidases A
and B.
368
Mutant knockout mice that produce only
ganglioside GM
3
as the major ganglioside in their
central nervous system die suddenly from seizures if
they hear a loud sound. This provides further evidence
of the essential nature of these components of nerve
membranes.
369a
3. Causes of Lysosomal Diseases
The descriptions given here have been simplified.
For many lysosomal diseases there are mild and severe
forms and infantile or juvenile forms to be contrasted
with adult forms. Some of the enzymes exist as multiple
isozymes. An enzyme may be completely lacking or
O
O
HO
O HO
O
NH
O
OH
O
SO
3
O
CH
3
COO
O
3
S
O
OH
O
OH
OH
O
NH
O
CH
3

HO

COO

O
H
3. GalNAc 4-Sulfatase
MPS VI, Maroteaux-Lamy
5. -Glucuronidase
MPS VII, Sly
4. -Hexosaminidase A,B
Sandhoff
2. -d-Iduronidase
MPS I, Hurler
1. Iduronate sulfatase
MPS II, Hunter
Ch 20 IND page-E- 2/14/03, 11:31 AM 1170
1171
a
A general reference is Scriver, C. R., Beaudet, A. L., Sly, W. S., and Valle, D., eds. (1995) The Metabolic and Molecular
Bases of Inherited Disease, 7th ed., Vol. 1, McGraw-Hill, New York (pp. 24272879)
b
Wenger, D. A., Sattler, M., Kudoh, T., Snyder, S. P., and Kingston, R. S. (1980) Science 208, 14711473
c
See main text
d
Hoogeveen, A. T., Reuser, A. J. J., Kroos, M., and Galjaard, H. (1986) J. Biol. Chem. 261, 57025704
e
Gravel, R. A., Clarke, J. T. R., Kaback, M. M., Mahuran, D., Sandhoff, K., and Suzuki, K. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 2839
2879, McGraw-Hill, New York
f
See Box 20-D
g
Neufeld, E. F., and Muenzer, J. (1995) in The Metabolic and Molecular Bases of Inherited Disease, 7th ed., Vol. 2 (Scriver, C.
R., Beaudet, A. L., Sly, W. S., and Valle, D., eds), pp. 2465 2494, McGraw-Hill, New York
h
Wu, B. M., Tomatsu, S., Fukuda, S., Sukegawa, K., Orii, T., and Sly, W. S. (1994) J. Biol. Chem. 269, 2368123688
i
Mononen, I., Fisher, K. J., Kaartinen, V., and Aronson, N. N., Jr. (1993) FASEB J. 7, 12471256
j
Seppala, R., Tietze, F., Krasnewich, D., Weiss, P., Ashwell, G., Barsh, G., Thomas, G. H., Packman, S., and Gahl, W. A.
(1991) J. Biol. Chem. 266, 74567461
1. NiemannPick disease
b
Sphingomyelinase
2. Farber disease (lipogranulomatosis) Ceramidase
3. Gaucher disease
c
-Glucocerebrosidase
4. Lactosyl ceramidosis -Galactosyl hydrolase
5. TaySachs disease
c
-Hexosaminidase A
6. G
M1
gangliosidosis
d
-Galactosidase
7. Fabry disease
c
-Galactosidase
8. Sandhoff disease
e
-Hexosaminidases A and B
9. Globoid cell leukodystrophy Galactocerebrosidase
10. Metachromatic leukodystrophy Arylsulfatase A
13. Hematoside (G
M3
) accumulation G
M3
-N-acetylgalactosaminyltransferase
Pompe disease
f
-Glucosidase
Hurler syndrome (MPS I)
c
-L-Iduronidase
Hunter syndrome (MPS II)
c
Iduronate 2-sulfate sulfatase
Sanfilippo disease
c,g
Type A (MPS III) Heparan N-sulfatase
Type B N-Acetylglucosaminidase
Type C Acetyl-CoA: -glucosaminide N-acetyltransferase
Type D GlcNAc-6-sulfate sulfatase
MaroteauxLamy syndrome (MPS VI)
g
Arylsulfatase B
Sly syndrome (MPS VII)
g,h
-Glucuronidase
Aspartylglycosaminuria
i
Aspartylglucosaminidase
Mannosidosis -Mannosidase
Fucosidosis -L-Fucosidase
Mucolipidosis -N-Acetylneuraminidase
Sialidosis
j
TABLE 20-1
Lysosomal Storage Diseases: Sphingolipidoses and Mucopolysaccharidoses
a
No. in
Fig. 20-11 Name Defective enzyme
G. The Intracellular Breakdown of Polysaccharides and Glycolipids
Ch 20 IND page-E- 2/14/03, 11:31 AM 1171
1172 Chapter 20. Some Pathways of Carbohydrate Metabolism
may be low in concentration. The causes of the defi-
ciencies may include total absence of the gene, absence
of the appropriate mRNA, impaired conversion of a
proenzyme to active enzyme, rapid degradation of a
precursor or of the enzyme itself, incorrect transport
of the enzyme precursor to its proper destination,
presence of mutations that inactivate the enzyme,
or absence of protective proteins. Several lysosomal
hydrolases require auxiliary activator proteins that
allow them to react with membrane-bound sub-
strates.
350,351,365,370
4. Can Lysosomal Diseases Be Treated?
There has been some success in using enzyme
replacement therapy for lysosomal deficiency dis-
eases.
347,371-371b
One approach makes use of the fact
that the mannose-6-P receptors of the plasma mem-
brane take up suitably marked proteins and transfer
them into lysosomes (Section C,2). The missing en-
zyme might simply be injected into the patients
bloodstream from which it could be taken up into the
lysosomes.
372
This carries a risk of allergic reaction,
and it may be safer to attempt microencapsulation of
the enzyme, perhaps in ghosts from the patients own
erythrocytes.
347
A second approach, which has had
limited success, is transplantation of an organ
371
or of
bone marrow
373
from a donor with a normal gene for
the missing enzyme. This is dangerous and is little
used at present. However, new hope is offered by
the possibility of transferring a gene for the missing
enzyme into some of the patients cells. For example,
the cloned gene for the transferase missing in Gaucher
disease has been transferred into cultured cells from
Gaucher disease patients with apparent correction of
the defect.
374
Long-term correction of the Hurler syn-
drome in bone marrow cells also provides hope for an
effective therapy involving gene transfer into a patients
own bone marrow cells
375
or transplantation of selected
hematopoietic cells.
375a
In the cases of Gaucher disease and Fabry disease,
it is hoped that treatment of infants and young children
may prevent brain damage. However, in TaySachs
disease the primary sites of accumulation of the
ganglioside GM
2
are the ganglion and glial cells of
the brain. Because of the bloodbrain barrier and
the severity of the damage it seems less likely that the
disease can be treated successfully.
The approach presently used most often consists
of identifying carriers of highly undesirable genetic
traits and offering genetic counseling. For example,
if both parents are carriers the risk of bearing a child
with TaySachs disease is one in four. Women who
have borne a previous child with the disease usually
have the genetic status of the fetus checked by amnio-
centesis. A sample of the amniotic fluid surrounding
the fetus is withdrawn during the 16th to 18th week
of pregnancy. The fluid contains fibroblasts that have
become detached from the surface of the fetus. These
cells are cultured for 2 3 weeks to provide enough
cells for a reliable assay of the appropriate enzymes.
Such tests for a variety of defects are becoming faster
and more sensitive as new techniques are applied.
376
In the case of TaySachs disease, most women who
have one child with the disease choose abortion if a
subsequent fetus has the disease.
The diseases considered here affect only a small
fraction of the problems in the catabolism of body
constituents. On the other hand, fewer cases are on
record of deficiencies in biosynthetic pathways. These
are more often absolutely lethal and lead to early
spontaneous abortion. However, blockages in the
biosynthesis of cerebrosides are known in the special
strains of mice known as Jimpy, Quaking, and msd
(myelin synthesis deficient).
377,378
The transferases
(points 11 and 12 of Fig. 20-11) are not absent but are
of low activity. The mice have distinct neurological
defects and poor myelination of nerves in the brain.
A human ailment involving impaired conversion of
GM
3
to GM
2
(with accumulation of the former; point
13 of Fig. 20-11) has been reported. Excessive synthesis
of sialic acid causes the rare human sialuria.
379
This
is apparently a result of a failure in proper feedback
inhibition.
Animals suffer many of the same metabolic diseases
as humans. Among these are a large number of lyso-
somal deficiency diseases.
380
Their availability means
that new methods of treating the diseases may, in
many cases, be tried first on animals. For example,
enzyme replacement therapy for the Hurler syndrome
is being tested in dogs.
381
Bone marrow transplantation
for human -mannosidosis is being tested in cats with
a similar disease.
382
Mice with a hereditary deficiency
of -glucuronidase are being treated by gene transfer
from normal humans.
357
Ch 20 IND page-E- 2/14/03, 11:31 AM 1172
1173
43a. Benson, T. E., Harris, M. S., Choi, G. H.,
Cialdella, J. I., Herberg, J. T., Martin, J. P., Jr.,
and Baldwin, E. T. (2001) Biochemistry 40,
2340 2350
44. Pastuszak, I., ODonnell, J., and Elbein, A. D.
(1996) J. Biol. Chem. 271, 2365323656
44a. Zhao, X., Creuzenet, C., Blanger, M.,
Egbosimba, E., Li, J., and Lam, J. S. (2000) J.
Biol. Chem. 275, 33252 33259
45. Schauer, R. (1985) Trends Biochem. Sci. 10, 357
360
46. Simon, E. S., Bednarski, M. D., and Whitesides,
G. M. (1988) J. Am. Chem. Soc. 110, 71597163
47. Morgan, P. M., Sala, R. F., and Tanner, M. E.
(1997) J. Am. Chem. Soc. 119, 1026910277
47a. Campbell, R. E., Mosimann, S. C., Tanner, M.
E., and Strynadka, N. C. J. (2000) Biochemis-
try 39, 14993 15001
47b. Keppler, O. T., Hinderlich, S., Langner, J.,
Schwartz-Albiez, R., Reutter, W., and
Pawlita, M. (1999) Science 284, 1372 1376
47c. Itoh, T., Mikami, B., Maru, I., Ohta, Y.,
Hashimoto, W., and Murata, K. (2000) J. Mol.
Biol. 303, 733 744
47d. Jacobs, C. L., Goon, S., Yarema, K. J.,
Hinderlich, S., Hang, H. C., Chai, D. H., and
Bertozzi, C. R. (2001) Biochemistry 40, 12864
12874
48. Stsche, R., Hinderlich, S., Weise, C., Effertz,
K., Lucka, L., Moormann, P., and Reutter, W.
(1997) J. Biol. Chem. 272, 2431924324
48a. Jordan, P. A., Bohle, D. S., Ramilo, C. A., and
Evans, J. N. S. (2001) Biochemistry 40, 8387
8396
49. Dotson, G. D., Dua, R. K., Clemens, J. C.,
Wooten, E. W., and Woodard, R. W. (1995) J.
Biol. Chem. 270, 1369813705
50. Sheflyan, G. Y., Howe, D. L., Wilson, T. L., and
Woodard, R. W. (1998) J. Am. Chem. Soc. 120,
1102811032
51. Kaustov, L., Kababya, S., Du, S., Baasov, T.,
Gropper, S., Shoham, Y., and Schmidt, A.
(2000) Biochemistry 39, 14865 14876
52. Radaev, S., Dastidar, P., Patel, M., Woodard,
R. W., and Gatti, D. L. (2000) J. Biol. Chem.
275, 9476 9484
52a. Duewel, H. S., Radaev, S., Wang, J.,
Woodard, R. W., and Gatti, D. L. (2001) J.
Biol. Chem. 276, 8393 8402
52b. Wagner, T., Kretsinger, R. H., Bauerle, R.,
and Tolbert, W. D. (2000) J. Mol. Biol. 301,
233 238
52c. Mosimann, S. C., Gilbert, M., Dombroswki,
D., To, R., Wakarchuk, W., and Strynadka, N.
C. J. (2001) J. Biol. Chem. 276, 8190 8196
53. Kawano, T., Koyama, S., Takematsu, H.,
Kozutsumi, Y., Kawasaki, H., Kawashima, S.,
Kawasaki, T., and Suzuki, A. (1995) J. Biol.
Chem. 270, 1645816463
54. Nishino, S., Kuroyanagi, H., Terada, T., Inoue,
S., Inoue, Y., Troy, F. A., and Kitajima, K.
(1996) J. Biol. Chem. 271, 29092913
55. Inoue, S., Kitajima, K., and Inoue, Y. (1996) J.
Biol. Chem. 271, 2434124344
55a. Angata, T., Nakata, D., Matsuda, T.,
Kitajima, K., and Troy, F. A., II. (1999) J. Biol.
Chem. 274, 22949 22956
55b. Lawrence, S. M., Huddleston, K. A., Pitts, L.
R., Nguyen, N., Lee, Y. C., Vann, W. F.,
Coleman, T. A., and Betenbaugh, M. J. (2000)
J. Biol. Chem. 275, 17869 17877
56. Kohlbrenner, W. E., Nuss, M. M., and Fesik, S.
W. (1987) J. Biol. Chem. 262, 45344537
56a. Royo, J., Gmez, E., and Hueros, G. (2000) J.
Biol. Chem. 275, 24993 24999
56b. Jelakovic, S., and Schulz, G. E. (2001) J. Mol.
Biol. 312, 143 155
57. Kontrohr, T., and Kocsis, B. (1981) J. Biol.
Chem. 256, 77157718
1. Cardini, C. E., Paladini, A. C., Caputto, R.,
and Leloir, L. F. (1950) Nature (London) 165,
191192
2. Grisola, S. (1988) Nature (London) 331, 212
3. Hallfrisch, J. (1990) FASEB J. 4, 26522660
4. Jefferey, J., and Jornvall, H. (1983) Proc. Natl.
Acad. Sci. U.S.A. 80, 901905
5. Cox, T. M. (1994) FASEB J. 8, 6271
6. Gitzelmann, R., Steinmann, B., and Van den
Berghe, G. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 905935, McGraw-
Hill, New York
7. Gopher, A., Vaisman, N., Mandel, H., and
Lapidot, A. (1990) Proc. Natl. Acad. Sci. U.S.A.
87, 54495453
8. Hope, J. N., Bell, A. W., Hermodson, M. A.,
and Groarke, J. M. (1986) J. Biol. Chem. 261,
76637668
9. Ridley, W. P., Houchins, J. P., and Kirkwood,
S. (1975) J. Biol. Chem. 250, 87618767
9a. Campbell, R. E., Mosimann, S. C., van de
Rijn, I., Tanner, M. E., and Strynadka, N. C. J.
(2000) Biochemistry 39, 7012 7023
10. Li, J.-p, Hagner-McWhirter, ., Kjelln, L.,
Palgi, J., Jalkanen, M., and Lindahl, U. (1997)
J. Biol. Chem. 272, 2815828163
11. Segal, S., and Berry, G. T. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 1 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 967
1000, McGraw-Hill, New York
12. Ruzicka, F. J., Wedekind, J. E., Kim, J.,
Rayment, I., and Frey, P. A. (1995) Biochemistry
34, 56105617
13. Wedekind, J. E., Frey, P. A., and Rayment, I.
(1996) Biochemistry 35, 1156011569
14. Thoden, J. B., Ruzicka, F. J., Frey, P. A.,
Rayment, I., and Holden, H. M. (1997)
Biochemistry 36, 12121222
15. Frey, P. A. (1996) FASEB J. 10, 461470
15a. Lai, K., Willis, A. C., and Elsas, L. J. (1999) J.
Biol. Chem. 274, 6559 6566
16. Thoden, J. B., and Holden, H. M. (1998)
Biochemistry 37, 1146911477
16a. Thoden, J. B., Wohlers, T. M., Fridovich-Keil,
J. L., and Holden, H. M. (2001) J. Biol. Chem.
276, 15131 15136
16b. Thoden, J. B., Wohlers, T. M., Fridovich-Keil,
J. L., and Holden, H. M. (2001) J. Biol. Chem.
276, 20617 20623
17. Beebe, J. A., and Frey, P. A. (1998) Biochemistry
37, 1498914997
17a. Zhang, Q., and Liu, H.-w. (2001) J. Am.
Chem. Soc. 123, 6756 6766
18. Wong, Y.-H. H., and Sherman, W. R. (1985) J.
Biol. Chem. 260, 1108311090
19. Migaud, M. E., and Frost, J. W. (1996) J. Am.
Chem. Soc. 118, 495501
19a. Tian, F., Migaud, M. E., and Frost, J. W.
(1999) J. Am. Chem. Soc. 121, 5795 5796
19b. Chen, L., Zhou, C., Yang, H., and Roberts,
M. F. (2000) Biochemistry 39, 12415 12423
20. Loewus, M. W., Loewus, F. A., Brillinger, G.
U., Otsuka, H., and Floss, H. G. (1980) J. Biol.
Chem. 255, 1171011712
21. Lapan, E. A. (1975) Exp. Cell. Res. 94, 277282
22. Holub, B. J. (1992) N. Engl. J. Med. 326, 1285
1287
22a. Nestler, J. E., Jakubowicz, D. J., Reamer, P.,
Gunn, R. D., and Allan, G. (1999) N. Engl. J.
Med. 340, 1314 1320
23. Reddy, C. C., Swan, J. S., and Hamilton, G. A.
(1981) J. Biol. Chem. 256, 85108518
24. Nishikimi, M., and Yagi, K. (1996) in
Subcellular Biochemistry, Vol. 25 (Harris, J. R.,
ed), pp. 1739, Plenum, New York
25. Koshizaka, T., Nishikimi, M., Ozawa, T., and
Yagi, K. (1988) J. Biol. Chem. 263, 16191621
26. Nishikimi, M., Fukuyama, R., Minoshima, S.,
Shimizu, N., and Yagi, K. (1994) J. Biol. Chem.
269, 1368513688
26a. Maeda, N., Hagihara, H., Nakata, Y., Hiller,
S., Wilder, J., and Reddick, R. (2000) Proc.
Natl. Acad. Sci. U.S.A. 97, 841 846
26b. Tsukaguchi, H., Tokui, T., Mackenzie, B.,
Berger, U. V., Chen, X.-Z., Wang, Y.,
Brubaker, R. F., and Hediger, M. A. (1999)
Nature (London) 399, 70 75
27. Miller, J. V., Estell, D. A., and Lazarus, R. A.
(1987) J. Biol. Chem. 262, 90169020
28. Wheeler, G. L., Jones, M. A., and Smirnoff, N.
(1998) Nature (London) 393, 365369
29. Conklin, P. L., Norris, S. R., Wheeler, G. L.,
Williams, E. H., Smirnoff, N., and Last, R. L.
(1999) Proc. Natl. Acad. Sci. U.S.A. 96, 4198
4203
30. Hiatt, H. H. (1995) in The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1 (Scriver, C. R., Beaudet, A. L., Sly, W. S.,
and Valle, D., eds), pp. 10011010, McGraw-
Hill, New York
31. Massire, F., Badet-Denisot, M.-A., Ren, L.,
and Badet, B. (1997) J. Am. Chem. Soc. 119,
57485749
32. Bearne, S. L., and Wolfenden, R. (1995)
Biochemistry 34, 1151511520
33. Badet, B., Vermoote, P., Haumont, P. Y.,
Lederer, F., and Le Goffic, F. (1987) Biochemis-
try 26, 19401948
34. Golinelli-Pimpaneau, B., Le Goffic, F., and
Badet, B. (1989) J. Am. Chem. Soc. 111, 3029
3034
34a. Bearne, S. L., and Blouin, C. (2000) J. Biol.
Chem. 275, 135 140
35. Leriche, C., Badet-Denisot, M.-A., and Badet,
B. (1996) J. Am. Chem. Soc. 118, 17971798
36. Mio, T., Yamada-Okabe, T., Arisawa, M., and
Yamada-Okabe, H. (1999) J. Biol. Chem. 274,
424429
37. Mengin-Lecreulx, D., and van Heijenoort, J.
(1996) J. Biol. Chem. 271, 3239
37a. Sulzenbacher, G., Gal, L., Peneff, C., Fassy,
F., and Bourne, Y. (2001) J. Biol. Chem. 276,
11844 11851
37b. Olsen, L. R., and Roderick, S. L. (2001)
Biochemistry 40, 1913 1921
38. Montero-Morn, G. M., Horjales, E.,
Calcagno, M. L., and Altamirano, M. M.
(1998) Biochemistry 37, 78447849
39. Wolosker, H., Kline, D., Bian, Y., Blackshaw,
S., Cameron, A. M., Fralich, T. J., Schnaar, R.
L., and Snyder, S. H. (1998) FASEB J. 12, 91
99
40. Ramilo, C., Appleyard, R. J., Wanke, C.,
Krekel, F., Amrhein, N., and Evans, J. N. S.
(1994) Biochemistry 33, 1507115079
41. Kim, D. H., Lees, W. J., Kempsell, K. E., Lane,
W. S., Duncan, K., and Walsh, C. T. (1996)
Biochemistry 35, 49234928
41a. Krekel, F., Oecking, C., Amrhein, N., and
Macheroux, P. (1999) Biochemistry 38, 8864
8878
41b. Krekel, F., Samland, A. K., Macheroux, P.,
Amrhein, N., and Evans, J. N. S. (2000)
Biochemistry 39, 12671 12677
41c. Samland, A. K., Etezady-Esfarjani, T.,
Amrhein, N., and Macheroux, P. (2001)
Biochemistry 40, 1550 1559
42. Benson, T. E., Walsh, C. T., and Hogle, J. M.
(1997) Biochemistry 36, 806811
43. Benson, T. E., Walsh, C. T., and Massey, V.
(1997) Biochemistry 36, 796805
References
Ch 20 IND page-E- 2/14/03, 11:31 AM 1173
1174 Chapter 20. Some Pathways of Carbohydrate Metabolism
58. Ding, L., Seto, B. L., Ahmed, S. A., and
Coleman, W. G., Jr. (1994) J. Biol. Chem. 269,
2438424390
58a. Ni, Y., McPhie, P., Deacon, A., Ealick, S., and
Coleman, W. G., Jr. (2001) J. Biol. Chem. 276,
27329 27334
58b. Kneidinger, B., Graninger, M., Puchberger,
M., Kosma, P., and Messner, P. (2001) J. Biol.
Chem. 276, 20935 20944
59. Sharon, N. (1975) Complex Carbohydrates,
Addison-Wesley, Reading, Massachusetts
(pp. 131138)
59a. Blankenfeldt, W., Asuncion, M., Lam, J. S.,
and Naismith, J. H. (2000) EMBO J. 19, 6652
6663
59b. Hegeman, A. D., Gross, J. W., and Frey, P. A.
(2001) Biochemistry 40, 6598 6610
59c. Gross, J. W., Hegeman, A. D., Gerratana, B.,
and Frey, P. A. (2001) Biochemistry 40, 12497
12504
59d. Allard, S. T. M., Giraud, M.-F., Whitfield, C.,
Graninger, M., Messner, P., and Naismith, J.
H. (2001) J. Mol. Biol. 307, 283 295
59e. Christendat, D., Saridakis, V., Dharamsi, A.,
Bochkarev, A., Pai, E. F., Arrowsmith, C. H.,
and Edwards, A. M. (2000) J. Biol. Chem. 275,
24608 24612
59f. Kneidinger, B., Graninger, M., Adam, G.,
Puchberger, M., Kosma, P., Zayni, S., and
Messner, P. (2001) J. Biol. Chem. 276, 5577
5583
60. Chan, J. Y., Nwokoro, N. A., and Schachter, H.
(1979) J. Biol. Chem. 251, 70607068
60a. He, X. M., and Liu, H.-w. (2002) Ann. Rev.
Biochem. 71, 701754
61. Bonin, C. P., Potter, I., Vanzin, G. F., and Reiter,
W.-D. (1997) Proc. Natl. Acad. Sci. U.S.A. 94,
20852090
61a. Menon, S., Stahl, M., Kumar, R., Xu, G.-Y.,
and Sullivan, F. (1999) J. Biol. Chem. 274,
26743 26750
61b. Rosano, C., Bisso, A., Izzo, G., Tonetti, M.,
Sturla, L., De Flora, A., and Bolognesi, M.
(2000) J. Mol. Biol. 303, 77 91
62. Rubenstein, P. A., and Strominger, J. L. (1974)
J. Biol. Chem. 249, 37763781
63. Pieper, P. A., Guo, Z., and Liu, H.-w. (1995)
J. Am. Chem. Soc. 117, 51585159
64. Johnson, D. A., Gassner, G. T., Bandarian, V.,
Ruzicka, F. J., Ballou, D. P., Reed, G. H., and
Liu, H.-w. (1996) Biochemistry 35, 1584615856
65. Chen, X. M. H., Ploux, O., and Liu, H.-w.
(1996) Biochemistry 35, 1641216420
65a. Hallis, T. M., Zhao, Z., and Liu, H.-w. (2000)
J. Am. Chem. Soc. 122, 10493 10503
66. Chang, C.-W. T., Chen, X. H., and Liu, H.-w.
(1998) J. Am. Chem. Soc. 120, 96989699
67. Zhao, L., Que, N. L. S., Xue, Y., Sherman, D.
H., and Liu, H.-w. (1998) J. Am. Chem. Soc.
120, 1215912160
68. Toshima, K., Nozaki, Y., Mukaiyama, S.,
Tamai, T., Nakata, M., Tatsuta, K., and
Kinoshita, M. (1995) J. Am. Chem. Soc. 117,
37173727
69. Benning, C., Beatty, J. T., Prince, R. C., and
Somerville, C. R. (1993) Proc. Natl. Acad. Sci.
U.S.A. 90, 15611565
70. Benson, A. A. (1963) Adv. Lipid Res. 1, 387394
70a. Mulichak, A. M., Theisen, M. J., Essigmann,
B., Benning, C., and Garavito, R. M. (1999)
Proc. Natl. Acad. Sci. U.S.A. 96, 13097 13102
70b. Sanda, S., Leustek, T., Theisen, M. J., Garavito,
R. M., and Benning, C. (2001) J. Biol. Chem. 276,
39413946
71. Preiss, J. (1984) Trends Biochem. Sci. 9, 2427
71a. Lunn, J. E., Ashton, A. R., Hatch, M. D., and
Heldt, H. W. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 12914 12919
References
72. Singh, A. N., Hester, L. S., and Raushel, F. M.
(1987) J. Biol. Chem. 262, 25542557
73. Hunziker, W., Spiess, M., Semenza, G., and
Lodish, H. F. (1986) Cell 46, 227234
74. Naim, H. Y., Sterchi, E. E., and Lentze, M. J.
(1988) J. Biol. Chem. 263, 1970919717
75. Wolschek, M. F., and Kubicek, C. P. (1997) J.
Biol. Chem. 272, 27292735
76. Horlacher, R., and Boos, W. (1997) J. Biol.
Chem. 272, 1302623032
76a. Hars, U., Horlacher, R., Boos, W., Welte, W.,
and Diederichs, K. (1998) Protein Sci. 7, 2511
2521
77. Kandror, O., DeLeon, A., and Goldberg, A. L.
(2002) Proc. Natl. Acad. Sci. U.S.A. 99, 9727
9732
77a. Diederichs, K., Diez, J., Greller, G., Mller,
C., Breed, J., Schnell, C., Vonrhein, C., Boos,
W., and Welte, W. (2000) EMBO J. 19, 5951
961
77b. Diez, J., Diederichs, K., Greller, G.,
Horlacher, R., Boos, W., and Welte, W. (2001)
J. Mol. Biol. 305, 905 915
78. Shaper, N. L., Hollis, G. F., Douglas, J. G.,
Kirsch, I. R., and Shaper, J. H. (1988) J. Biol.
Chem. 263, 1042010428
79. Rajput, B., Shaper, N. L., and Shaper, J. H.
(1996) J. Biol. Chem. 271, 51315142
80. Yadav, S. P., and Brew, K. (1991) J. Biol. Chem.
266, 698703
80a. Gastinel, L. N., Cambillau, C., and Bourne,
Y. (1999) EMBO J. 18, 3546 3557
80b. Ramakrishnan, B., Shah, P. S., and Qasba, P.
K. (2001) J. Biol. Chem. 276, 37665 37671
81. Montgomery, R. K., Bller, H. A., Rings, E. H.
H. M., and Grand, R. J. (1991) FASEB J. 5,
28242832
81a. Potera, C. (1998) Science 281, 1793
81b. Barbier, O., Girard, C., Breton, R., Blanger,
A., and Hum, D. W. (2000) Biochemistry 39,
11540 11552
81c. Lvesque, E., Turgeon, D., Carrier, J.-S.,
Montminy, V., Beaulieu, M., and Blanger, A.
(2001) Biochemistry 40, 3869 3881
82. Lewis, D. A., and Armstrong, R. N. (1983)
Biochemistry 22, 62976303
83. Hanessian, S., and Haskell, T. H. (1970) in The
Carbohydrates, 2nd ed., Vol. 2A (Pigman, W.,
and Horton, D., eds), pp. 139211, Academic
Press, New York
84. Snell, J. F. (1966) Biosynthesis of Antibiotics, Vol.
1, Academic Press, New York
85. Botti, M. G., Taylor, M. G., and Botting, N. P.
(1995) J. Biol. Chem. 270, 2053020535
86. Cottaz, S., Henrissat, B., and Driguez, H.
(1996) Biochemistry 35, 1525615259
86a. Burmeister, W. P., Cottaz, S., Rollin, P.,
Vasella, A., and Henrissat, B. (2000) J. Biol.
Chem. 275, 39385 39393
87. Lomako, J., Lomako, W. M., and Whelan, W. J.
(1988) FASEB J. 2, 30973103
87a. Cid, E., Gomis, R. R., Geremia, R. A.,
Guinovart, J. J., and Ferrer, J. C. (2000) J. Biol.
Chem. 275, 33614 33621
88. Baecker, P. A., Greenberg, E., and Preiss, J.
(1986) J. Biol. Chem. 261, 87388743
89. Hill, M. A., Kaufmann, K., Otero, J., and Preiss,
J. (1991) J. Biol. Chem. 266, 1245512460
90. Furukawa, K., Tagaya, M., Tanizawa, K., and
Fukui, T. (1994) J. Biol. Chem. 269, 868871
91. Ochoa, S. (1985) Trends Biochem. Sci. 10,
147150
92. Kim, S. C., Singh, A. N., and Raushel, F. M.
(1988) J. Biol. Chem. 263, 1015110154
93. Skurat, A. V., Wang, Y., and Roach, P. J. (1994)
J. Biol. Chem. 269, 2553425542
94. Shulman, R. G., Bloch, G., and Rothman, D. L.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 8535
8542
95. Villar-Palasi, C., and Guinovart, J. J. (1997)
FASEB J. 11, 544558
96. Printen, J. A., Brady, M. J., and Saltiel, A. R.
(1997) Science 275, 14751478
96a. Halse, R., Rochford, J. J., McCormack, J. G.,
Vandenheede, J. R., Hemmings, B. A., and
Yeaman, S. J. (1999) J. Biol. Chem. 274, 776780
96b. Oikonomakos, N. G., Schnier, J. B.,
Zographos, S. E., Skamnaki, V. T., Tsitsanou,
K. E., and Johnson, L. N. (2000) J. Biol. Chem.
275, 34566 34573
97. Thon, V. J., Khalil, M., and Cannon, J. F. (1993)
J. Biol. Chem. 268, 75097513
98. Takrama, J., and Madsen, N. B. (1988)
Biochemistry 27, 33083314
99. Yang, B.-Z., Ding, J.-H., Enghild, J. J., Bao, Y.,
and Chen, Y.-T. (1992) J. Biol. Chem. 267,
92949299
99a. Nakayama, A., Yamamoto, K., and Tabata, S.
(2001) J. Biol. Chem. 276, 28824 28828
100. Alonso, M. D., Lomako, J., Lomako, W. M.,
and Whelan, W. J. (1995) FASEB J. 9, 11261137
101. Blumenfeld, M. L., and Krisman, C. R. (1985)
J. Biol. Chem. 260, 1156011566
101a. Pederson, B. A., Cheng, C., Wilson, W. A.,
and Roach, P. J. (2000) J. Biol. Chem. 275,
27753 27761
102. Alonso, M. D., Lomako, J., Lomako, W. M.,
and Whelan, W. J. (1995) J. Biol. Chem. 270,
1531515319
103. Ercan, N., Gannon, M. C., and Nuttall, F. Q.
(1994) J. Biol. Chem. 269, 2232822333
104. Mu, J., and Roach, P. J. (1998) J. Biol. Chem.
273, 3485034856
105. Mu, J., Skurat, A. V., and Roach, P. J. (1997) J.
Biol. Chem. 272, 2758927597
106. Charng, Y.-y, Iglesias, A. A., and Preiss, J.
(1994) J. Biol. Chem. 269, 2410724113
107. Nakata, P. A., Anderson, J. M., and Okita, T.
W. (1994) J. Biol. Chem. 269, 3079830807
108. Van den Koornhuyse, N., Libessart, N.,
Delrue, B., Zabawinski, C., Decq, A., Iglesias,
A., Carton, A., Preiss, J., and Ball, S. (1996) J.
Biol. Chem. 271, 1628116287
109. Pozueta-Romero, J., Frehner, M., Viale, A. M.,
and Akazawa, T. (1991) Proc. Natl. Acad. Sci.
U.S.A. 88, 57695773
110. Yu, Y., Mu, H. H., Mu-Forster, C., and
Wasserman, B. P. (1998) Plant Physiol. 116,
14511460
111. Calvert, P. (1997) Nature (London) 389, 338339
112. Ball, S., Guan, H.-P., James, M., Myers, A.,
Keeling, P., Mouille, G., Bulon, A., Colonna,
P., and Preiss, J. (1996) Cell 86, 349352
113. Waigh, T. A., Hopkinson, I., Donald, A. M.,
Butler, M. F., Heidelbach, F., and Riekel, C.
(1997) Macromolecules 30, 38133820
114. Gallant, D. J., Bouchet, B., and Baldwin, P. M.
(1997) Carbo. Polymers 32, 177191
115. Fontaine, T., DHulst, C., Maddelein, M.-L.,
Routier, F., Ppin, T. M., Decq, A.,
Wieruszeski, J.-M., Delrue, B., Van den
Koornhuyse, N., Bossu, J.-P., Fournet, B., and
Ball, S. (1993) J. Biol. Chem. 268, 1622316230
116. Gao, M., Wanat, J., Stinard, P. S., James, M. G.,
and Myers, A. M. (1998) Plant Cell 10, 399412
117. Cao, H., Imparl-Radosevich, J., Guan, H. P.,
Keeling, P. L., James, M. G., and Myers, A. M.
(1999) Plant Physiol. 120, 111
118. Rahman, A., Wong, K.-s, Jane, J.-l, Myers, A.
M., and James, M. G. (1998) Plant Physiol. 117,
425435
119. Guan, H., Kuriki, T., Sivak, M., and Preiss, J.
(1995) Proc. Natl. Acad. Sci. U.S.A. 92, 964967
120. Beatty, M. K., Rahman, A., Cao, H., Woodman,
W., Lee, M., Myers, A. M., and James, M. G.
(1999) Plant Physiol. 119, 255266
121. Robyt, J. F. (1998) Essentials of Carbohydrate
Chemistry, Springer, New York
Ch 20 IND page-E- 2/14/03, 11:31 AM 1174
1175
165. Robertson, D., Mitchell, G. P., Gilroy, J. S.,
Gerrish, C., Bolwell, G. P., and Slabas, A. R.
(1997) J. Biol. Chem. 272, 1584115848
166. Averyhart-Fullard, V., Datta, K., and Marcus,
A. (1988) Proc. Natl. Acad. Sci. U.S.A. 85,
10821085
167. Condit, C. M., and Meagher, R. B. (1986)
Nature (London) 323, 178181
168. Cosgrove, D. J. (1997) Proc. Natl. Acad. Sci.
U.S.A. 94, 55045505
169. Rose, J. K. C., Lee, H. H., and Bennett, A. B.
(1997) Proc. Natl. Acad. Sci. U.S.A. 94, 59555960
170. Fleming, A. J., McQueen-Mason, S., Mandel,
T., and Kuhlemeier, C. (1997) Science 276,
14151418
171. Stanley, P., and Ioffe, E. (1995) FASEB J. 9,
14361444
172. Baenziger, J. U. (1994) FASEB J. 8, 10191025
172a. DeAngelis, P. L., and Padgett-McCue, A. J.
(2000) J. Biol. Chem. 275, 24124 24129
172b. Pummill, P. E., Kempner, E. S., and
DeAngelis, P. L. (2001) J. Biol. Chem. 276,
39832 39835
173. Laurent, T. C., and Fraser, J. R. E. (1992)
FASEB J. 6, 23972404
174. Tlapak-Simmons, V. L., Baggenstoss, B. A.,
Kumari, K., Heldermon, C., and Weigel, P. H.
(1999) J. Biol. Chem. 274, 42464253
175. Spicer, A. P., and McDonald, J. A. (1998) J.
Biol. Chem. 273, 19231932
176. Robyt, J. F. (1979) Trends Biochem. Sci. 4, 4749
177. Robyt, J. F. (1995) Adv. Carbohydr. Chem.
Biochem. 51, 133168
178. Robyt, J. F., and Martin, P. J. (1983) Carbohydr.
Res. 113, 301315
179. Mooser, G., Hefta, S. A., Paxton, R. J., Shively,
J. E., and Lee, T. D. (1991) J. Biol. Chem. 266,
89168922
180. Funane, K., Shiraiwa, M., Hashimoto, K.,
Ichishima, E., and Kobayashi, M. (1993)
Biochemistry 32, 1369613702
181. Sprenger, N., Bortlik, K., Brandt, A., Boller, T.,
and Wiemken, A. (1995) Proc. Natl. Acad. Sci.
U.S.A. 92, 1165211656
182. Beale, J. M., Jr., and Foster, J. L. (1996)
Biochemistry 35, 44924501
183. Hardingham, T. E., and Fosang, A. J. (1992)
FASEB J. 6, 861870
184. Huang, S., Wang, Y.-X., and Draper, D. E.
(1996) J. Mol. Biol. 258, 308321
185. Kokenyesi, R., and Bernfield, M. (1994) J. Biol.
Chem. 269, 1230412309
186. Kearns, A. E., Campbell, S. C., Westley, J., and
Schwartz, N. B. (1991) Biochemistry 30, 7477
7483
187. Shworak, N. W., Shirakawa, M., Mulligan, R.
C., and Rosenberg, R. D. (1994) J. Biol. Chem.
269, 2120421214
188. Vertel, B. M., Walters, L. M., Flay, N., Kearns,
A. E., and Schwartz, N. B. (1993) J. Biol. Chem.
268, 1110511112
189. Fernndez, C. J., and Warren, G. (1998) J. Biol.
Chem. 273, 1903019039
190. Lugemwa, F. N., Sarkar, A. K., and Esko, J. D.
(1996) J. Biol. Chem. 271, 1915919165
190a. Bai, X., Wei, G., Sinha, A., and Esko, J. D.
(1999) J. Biol. Chem. 274, 13017 13024
190b. Gtting, C., Kuhn, J., Zahn, R., Brinkmann,
T., and Kleesiek, K. (2000) J. Mol. Biol. 304,
517 528
190c. Pedersen, L. C., Tsuchida, K., Kitagawa, H.,
Sugahara, K., Darden, T. A., and Negishi, M.
(2000) J. Biol. Chem. 275, 34580 34585
191. Sugahara, K., Ohkita, Y., Shibata, Y., Yoshida,
K., and Ikegami, A. (1995) J. Biol. Chem. 270,
72047212
122. Ball, S. G., van de Wal, M. H. B. J., and Visser,
R. G. F. (1998) Trends Plant Sci. 3, 462467
122a. Sehnke, P. C., Chung, H.-J., Wu, K., and Ferl,
R. J. (2001) Proc. Natl. Acad. Sci. U.S.A. 98,
765 770
123. French, D. and Robyt, J. F. (1973) Abstr., 166th
Natl. Meet., Am. Chem. Soc., Abstract 65 BIOL
123a. Robyt, J. F. (2000) Abstr., 220th Natl. Meet.,
Am. Chem. Soc., Abstract 84 CARB
123b. Mukerjea, R., Yu, L., and Robyt, J. F. (2002)
Carbohydr. Res. 337, 10151022
124. McGuire, V., and Alexander, S. (1996) J. Biol.
Chem. 271, 1459614603
125. Bureau, T. E., and Brown, R. M., Jr. (1987)
Proc. Natl. Acad. Sci. U.S.A. 84, 69856989
126. Lloyd, C. (1980) Nature (London) 284, 596597
127. Brown, R. M., Jr., Willison, J. H. M., and
Richardson, C. L. (1976) Proc. Natl. Acad. Sci.
U.S.A. 73, 45654569
128. Lin, F. C., Brown, R. M., Jr., Cooper, J. B., and
Delmer, D. P. (1985) Science 230, 822825
129. Ross, P., Mayer, R., and Benziman, M. (1991)
Microbiol. Rev. 55, 3558
130. Bokelman, G. H., Ruben, G. C., and Krakow,
W. (1988) J. Cell Biol. 107, 147a
131. Carpita, N., and Vergara, C. (1998) Science 279,
672673
131a. Scheible, W.-R., Eshed, R., Richmond, T.,
Delmer, D., and Somerville, C. (2001) Proc.
Natl. Acad. Sci. U.S.A. 98, 10079 10084
131b. Kurek, I., Kawagoe, Y., Jacob-Wilk, D., Doblin,
M., and Delmer, D. (2002) Proc. Natl. Acad. Sci.
U.S.A. 99, 1110911114
132. Haigler, C. H., and Blanton, R. L. (1996) Proc.
Natl. Acad. Sci. U.S.A. 93, 1208212085
133. Arioli, T., Peng, L., Betzner, A. S., Burn, J.,
Wittke, W., Herth, W., Camilleri, C., Hfte, H.,
Plazinski, J., Birch, R., Cork, A., Glover, J.,
Redmond, J., and Williamson, R. E. (1998)
Science 279, 717720
133a. Cosgrove, D. J. (2000) Nature (London) 407,
321 326
133b. Peng, L., Kawagoe, Y., Hogan, P., and Delmer,
D. (2002) Science 295, 147150
134. Koyama, M., Helbert, W., Imai, T., Sugiyama,
J., and Henrissat, B. (1997) Proc. Natl. Acad.
Sci. U.S.A. 94, 90919095
135. Han, N. S., and Robyt, J. F. (1998) Carbohydr.
Res. 313, 125133
136. Sutherland, I. W. (1993) in Industrial Gums,
3rd ed. (Whistler, R. L., and BeMiller, J. N.,
eds), pp. 6985, Academic Press, San Diego,
California
137. Sugiyama, J., Boisset, C., Hashimoto, M., and
Watanabe, T. (1999) J. Mol. Biol. 286, 247255
138. Mayer, R., Ross, P., Weinhouse, H., Amikam,
D., Volman, G., Ohana, P., Calhoon, R. D.,
Wong, H. C., Emerick, A. W., and Benziman,
M. (1991) Proc. Natl. Acad. Sci. U.S.A. 88,
54725476
139. Egli, M., Gessner, R. V., Williams, L. D.,
Quigley, G. J., van der Marel, G. A., van Boom,
J. H., Rich, A., and Frederick, C. A. (1990) Proc.
Natl. Acad. Sci. U.S.A. 87, 32353239
139a. Chang, A. L., Tuckerman, J. R., Gonzalez, G.,
Mayer, R., Weinhouse, H., Volman, G., Amikam,
D., Benziman, M., and Gilles-Gonzalez, M.-A.
(2001) Biochemistry 40, 34203426
140. Amor, Y., Haigler, C. H., Johnson, S.,
Wainscott, M., and Delmer, D. P. (1995) Proc.
Natl. Acad. Sci. U.S.A. 92, 93539357
141. Nakai, T., Tonouchi, N., Konishi, T., Kojima,
Y., Tsuchida, T., Yoshinaga, F., Sakai, F., and
Hayashi, T. (1999) Proc. Natl. Acad. Sci. U.S.A.
96, 1418
142. Martin, M. M., and Martin, J. S. (1978) Science
199, 14531455
143. Skipper, N., Sutherland, M., Davies, R. W.,
Kilburn, D., Miller, R. C., Jr., Warren, A., and
Wong, R. (1985) Science 230, 958960
144. Ohana, P., Delmer, D. P., Steffens, J. C.,
Matthews, D. E., Mayer, R., and Benziman, M.
(1991) J. Biol. Chem. 266, 1374213745
145. Arellano, M., Durn, A., and Prez, P. (1996)
EMBO J. 15, 45844591
146. Hrmova, M., Garrett, T. P. J., and Fincher, G.
B. (1995) J. Biol. Chem. 270, 1455614563
147. Cabib, E. (1987) Adv. Enzymol. 59, 59101
148. Orlean, P. (1987) J. Biol. Chem. 262, 57325739
149. Silverman, S. J., Sburlati, A., Slater, M. L., and
Cabib, E. (1988) Proc. Natl. Acad. Sci. U.S.A.
85, 47354739
150. Machida, S., and Saito, M. (1993) J. Biol. Chem.
268, 17021707
151. Davis, L. L., and Bartnicki-Garcia, S. (1984)
Biochemistry 23, 10651073
152. Kafetzopoulos, D., Martinou, A., and
Bouriotis, V. (1993) Proc. Natl. Acad. Sci. U.S.A.
90, 25642568
153. Marx, J. L. (1977) Science 197, 11701172
154. Bassler, B. L., Gibbons, P. J., Yu, C., and
Roseman, S. (1991) J. Biol. Chem. 266, 24268
24275
155. Sticher, L., Hofsteenge, J., Milani, A.,
Neuhaus, J.-M., and Meins, F., Jr. (1992)
Science 257, 655657
156. Boot, R. G., Renkema, G. H., Verhoek, M.,
Strijland, A., Bliek, J., de Meulemeester, T. M.
A. M. O., Mannens, M. M. A. M., and Aerts, J.
M. F. G. (1998) J. Biol. Chem. 273, 2568025685
157. Semino, C. E., Specht, C. A., Raimondi, A.,
and Robbins, P. W. (1996) Proc. Natl. Acad. Sci.
U.S.A. 93, 45484553
158. Kamst, E., Bakkers, J., Quaedvlieg, N. E. M.,
Pilling, J., Kijne, J. W., Lugtenberg, B. J. J., and
Spaink, H. P. (1999) Biochemistry 38, 4045
4052
159. Gibeaut, D., and Carpita, N. C. (1994) FASEB
J. 8, 904915
160. Darvill, S., McNeil, M., Albersheim, P., and
Delmer, D. P. (1980) in The Biochemisty of
Plants, Vol. I (Tolbert, N. E., ed), pp. 91162,
Academic Press, New York
161. Preston, R. D. (1979) Ann. Rev. Plant Physiol.
30, 55
162. Brett, C. T., and Hillman, J. R., eds. (1985)
Biochemistry of Plant Cell Walls, Cambridge
Univ. Press, Cambridge
163. MacKay, A. L., Wallace, J. C., Sasaki, K., and
Taylor, I. E. P. (1988) Biochemistry 27, 14671473
163a. Carpita, N., and McCann, M. (2000) in
Biochemistry and Molecular Biology of Plants
(Buchanan, B. B., Gruissem, W., and Jones,
R. L., eds), pp. 52 108, American Society of
Plant Physiologists, Rockville, Maryland
164. Northcote, D. H. (1972) Ann. Rev. Plant
Physiol. 23, 113
164a. Perrin, R. M., DeRocher, A. E., Bar-Peled, M.,
Zeng, W., Norambuena, L., Orellana, A.,
Raikhel, N. V., and Keegstra, K. (1999)
Science 284, 1976 1979
164b. Kofod, L. V., Kauppinen, S., Christgau, S.,
Andersen, L. N., Heldt-Hansen, H. P.,
Drreich, K., and Dalbge, H. (1994) J. Biol.
Chem. 269, 29182 29189
164c. ONeill, M. A., Warrenfeltz, D., Kates, K.,
Pellerin, P., Doco, T., Darvill, A. G., and
Albersheim, P. (1996) J. Biol. Chem. 271,
22923 22930
164d. Ishii, T., Matsunaga, T., Pellerin, P., ONeill,
M. A., Darvill, A., and Albersheim, P. (1999)
J. Biol. Chem. 274, 13098 13104
164e. Hfte, H. (2001) Science 294, 795 797
164f. ONeill, M. A., Eberhard, S., Albersheim, P.,
and Darvill, A. G. (2001) Science 294, 846 849
References
Ch 20 IND page-E- 2/14/03, 11:31 AM 1175
1176 Chapter 20. Some Pathways of Carbohydrate Metabolism
192. Pavo, M. S. G., Aiello, K. R. M., Werneck, C.
C., Silva, L. C. F., Valente, A.-P., Mulloy, B.,
Colwell, N. S., Tollefsen, D. M., and Mouro,
P. A. S. (1998) J. Biol. Chem. 273, 2784827857
193. Sugumaran, G., Katsman, M., and Drake, R.
R. (1995) J. Biol. Chem. 270, 2248322487
194. Yamauchi, S., Hirahara, Y., Usui, H., Takeda,
Y., Hoshino, M., Fukuta, M., Kimura, J. H.,
and Habuchi, O. (1999) J. Biol. Chem. 274,
24562463
195. Kobayashi, M., Habuchi, H., Habuchi, O.,
Saito, M., and Kimata, K. (1996) J. Biol. Chem.
271, 76457653
196. Razi, N., and Lindahl, U. (1995) J. Biol. Chem.
270, 1126711275
197. Shworak, N. W., Liu, J., Petros, L. M., Zhang,
L., Kobayashi, M., Copeland, N. G., Jenkins,
N. A., and Rosenberg, R. D. (1999) J. Biol.
Chem. 274, 51705184
198. Toma, L., Berninsone, P., and Hirschberg, C.
B. (1998) J. Biol. Chem. 273, 2245822465
199. Aikawa, J.-i, and Esko, J. D. (1999) J. Biol.
Chem. 274, 26902695
200. Scott, J. E., Heatley, F., and Wood, B. (1995)
Biochemistry 34, 1546715474
201. Yanagashita, M., and Hascall, V. C. (1992) J.
Biol. Chem. 267, 94519454
202. Lane, D. A., and Bjrk, I., eds. (1992) Heparin
and Related Polysaccharides, Plenum, New York
203. Lindahl, U., Kusche-Gullberg, M., and Kjelln,
L. (1998) J. Biol. Chem. 273, 2497924982
203a. Safaiyan, F., Lindahl, U., and Salmivirta, M.
(2000) Biochemistry 39, 10823 10830
203b. Zhang, L., Lawrence, R., Schwartz, J. J., Bai,
X., Wei, G., Esko, J. D., and Rosenberg, R. D.
(2001) J. Biol. Chem. 276, 28806 28813
204. Kinoshita, A., Yamada, S., Haslam, S. M.,
Morris, H. R., Dell, A., and Sugahara, K.
(1997) J. Biol. Chem. 272, 1965619665
205. Nadanaka, S., Clement, A., Masayama, K.,
Faissner, A., and Sugahara, K. (1998) J. Biol.
Chem. 273, 32963307
206. Mouro, P. A. S., Pereira, M. S., Pavo, M. S.
G., Mulloy, B., Tollefsen, D. M., Mowinckel,
M.-C., and Abildgaard, U. (1996) J. Biol. Chem.
271, 2397323984
207. Petitou, M., Hrault, J.-P., Bernat, A., Driguez,
P.-A., Duchaussoy, P., Lormeau, J.-C., and
Herbert, J.-M. (1999) Nature (London) 398,
417422
208. Sina, P. (1999) Nature (London) 398, 377378
208a. Zehnder, J. L., and Galli, S. J. (1999) Nature
(London) 400, 714 715
208b. Hondal, R. J., Ma, S., Caprioli, R. M., Hill, K.
E., and Burk, R. F. (2001) J. Biol. Chem. 276,
15823 15831
209. Spiro, R. C., Casteel, H. E., Laufer, D. M.,
Reisfeld, R. A., and Harper, J. R. (1989) J. Biol.
Chem. 264, 17791786
210. Iozzo, R. V., and Murdoch, A. D. (1996) FASEB
J. 10, 598614
211. Cheng, F., Heinegrd, D., Fransson, L.-.,
Bayliss, M., Bielicki, J., Hopwood, J., and Yoshida,
K. (1996) J. Biol. Chem. 271, 2857228580
212. Hauser, N., Paulsson, M., Heinegrd, D., and
Mrgelin, M. (1996) J. Biol. Chem. 271, 32247
32252
213. Zheng, J., Luo, W., and Tanzer, M. L. (1998) J.
Biol. Chem. 273, 1299913006
213a. Olin, A. I., Mrgelin, M., Sasaki, T., Timpl,
R., Heinegrd, D., and Aspberg, A. (2001) J.
Biol. Chem. 276, 1253 1261
214. Arner, E. C., Pratta, M. A., Trzaskos, J. M.,
Decicco, C. P., and Tortorella, M. D. (1999) J.
Biol. Chem. 274, 65946601
215. Brown, G. M., Huckerby, T. N., Bayliss, M. T.,
and Nieduszynski, I. A. (1998) J. Biol. Chem.
273, 2640826414
References
216. Lindahl, B., Eriksson, L., Spillmann, D.,
Caterson, B., and Lindahl, U. (1996) J. Biol.
Chem. 271, 1699116994
217. Dours-Zimmermann, M. T., and
Zimmermann, D. R. (1994)
J. Biol. Chem. 269, 3299232998
217a. Yang, B. L., Cao, L., Kiani, C., Lee, V., Zhang,
Y., Adams, M. E., and Yang, B. B. (2000) J.
Biol. Chem. 275, 21255 21261
217b. Pangalos, M. N., Efthimiopoulos, S., Shioi, J.,
and Robakis, N. K. (1995) J. Biol. Chem. 270,
10388 10391
218. Halfter, W., Dong, S., Schurer, B., and Cole, G.
J. (1998) J. Biol. Chem. 273, 2540425412
219. Scott, J. E. (1996) Biochemistry 35, 87958799
220. Weber, I. T., Harrison, R. W., and Iozzo, R. V.
(1996) J. Biol. Chem. 271, 3176731770
221. Tai, G.-H., Huckerby, T. N., and Nieduszynski,
I. A. (1996) J. Biol. Chem. 271, 2353523546
222. Ungefroren, H., and Krull, N. B. (1996) J. Biol.
Chem. 271, 1578715795
222a. Yang, V. W.-C., LaBrenz, S. R., Rosenberg, L.
C., McQuillan, D., and Hk, M. (1999) J.
Biol. Chem. 274, 12454 12460
223. Sommarin, Y., Wendel, M., Shen, Z., Hellman,
U., and Heinegrd, D. (1998) J. Biol. Chem. 273,
1672316729
223a. Iozzo, R. V. (1999) J. Biol. Chem. 274, 18843
18846
223b. Saika, S., Shiraishi, A., Saika, S., Liu, C.-Y.,
Funderburgh, J. L., Kao, C. W.-C., Converse,
R. L., and Kao, W. W.-Y. (2000) J. Biol. Chem.
275, 2607 2612
223c. Bengtsson, E., Aspberg, A., Heinegrd, D.,
Sommarin, Y., and Spillmann, D. (2000) J.
Biol. Chem. 275, 40695 40702
224. David, C. L., Orpiszewski, J., Zhu, X.-C.,
Reissner, K. J., and Aswad, D. W. (1998) J. Biol.
Chem. 273, 3206332070
225. Grumet, M., Milev, P., Sakurai, T.,
Karthikeyan, L., Bourdon, M., Margolis, R. K.,
and Margolis, R. U. (1994) J. Biol. Chem. 269,
1214212146
226. Fernndez-Busquets, X., Kammerer, R. A.,
and Burger, M. M. (1996) J. Biol. Chem. 271,
2355823565
227. Varner, J. A. (1996) J. Biol. Chem. 271, 16119
16125
228. Hart, G. W., Holt, G. D., and Haltiwanger, R.
S. (1988) Trends Biochem. Sci. 13, 380384
229. Sadler, J. E. (1984) in Biology of Carbohy-
drates, Vol. 2 (Ginsburg, V., and Robbins, P.
W., eds), pp. 199288, Wiley, New York
230. Jentoft, N. (1990) Trends Biochem. Sci. 15, 291
294
231. Dong, D. L.-Y., Xu, Z.-S., Chevrier, M. R.,
Cotter, R. J., Cleveland, D. W., and Hart, G. W.
(1993) J. Biol. Chem. 268, 1667916687
231a. Varki, A., Cummings, R., Esko, J., Freeze, H.,
Hart, G., and Marth, J., eds. (1999) Essentials
of Glycobiology, Cold Spring Harbor Lab.
Press, Plainview, New York
231b. Roseman, S. (2001) J. Biol. Chem. 276, 41527
41542
232. Hanisch, F.-G., Mller, S., Hassan, H.,
Clausen, H., Zachara, N., Gooley, A. A.,
Paulsen, H., Alving, K., and Peter-Katalinic, J.
(1999) J. Biol. Chem. 274, 99469954
233. Schwientek, T., Nomoto, M., Levery, S. B.,
Merkx, G., van Kessel, A. G., Bennett, E. P.,
Hollingsworth, M. A., and Clausen, H. (1999)
J. Biol. Chem. 274, 45044512
233a. Comer, F. I., and Hart, G. W. (2000) J. Biol.
Chem. 275, 29179 29182
233b. Wells, L., Vosseller, K., and Hart, G. W.
(2001) Science 291, 2376 2378
233c. Hanover, J. A. (2001) FASEB J. 15, 1865 1876
234. Abeijon, C., and Hirschberg, C. B. (1992)
Trends Biochem. Sci. 17, 3236
235. Moloney, D. J., Lin, A. I., and Haltiwanger, R.
S. (1997) J. Biol. Chem. 272, 1904619050
236. Allen, A. (1983) Trends Biochem. Sci. 8, 169173
237. Yeh, J.-C., Ong, E., and Fukuda, M. (1999) J.
Biol. Chem. 274, 32153221
238. Gum, J. R., Jr., Ho, J. J. L., Pratt, W. S., Hicks, J.
W., Hill, A. S., Vinall, L. E., Roberton, A. M.,
Swallow, D. M., and Kim, Y. S. (1997) J. Biol.
Chem. 272, 266786686
239. Desseyn, J.-L., Buisine, M.-P., Porchet, N., and
Aubert, J.-P. (1998) J. Biol. Chem. 273, 30157
30164
240. Perez-Vilar, J., and Hill, R. L. (1998) J. Biol.
Chem. 273, 3452734534
241. Sagami, H., Kurisaki, A., and Ogura, K. (1993)
J. Biol. Chem. 268, 1010910113
242. Opdenakker, G., Rudd, P. M., Ponting, C. P.,
and Dwek, R. A. (1993) FASEB J. 7, 13301337
243. Gahmberg, C. G., and Tolvanen, M. (1996)
Trends Biochem. Sci. 21, 308311
244. Manzella, S. M., Hooper, L. V., and Baenziger,
J. U. (1996) J. Biol. Chem. 271, 1211712120
245. Drickamer, K., and Taylor, M. E. (1998) Trends
Biochem. Sci. 23, 321324
246. Elbein, A. D. (1981) Trends Biochem. Sci. 6,
219221
247. Zhu, X., Zeng, Y., and Lehrman, M. A. (1992)
J. Biol. Chem. 267, 88958902
248. Rajput, B., Muniappa, N., and Vijay, I. K.
(1994) J. Biol. Chem. 269, 1605416061
249. Zara, J., and Lehrman, M. A. (1994) J. Biol.
Chem. 269, 1910819115
249a. nligil, U. M., Zhou, S., Yuwaraj, S., Sarkar,
M., Schachter, H., and Rini, J. M. (2000)
EMBO J. 19, 5269 5280
250. Silberstein, S., and Gilmore, R. (1996) FASEB J.
10, 849858
251. Imperiali, B., and Shannon, K. L. (1991)
Biochemistry 30, 43744380
252. Yan, Q., Prestwich, G. D., and Lennarz, W. J.
(1999) J. Biol. Chem. 274, 50215025
252a. Chen, X., VanValkenburgh, C., Liang, H.,
Fang, H., and Green, N. (2001) J. Biol. Chem.
276, 2411 2416
252b. Karaoglu, D., Kelleher, D. J., and Gilmore, R.
(2001) Biochemistry 40, 12193 12206
253. Schwarz, R. T., and Datema, R. (1984) Trends
Biochem. Sci. 9, 3234
254. Elbein, A. D. (1987) Ann. Rev. Biochem. 56,
497534
255. Kaushal, G. P., Pan, Y. T., Tropea, J. E.,
Mitchell, M., Liu, P., and Elbein, A. D. (1988) J.
Biol. Chem. 263, 1727817283
256. Kukuruzinska, M. A., Bergh, M. L. E., and
Jackson, B. J. (1987) Ann. Rev. Biochem. 56,
915944
257. Hatton, M. W. C., Marz, L., and Regoeczi, E.
(1983) Trends Biochem. Sci. 8, 287291
257a. Valle, F., Karaveg, K., Herscovics, A.,
Moremen, K. W., and Howell, P. L. (2000) J.
Biol. Chem. 275, 41287 41298
257b. Van Petegem, F., Contreras, H., Contreras,
R., and Van Beeumen, J. (2001) J. Mol. Biol.
312, 157 165
258. Herscovics, A., and Orlean, P. (1993) FASEB J.
7, 540550
258a. Dell, A., and Morris, H. R. (2001) Science 291,
2351 2356
259. Dolci, E. D., and Palade, G. E. (1985) J. Biol.
Chem. 260, 1072810735
260. Gonatas, J. O., Mezitis, S. G. E., Stieber, A.,
Fleischer, B., and Gonatas, N. K. (1989) J. Biol.
Chem. 264, 646653
261. Hooper, L. V., Hindsgaul, O., and Baenziger, J.
U. (1995) J. Biol. Chem. 270, 1632716332
262. Okajima, T., Fukumoto, S., Miyazaki, H.,
Ishida, H., Kiso, M., Furukawa, K., Urano, T.,
and Furukawa, K. (1999) J. Biol. Chem. 274,
1147911486
Ch 20 IND page-E- 2/14/03, 11:31 AM 1176
1177
263. Lee, Y.-C., Kaufmann, M., Kitazume-
Kawaguchi, S., Kono, M., Takashima, S.,
Kurosawa, N., Liu, H., Pircher, H., and Tsuji,
S. (1999) J. Biol. Chem. 274, 1195811967
264. Trimble, R. B., and Atkinson, P. H. (1986)
J. Biol. Chem. 261, 98159824
265. Chiba, Y., Suzuki, M., Yoshida, S., Yoshida, A.,
Ikenaga, H., Takeuchi, M., Jigami, Y., and
Ichishima, E. (1998) J. Biol. Chem. 273, 26298
26304
266. Kollr, R., Reinhold, B. B., Petrkova, E., Yeh,
H. J. C., Ashwell, G., Drgonov, J., Kapteyn, J.
C., Klis, F. M., and Cabib, E. (1997) J. Biol.
Chem. 272, 1776217775
266a. Helenius, A., and Aebi, M. (2001) Science
291, 2364 2369
266b. Lehrman, M. A. (2001) J. Biol. Chem. 276,
8623 8626
266c. Bertozzi, C. R., and Kiessling, L. L. (2001)
Science 291, 2357 2364
266d. Plante, O. J., Palmacci, E. R., and Seeberger,
P. H. (2001) Science 291, 1523 1527
267. Mazzarello, P., and Bentivoglio, M. (1998)
Nature (London) 392, 543544
268. Featherstone, C. (1998) Science 282, 21722174
269. Berger, E. G., and Roth, J., eds. (1997) The
Golgi Apparatus, Birkhuser Verlag, Basel
270. Driouich, A., Faye, L., and Staehelin, L. A.
(1993) Trends Biochem. Sci. 18, 210214
271. Lazar, T., Gtte, M., and Gallwitz, D. (1997)
Trends Biochem. Sci. 22, 468472
272. Farquhar, M. G., and Hauri, H.-P. (1997) in
The Golgi Apparatus (Berger, E. G., and Roth,
J., eds), pp. 63129, Birkhuser Verlag, Basel,
Switzerland
273. Springer, S., and Schekman, R. (1998) Science
281, 698700
274. Walter, D. M., Paul, K. S., and Waters, M. G.
(1998) J. Biol. Chem. 273, 2956529576
275. Dice, J. F. (1990) Trends Biochem. Sci. 15, 305309
276. Rambourg, A., and Clermont, Y. (1997) in The
Golgi Apparatus (Berger, E. G., and Roth, J.,
eds), pp. 3761, Birkhuser Verlag, Basel,
Switzerland
277. Ungermann, C., and Wickner, W. (1998)
EMBO J. 17, 32693276
278. Wu, S.-K., Zeng, K., Wilson, I. A., and Balch,
W. E. (1996) Trends Biochem. Sci. 21, 472476
279. Webb, R. J., East, J. M., Sharma, R. P., and Lee,
A. G. (1998) Biochemistry 37, 673679
280. Ivessa, N. E., De Lemos-Chiarandini, C.,
Gravotta, D., Sabatini, D. D., and Kreibich, G.
(1995) J. Biol. Chem. 270, 2596025967
280a. Todorow, Z., Spang, A., Carmack, E., Yates,
J., and Schekman, R. (2000) Proc. Natl. Acad.
Sci. U.S.A. 97, 13643 13648
280b. Marsh, B. J., Mastronarde, D. N., Buttle, K.
F., Howell, K. E., and McIntosh, J. R. (2001)
Proc. Natl. Acad. Sci. U.S.A. 98, 2399 2406
280c. Jakymiw, A., Raharjo, E., Rattner, J. B.,
Eystathioy, T., Chan, E. K. L., and Fujita, D. J.
(2000) J. Biol. Chem. 275, 4137 4144
280d. Hirschberg, C. B., Robbins, P. W., and Abeijon,
C. (1998) Ann. Rev. Biochem. 67, 4969
280e. Gao, X.-D., and Dean, N. (2000) J. Biol. Chem.
275, 17718 17727
280f. Bell, A. W., and 16 other authors. (2001) J. Biol.
Chem. 276, 5152 5165
280g. Alvarez, C., Garcia-Mata, R., Hauri, H.-P., and
Sztul, E. (2001) J. Biol. Chem. 276, 26932700
280h. Charest, A., Lane, K., McMahon, K., and
Housman, D. E. (2001) J. Biol. Chem. 276,
29456 29465
280i. Demaurex, N., Furuya, W., DSouza, S.,
Bonifacino, J. S., and Grinstein, S. (1998)
J. Biol. Chem. 273, 2044 2051
281. Fritzler, M. J., Lung, C.-C., Hamel, J. C.,
Griffith, K. J., and Chan, E. K. L. (1995) J. Biol.
Chem. 270, 3126231268
References
282. Bascon, R. A., Srinivasan, S., and Nussbaum,
R. L. (1999) J. Biol. Chem. 274, 29532962
283. Kain, R., Angata, K., Kerjaschki, D., and
Fukuda, M. (1998) J. Biol. Chem. 273, 981988
284. Johannes, L., Tenza, D., Antony, C., and Goud,
B. (1997) J. Biol. Chem. 272, 1955419561
285. Drickamer, K. (1988) J. Biol. Chem. 263, 9557
9560
286. Cantor, A. B., and Kornfeld, S. (1992) J. Biol.
Chem. 267, 2335723363
287. Kornfeld, S. (1992) Ann. Rev. Biochem. 61,
307330
288. Pohlmann, R., Boeker, M. W. C., and von
Figura, K. (1995) J. Biol. Chem. 270, 27311
27318
289. Sleat, D. E., and Lobel, P. (1997) J. Biol. Chem.
272, 731738
290. York, S. J., Arneson, L. S., Gregory, W. T.,
Dahms, N. M., and Kornfeld, S. (1999) J. Biol.
Chem. 274, 11641171
290a. Zhu, Y., Doray, B., Poussu, A., Lehto, V.-P.,
and Kornfeld, S. (2001) Science 292, 1716
1718
291. Sleat, D. E., Sohar, I., Lackland, H., Majercak,
J., and Lobel, P. (1996) J. Biol. Chem. 271,
1919119198
292. Klionsky, D. J., and Emr, S. D. (1990) J. Biol.
Chem. 265, 53495352
293. Lodish, H. F. (1991) Trends Biochem. Sci. 16,
374377
294. Chao, W., Liu, H., Hanahan, D. J., and Olson,
M. S. (1992) J. Biol. Chem. 267, 67256735
295. Chiu, M. H., Thomas, V. H., Stubbs, H. J., and
Rice, K. G. (1995) J. Biol. Chem. 270, 24024
24031
296. Reichner, J. S., Whiteheart, S. W., and Hart, G.
W. (1988) J. Biol. Chem. 263, 1631616326
296a. Filipe, S. R., Severina, E., and Tomasz, A.
(2001) J. Biol. Chem. 276, 39618 39628
297. Emanuele, J. J., Jr., Jin, H., Yanchunas, J., Jr.,
and Villafranca, J. J. (1997) Biochemistry 36,
72647271
298. Anderson, M. S., Eveland, S. S., Onishi, H. R.,
and Pompliano, D. L. (1996) Biochemistry 35,
1626416269
299. Duncan, K., van Heijenoort, J., and Walsh, C.
T. (1990) Biochemistry 29, 23792386
300. Van Heijenoort, J. (1994) in Bacterial Cell Wall
(New Comprehensive Biochemistry), Vol. 27
(Ghuysen, J.-M., and Hakenbeck, R., eds), pp.
3954, Elsevier, Amsterdam
301. Matsuhashi, M. (1994) in Bacterial Cell Wall
(New Comprehensive Biochemistry), Vol. 27
(Ghuysen, J.-M., and Hakenbeck, R., eds), pp.
5571, Elsevier, Amsterdam
301a. Ha, S., Chang, E., Lo, M.-C., Men, H., Park,
P., Ge, M., and Walker, S. (1999) J. Am. Chem.
Soc. 121, 8417 8426
301b. Marmor, S., Petersen, C. P., Reck, F., Yang,
W., Gao, N., and Fisher, S. L. (2001)
Biochemistry 40, 12207 12214
301c. Gordon, E., Flouret, B., Chantalat, L., van
Heijenoort, J., Mengin-Lecreulx, D., and
Dideberg, O. (2001) J. Biol. Chem. 276, 10999
11006
301d. Yan, Y., Munshi, S., Leiting, B., Anderson, M.
S., Chrzas, J., and Chen, Z. (2000) J. Mol. Biol.
304, 435 445
301e. Lee, W., McDonough, M. A., Kotra, L. P., Li,
Z.-H., Silvaggi, N. R., Takeda, Y., Kelly, J. A.,
and Mobashery, S. (2001) Proc. Natl. Acad.
Sci. U.S.A. 98, 1427 1431
302. Koch, A. L. (1985) Trends Biochem. Sci. 10, 1114
303. Archibald, A. R., Hancock, I. C., and
Harwood, C. R. (1993) in Bacillus subtilis and
Other Gram-Positive Bacteria (Sonenshein, A.
L., Hoch, J. A., and Losick, R., eds), pp. 381
410, American Society for Microbiology,
Washington, D. C.
304. Jacobs, C. (1997) Science 278, 17311732
305. Jacobs, C., Huang, L.-j, Bartowsky, E.,
Normark, S., and Park, J. T. (1994) EMBO J.
13, 46844694
305a. Templin, M. F., Ursinus, A., and Hltje, J.-V.
(1999) EMBO J. 18, 4108 4117
306. Keenleyside, W. J., and Whitfield, C. (1996) J.
Biol. Chem. 271, 2858128592
307. Raetz, C. R. H., Ulevitch, R. J., Wright, S. D.,
Sibley, C. H., Ding, A., and Nathan, C. F.
(1991) FASEB J. 5, 26522660
308. Odegaard, T. J., Kaltashov, I. A., Cotter, R. J.,
Steeghs, L., van der Ley, P., Khan, S., Maskell,
D. J., and Raetz, C. R. H. (1997) J. Biol. Chem.
272, 1968819696
308a. Raetz, C. R. H., and Whitfield, C. (2002) Ann.
Rev. Biochem. 71, 635700
309. Yethon, J. A., Heinrichs, D. E., Monteiro, M.
A., Perry, M. B., and Whitfield, C. (1998) J.
Biol. Chem. 273, 2631026316
310. Raetz, C. R. H., and Roderick, S. L. (1995)
Science 270, 9971000
311. Jackman, J. E., Raetz, C. R. H., and Fierke, C.
A. (1999) Biochemistry 38, 19021911
312. Zhou, Z., White, K. A., Polissi, A., Georgopoulos,
C., and Raetz, C. R. H. (1998) J. Biol. Chem.
273, 1246612475
313. Luk, J. M. C., Lind, S. M., Tsang, R. S. W., and
Lindberg, A. A. (1991) J. Biol. Chem. 266,
2321523225
314. Kerwood, D. E., Schneider, H., and Yamasaki,
R. (1992) Biochemistry 31, 1276012768
315. Basu, S. S., York, J. D., and Raetz, C. R. H.
(1999) J. Biol. Chem. 274, 1113911149
316. Rietschel, E. T., Kirikae, T., Schade, F. U.,
Mamat, U., Schmidt, G., Loppnow, H., Ulmer,
A. J., Zhringer, U., Seydel, U., Di Padova, F.,
Schreier, M., and Brade, H. (1994) FASEB J. 8,
217225
317. White, K. A., Kaltashov, I. A., Cotter, R. J.,
and Raetz, C. R. H. (1997) J. Biol. Chem. 272,
1655516563
318. Brooke, J. S., and Valvano, M. A. (1996) J. Biol.
Chem. 271, 36083614
319. Ssskind, M., Brade, L., Brade, H., and Holst,
O. (1998) J. Biol. Chem. 273, 70067017
320. Pavliak, V., Brisson, J.-R., Michon, F., Uhrn,
D., and Jennings, H. J. (1993) J. Biol. Chem.
268, 1414614152
321. Phillips, N. J., Apicella, M. A., Griffiss, J. M.,
and Gibson, B. W. (1993) Biochemistry 32,
20032012
321a. White, K. A., Lin, S., Cotter, R. J., and Raetz,
C. R. H. (1999) J. Biol. Chem. 274, 31391 31400
321b. Kooistra, O., Lneberg, E., Lindner, B.,
Knirel, Y. A., Frosch, M., and Zhringer, U.
(2001) Biochemistry 40, 7630 7640
322. Ganfield, M.-C. W., and Pieringer, R. A. (1980)
J. Biol. Chem. 255, 51645169
322a. Volkman, B. F., Zhang, Q., Debabov, D. V.,
Rivera, E., Kresheck, G. C., and Neuhaus, F.
C. (2001) Biochemistry 40, 7964 7972
323. Ton-That, H., Labischinski, H., Berger-Bchi,
B., and Schneewind, O. (1998) J. Biol. Chem.
273, 2914329149
324. Fischetti, V. A. (1991) Sci. Am. 264(Jun), 5865
325. Michon, F., Brisson, J.-R., Dell, A., Kasper, D.
L., and Jennings, H. J. (1988) Biochemistry 27,
53415351
326. Cassels, F. J., Fales, H. M., London, J., Carlson,
R. W., and van Halbeek, H. (1990) J. Biol.
Chem. 265, 1412714135
327. Wolucka, B. A., McNeil, M. R., de Hoffmann,
E., Chonjnacki, T., and Brennan, P. J. (1994) J.
Biol. Chem. 269, 2332823335
328. Besra, G. S., McNeil, M. R., Rivoire, B., Khoo,
K.-H., Morris, H. R., Dell, A., and Brennan, P.
J. (1993) Biochemistry 32, 347355
Ch 20 IND page-E- 2/14/03, 11:31 AM 1177
1178 Chapter 20. Some Pathways of Carbohydrate Metabolism
References
329. Beveridge, T. J., and Koval, S. F., eds. (1993)
Advances in Bacterial Paracrystalline Surface
Layers, Plenum, New York
329a. Maeda, Y., Watanabe, R., Harris, C. L., Hong,
Y., Ohishi, K., Kinoshita, K., and Kinoshita,
T. (2001) EMBO J. 20, 250 261
330. Takeda, J., and Kinoshita, T. (1995) Trends
Biochem. Sci. 20, 367371
331. Udenfriend, S., and Kodukula, K. (1995) Ann.
Rev. Biochem. 64, 563591
332. Tarutani, M., Itami, S., Okabe, M., Ikawa, M.,
Tezuka, T., Yoshikawa, K., Kinoshita, T., and
Takeda, J. (1997) Proc. Natl. Acad. Sci. U.S.A.
94, 74007405
333. Menon, A. K., and Stevens, V. L. (1992) J. Biol.
Chem. 267, 1527715280
334. Medof, M. E., Nagarajan, S., and Tykocinski,
M. L. (1996) FASEB J. 10, 574586
334a. Ferguson, M. A. J. (2000) Proc. Natl. Acad. Sci.
U.S.A. 97, 10673 10675
334b. Mahoney, A. B., Sacks, D. L., Saraiva, E.,
Modi, G., and Turco, S. J. (1999) Biochemistry
38, 9813 9823
334c. Smith, T. K., Gerold, P., Crossman, A.,
Paterson, M. J., Borissow, C. N., Brimacombe,
J. S., Ferguson, M. A. J., and Schwarz, R. T.
(2002) Biochemistry 41, 1239512406
335. Ralton, J. E., and McConville, M. J. (1998) J.
Biol. Chem. 273, 42454257
336. Gilleron, M., Nigou, J., Cahuzac, B., and Puzo,
G. (1999) J. Mol. Biol. 285, 21472160
337. Benghezal, M., Benachour, A., Rusconi, S.,
Aebi, M., and Conzelmann, A. (1996) EMBO J.
15, 65756583
338. Zawadzki, J., Scholz, C., Currie, G., Coombs,
G. H., and McConville, M. J. (1998) J. Mol.
Biol. 282, 287299
339. Smith, T. K., Sharma, D. K., Crossman, A.,
Dix, A., Brimacombe, J. S., and Ferguson, M.
A. J. (1997) EMBO J. 16, 66676675
339a. Morita, Y. S., Acosta-Serrano, A., Buxbaum,
L. U., and Englund, P. T. (2000) J. Biol. Chem.
275, 14147 14154
340. Leidich, S. D., and Orlean, P. (1996) J. Biol.
Chem. 271, 2782927837
341. Chien, J.-L., and Hogan, E. L. (1983) J. Biol.
Chem. 258, 1072710730
342. van Echten, G., and Sandhoff, K. (1993) J. Biol.
Chem. 268, 53415344
343. Sprong, H., Kruithof, B., Leijendekker, R.,
Slot, J. W., van Meer, G., and van der Sluijs, P.
(1998) J. Biol. Chem. 273, 2588025888
344. Schwientek, T., Almeida, R., Levery, S. B.,
Holmes, E. H., Bennett, E., and Clausen, H.
(1998) J. Biol. Chem. 273, 2933129340
345. Lannert, H., Gorgas, K., Meissner, I., Wieland,
F. T., and Jeckel, D. (1998) J. Biol. Chem. 273,
29392946
346. Jaskiewicz, E., Zhu, G., Bassi, R., Darling, D.
S., and Young, W. W., Jr. (1996) J. Biol. Chem.
271, 2639526403
347. Neufeld, E. F., Lim, T. W., and Shapiro, L. J.
(1975) Ann. Rev. Biochem. 44, 357376
348. Neufeld, E. F. (1991) Ann. Rev. Biochem. 60,
257280
349. Tager, J. M. (1985) Trends Biochem. Sci. 10, 324
326
350. Scriver, C. R., Beaudet, A. L., Sly, W. S., and
Valle, D., eds (1995) The Metabolic and
Molecular Bases of Inherited Disease, 7th ed.,
Vol. 1, McGraw-Hill, New York (pp. 2427
2879)
351. von Figura, K., and Hasilik, A. (1984) Trends
Biochem. Sci. 9, 2931
352. Neufeld, E. F., and Muenzer, J. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2465
2494, McGraw-Hill, New York
353. Stoltzfus, L. J., Sosa-Pineda, B., Moskowitz, S.
M., Menon, K. P., Dlott, B., Hooper, L.,
Teplow, D. B., Shull, R. M., and Neufeld, E. F.
(1992) J. Biol. Chem. 267, 65706575
354. Kresse, H., Paschke, E., von Figura, K.,
Gilberg, W., and Fuchs, W. (1980) Proc. Natl.
Acad. Sci. U.S.A. 77, 68226826
355. Wilson, P. J., Morris, C. P., Anson, D. S.,
Occhiodoro, T., Bielicki, J., Clements, P. R.,
and Hopwood, J. J. (1990) Proc. Natl. Acad. Sci.
U.S.A. 87, 85318535
356. Wicker, G., Prill, V., Brooks, D., Gibson, G.,
Hopwood, J., von Figura, K., and Peters, C.
(1991) J. Biol. Chem. 266, 2138621391
357. Wu, B. M., Tomatsu, S., Fukuda, S.,
Sukegawa, K., Orii, T., and Sly, W. S. (1994) J.
Biol. Chem. 269, 2368123688
358. McGovern, M. M., Aula, P., and Desnick, R. J.
(1983) J. Biol. Chem. 258, 1074310747
359. Mononen, I., Fisher, K. J., Kaartinen, V., and
Aronson, N. N., Jr. (1993) FASEB J. 7, 12471256
360. Saarela, J., Laine, M., Tikkanen, R., Oinonen,
C., Jalanko, A., Rouvinen, J., and Peltonen, L.
(1998) J. Biol. Chem. 273, 2532025328
361. Rothman, J. E., and Lenard, J. (1984) Trends
Biochem. Sci. 9, 176178
362. Beutler, E. (1992) Science 256, 794799
363. Beutler, E. (1993) Proc. Natl. Acad. Sci. U.S.A.
90, 53845390
364. Beutler, E., and Grabowski, G. A. (1995) in The
Metabolic and Molecular Bases of Inherited
Disease, 7th ed., Vol. 2 (Scriver, C. R., Beaudet,
A. L., Sly, W. S., and Valle, D., eds), pp. 2641
2670, McGraw-Hill, New York
365. Sano, A., Radin, N. S., Johnson, L. L., and Tarr,
G. E. (1988) J. Biol. Chem. 263, 1959719601
366. Myerowitz, R., and Costigan, F. C. (1988) J.
Biol. Chem. 263, 1858718589
366a. Mark, B. L., Vocadlo, D. J., Knapp, S., Triggs-
Raine, B. L., Withers, S. G., and James, M. N.
G. (2001) J. Biol. Chem. 276, 10330 10337
367. Neufeld, E. F. (1989) J. Biol. Chem. 264, 10927
10930
368. Gravel, R. A., Clarke, J. T. R., Kaback, M. M.,
Mahuran, D., Sandhoff, K., and Suzuki, K.
(1995) in The Metabolic and Molecular Bases of
Inherited Disease, 7th ed., Vol. 1 (Scriver, C. R.,
Beaudet, A. L., Sly, W. S., and Valle, D., eds),
pp. 28392879, McGraw-Hill, New York
369. Paw, B. H., Moskowitz, S. M., Uhrhammer,
N., Wright, N., Kaback, M. M., and Neufeld,
E. F. (1990) J. Biol. Chem. 265, 94529457
369a. Kawai, H., Allende, M. L., Wada, R., Kono,
M., Sango, K., Deng, C., Miyakawa, T.,
Crawley, J. N., Werth, N., Bierfreund, U.,
Sandhoff, K., and Proia, R. L. (2001) J. Biol.
Chem. 276, 6885 6888
370. Hama, Y., Li, Y.-T., and Li, S.-C. (1997) J. Biol.
Chem. 272, 28282833
371. Beutler, E. (1981) Trends Biochem. Sci. 6, 9597
371a. Schiffmann, R., and 21 other authors (2000)
Proc. Natl. Acad. Sci. U.S.A. 97, 365 370
371b. Sly, W. S., Vogler, C., Grubb, J. H., Zhou, M.,
Jiang, J., Zhou, X. Y., Tomatsu, S., Bi, Y., and
Snella, E. M. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 2205 2210
372. Furbish, F. S., Steer, C. J., Barranger, J. A.,
Jones, E. A., and Brady, R. O. (1978) Biochem.
Biophys. Res. Commun. 81, 10471053
373. Krivit, W., Pierpont, M. E., Ayaz, K., Tsai, M.,
Ramsay, N. K. C., Kersey, J. H., Weisdorf, S.,
Sibley, R., Snover, D., McGovern, M. M.,
Schwartz, M. F., and Desnick, R. J. (1984) N.
Engl. J. Med. 311, 16061611
374. Sorge, J., Kuhl, W., West, C., and Beutler, E.
(1987) Proc. Natl. Acad. Sci. U.S.A. 84, 906909
375. Fairbairn, L. J., Lashford, L. S., Spooncer, E.,
McDermott, R. H., Lebens, G., Arrand, J. E.,
Arrand, J. R., Bellantuono, I., Holt, R., Hatton,
C. E., Cooper, A., Besley, G. T. N., Wraith, J. E.,
Anson, D. S., Hopwood, J. J., and Dexter, T.
M. (1996) Proc. Natl. Acad. Sci. U.S.A. 93,
20252030
375a. Qin, G., Takenaka, T., Telsch, K., Kelley, L.,
Howard, T., Levade, T., Deans, R., Howard,
B. H., Malech, H. L., Brady, R. O., and
Medin, J. A. (2001) Proc. Natl. Acad. Sci.
U.S.A. 98, 3428 3433
376. Beaudet, A. L., Scriver, C. R., Sly, W. S., and
Valle, D. (1995) in The Metabolic and Molecular
Bases of Inherited Disease, 7th ed., Vol. 1
(Scriver, C. R., Beaudet, A. L., Sly, W. S., and
Valle, D., eds), pp. 53118, McGraw-Hill,
New York
377. Brenkert, A., Arora, R. C., Radin, N. S., Meier,
H., and MacPike, A. D. (1972) Brain Res. 36,
195202
378. Morell, P., and Constantino-Ceccarini, E.
(1972) Lipids 7, 266268
379. Seppala, R., Tietze, F., Krasnewich, D., Weiss,
P., Ashwell, G., Barsh, G., Thomas, G. H.,
Packman, S., and Gahl, W. A. (1991)
J. Biol. Chem. 266, 74567461
380. Spellacy, E., Shull, R. M., Constantopoulos,
G., and Neufeld, E. F. (1983) Proc. Natl. Acad.
Sci. U.S.A. 80, 60916095
381. Shull, R. M., Kakkis, E. D., McEntee, M. F.,
Kania, S. A., Jonas, A. J., and Neufeld, E. F.
(1994) Proc. Natl. Acad. Sci. U.S.A. 91, 12937
12941
382. Walkley, S. U., Thrall, M. A., Dobrenis, K.,
Huang, M., March, P. A., Siegel, D. A., and
Wurzelmann, S. (1994) Proc. Natl. Acad. Sci.
U.S.A. 91, 29702974
Ch 20 IND page-E- 2/14/03, 11:31 AM 1178
1179
Study Questions
1. Constitution of cell surface oligosaccharides or
polysaccharides includes the following:
D-Glucose
D-Mannose
D-Galactose
L-Arabinose
L-Fucose
D-Glucuronic acid
D-Neuraminic acid
Outline pathways for biosynthesis of these com-
pounds from glucose.
2. Decarboxylation steps are required for synthesis
of UDP-xylulose and UDP-apiose (Fig. 21-1).
Propose chemical mechanisms for these reactions.
3. How do animals and plants differ with respect to
transport and storage of glucose?
4. Comment on unresolved questions about the
biosynthesis of cellulose, amylose, and amylopec-
tin. What glycosyl carrier groups are required?
5. Most 5- and 6-membered sugars are found in
nature as pyranose ring forms. Why is ribose in
RNA in the furanose ring form?
6. If the ratio [ NAD
+
]/ [ NADH ] in a cell were 500
and the ratio [ NADP
+
]/ [ NADPH ] were 0.002,
what concentrations of fructose and sorbitol
would be in equilibrium with 0.1 mM glucose?
See Box 20-A and Table 6-4.
7. Write a balanced equation for reaction of boric
acid (H
3
BO
3
) with two sugar rings to give a borate
diol ester linkage (Box 20-E).
8. Describe in general terms the process by which
N-linked oligosaccharides are synthesized and
attached to proteins. What are the functions of the
ER and the Golgi?
9. What, if any, restrictions do you think should be
applied to the use of antibiotics on farms?
Ch 20 IND page-E- 2/14/03, 11:31 AM 1179
1180 Chapter 21. Specific Aspects of Lipid Metabolism
Contents
Two large fat-filled adipose cells are seen in the foreground of this scanning
electron micrograph. They are part of a larger cluster of cells from rat tissue.
Delicate strands of connective tissue fibers intertwine the cells and hold them
together. While most of the connective tissue substance has been washed away
during preparation of the specimen, the remnants give a realistic impression of
the soft, loose nature of the intercellular material. From Porter and Bonneville
(1973) Fine Structure of Cells and Tissues, Lea and Febiger, Philadelphia,
Pennsylvania. Courtesy of Mary Bonneville.
A. Digestion, Synthesis, and Distribution of
Triacylglycerols in the Human Body
1. Plasma Lipoproteins
2. Movement of Lipid Materials Between Cells
B. The Biosynthesis of Fatty Acids and Their Esters
1. Fatty Acid Synthases
2. Control of Chain Length
3. Starter Pieces and Branches
4. Synthesis by the Oxoacid Chain Elongation
Process
5. Unsaturated Fatty Acids
6. Cyclopropane Fatty Acids and Mycolic Acids
7. The Lipids of Skin and Other Surfaces
Surface lipids of plants
Formation of hydrocarbons
Insect waxes, hydrocarbons, and pheromones
C. Synthesis of Triacylglycerols, Glycolipids, and
Phospholipids
1. Phospholipids
2. The Ether-Linked Lipids
3. Sphingolipids
4. Complex Lipids in Signaling
5. Peroxidation of Lipids and Rancidity
6. Some Nutritional Questions
D. Prostaglandins and Related Prostanoid
Compounds
1. Metabolism of the Prostaglandins
2. Thromboxanes and Prostacyclins
3. Lipoxygenases
4. Leukotrienes, Lipoxins, and Related Compounds
5. Physiological Effects of the Prostanoids
6. Inflammation
7. Plant Lipoxygenases and Jasmonic Acid
E. The Polyketides
1181
1181
1184
1185
1185
1188
1189
1189
1191
1193
1195
1196
1196
1196
1196
1197
1201
1202
1202
1204
1205
1206
1206
1208
1208
1210
1210
1211
1212
1212
1218
1223
1186
1190
1194
1200
1214
1183
1183
1189
References
Study Questions
Boxes
Box 21-A Lipocalins, Fatty Acid-Binding Proteins,
and Lipophorins
Box 21-B The Essential Fatty Acids
Box 21-C Tuberculosis
Box 21-D Poly--Hydroxybutyrate and
Biodegradable Plastics
Box 21-E How the Flowers Make Their Colors
Tables
Table 21-1 Classes of Lipoprotein Particles
Table 21-2 Properties of Major Plasma
Apolipoproteins
Table 21-3 Starter Pieces for Biosynthesis of
Fatty Acids
Ch 21 IND page - E 2/14/03, 11:34 AM 1180
1181
Specific Aspects of Lipid Metabolism
21
The basic pathways for both synthesis of fatty
acids and for their -oxidation (Fig. 17-1) have been
described in Chapter 17. However, there are many
variations to these pathways, and additional sets of
enzymes are needed to synthesize the complex array
of lipids present in most organisms. We will consider
these details in this chapter. Like most other organ-
isms, human beings are able to synthesize triacylglyc-
erols (triglycerides), phospholipids, and glycolipids
needed for cell membranes. Glucose can serve as the
starting material. However, dietary lipids are also a
major source. For this reason, we will start with a
discussion of the digestion and uptake of lipids and of
the distribution by way of the bloodstream of ingested
lipids and of lipids synthesized in the liver or in other
tissues.
A. Digestion, Synthesis, and Distribution of
Triacylglycerols in the Human Body
Digestion of triglycerides begins in the stomach
with emulsification and partial digestion by gastric
lipase. Within the small intestine the ~ 100-residue
protein called colipase
1 2a
binds to the surface of the
fat droplets and provides an attachment site for the
449-residue pancreatic lipase. This Ca
2+
-dependent
serine esterase cleaves each triglyceride to two mole-
cules of fatty acid and one of a 2-monoacylglycerol.
3 5
These products are emulsified by bile salts (Fig. 22-10)
and are then taken up by the cells of the intestinal
lining. The fatty acids are converted to acyl-CoA
esters which transfer their acyl groups to the monoa-
cylglycerols to resynthesize the triacylglycerols.
6
The
latter are incorporated into the very large lipoprotein
particles called chylomicrons (Table 21-1) and enter
the bloodstream via the lymphatic system (Fig. 21-1).
7
Free fatty acids are also transported as complexes with
serum albumin.
Synthesis of lipids from carbohydrates is an effi-
cient process, which occurs largely in the liver and also
in intestinal epithelial cells.
6
The newly synthesized
triacylglycerols, together with smaller amounts of
phospholipids and cholesterol, combine with specific
apolipoproteins, which are also synthesized in the
liver, to form very low density lipoprotein (VLDL)
particles which are secreted into the blood stream.
They transport the newly formed triacylglycerols from
the liver to other body cells including the adipocytes,
which store excess fat (Fig. 21-1).
1. Plasma Lipoproteins
The small particles of plasma lipoprotein, which
carry triacylglycerols, can be separated according to
their buoyant densities by centrifugation. They have
been classified into five groups of increasing density
but smaller size as chylomicrons, very low density
lipoproteins (VLDL), intermediate density lipopro-
teins (IDL), low density (LDL), and high density
lipoproteins (HDL) (Table 21-1 and Fig. 21-2). Each
lipoprotein particle contains one or more apolipopro-
teins (Table 21-2), whose sizes vary from the enormous
4536-residue apoB-100 to apoC-II and apoC-III, each of
which contains just 79 residues
7a
and the 57-residue
apoC-I.
7b
The larger lipoproteins are spherical micelles
containing a core of triacylglycerols and esters of
cholesterol surrounded by a 2- to 3-nm-thick layer
50 m
Ch 21 IND page - E 2/14/03, 11:34 AM 1181
1182 Chapter 21. Specific Aspects of Lipid Metabolism
consisting of phospholipid, free cholesterol, and the
apolipoprotein components.
8
The size of the lipopro-
tein particles also varies from a 200- to 500-nm diame-
ter for chylomicrons to as little as 5 nm for the smallest
HDL particles. The difference in volume is more
impressive. If, as has been estimated,
9
a 22-nm diame-
ter LDL particle contains about 2000 cholesterol and
cholesteryl ester molecules and 800 phospholipids, a
small HDL particle of 7-nm diameter will have room
for only about 60 molecules of cholesterol and 90 of
phospholipid, while a chylomicron may carry 10
million molecules of triacylglycerol. HDL particles are
quite heterogeneous. As is indicated in Table 21-1,
they are sometimes dividing into HDL2 and HDL3
density groups. In addition, there is a pre-HDL with
lower phospholipid content and discoid forms low in
cholesterol. Models of a reconstituted lipoprotein disc
contain two molecules of apoA-I and ~160 phosphati-
dylcholines that form a bilayer core.
10 10b
Each apolipoprotein has one or more distinct
functions. The apoB proteins probably stabilize the
lipoprotein micelles. In addition, apoB-100 is essential
to recognition of LDL by its receptors. The 79-residue
apoC-II has a specific function of activating the lipo-
protein lipase that hydrolyses the triacylglycerols of
chylomicrons and VLDL. Lack of either C-II or the
lipase results in a very high level of triacylglycerols in
the blood.
11
The large apolipoprotein B-100 is synthesized in
the liver and is a principal component of VLDL, IDL,
and LDL. It is the sole protein in LDL, accounting for
nearly 20% of the mass of LDL particles. Partly be-
cause of its insolubility in water, its detailed structure
is uncertain. If it were all coiled into an helix, it
would be 680 nm long and could encircle the LDL
particle nearly 10 times! While the true structure of
apoB-100 is unknown, it is thought to be extended and
to span at least a hemisphere of the LDL surface.
12
It
consists of at least five domains. Sixteen cysteines are
present in the first 25 residues at the N terminus,
forming a crosslinked high-cysteine region. There are
also 16 N-glycosylated sites. Domain IV (residues
3071 4011) is thought to contain the site that binds to
its specific receptor, the LDL receptor.
12
Heterogeneity
in the amide I band of the infrared absorption spec-
trum (Fig. 23-3) suggests that about 24% is helix,
23% sheet, and that a large fraction consists of turns,
and unordered and extended peptide structures.
13
In intestinal epithelial cells the same apoB gene
that is used to synthesize apoB-100 in the liver is used
to make the shorter apoB-48 (48%) protein. This is
accomplished in an unusual way that involves edit-
ing of the mRNA that is formed. Codon 2153 in the
mRNA for the protein is CAA, encoding glutamine.
However, the cytosine of the triplet is acted on by a
deaminase, an editing enzyme, to form UAA, a chain
termination codon.
14,15
A third form of apoB is found
in lipoprotein(a) (Lpa). This LDL-like particle con-
tains apoB-100 to which is covalently attached by a
single disulfide linkage (probably to Cys 3734 of apoB-
100) a second protein, apo(a). The latter consists
largely of a chain of from 11 to over 50 kringle do-
mains resembling the 78-residue kringle-4 of plasmi-
nogen (see Fig. 7-30C)
16 19a
as well as a protease
domain.
20
This additional chain may cause tighter
binding to LDL receptors and may cause lipoprotein(a)
to displace plasminogen from cell surface receptors.
21
The amount of Lp(a) varies over 1000-fold among
individuals and is genetically determined. The num-
ber of kringle domains also varies.
17
Although the
presence of high Lp(a) is associated with a high risk of
atherosclerosis and stroke,
21a
many healthy 100-year
olds also have high serum Lp(a).
22
Apolipoprotein A-I is the primary protein compo-
nent of HDL.
23 25b
Most of the 243 residues consist of
a nearly continuous amphipathic helix with kinks at
regularly spaced proline residues.
26 28
Two disulfide-
linked ApoA-I molecules may form a belt that encir-
cles the discoid lipoprotein.
25b
ApoA-II is the second
major HDL protein, but no clearly specialized function
has been identified.
29,30
ApoA-I, II, and IV, apoC-I, II,
and III, and apoE all have multiple repeats of 22 amino
acids with sequences that suggest amphipathic helices.
The 391-residue ApoA-IV has 13 tandem 22-residue
repeats. Proline and glycine are present in intervening
hinge regions.
23
This may enable these proteins to
spread over and penetrate the surfaces of the lipopro-
tein micelles. Most of these proteins are encoded by a
related multigene family.
7,30a
The 299-residue apolipoprotein E plays a key role
in metabolism of both triacylglycerols and cholesterol.
Like apoB-100 it binds to cell surface receptors.
31 33a
Absence of functional apoE leads to elevated plasma
triacylglycerol and cholesterol, a problem that is con-
sidered in Chapter 22, Section D. The N-terminal
domain, from residues 23 to 164, forms a 6.5 nm-long
four-helix bundle, which binds to the LDL receptors.
32
There are three common isoforms of alipoprotein E
(apoE2, apoE3, and apoE4). ApoE3 is most
common.
33b
The presence of apoE4 is associated with
an increased risk of Alzheimer disease (Chapter 30).
The major lipoproteins of insect hemolymph, the
lipophorins, transport diacylglycerols. The apolipo-
phorins have molecular masses of ~ 250, 80, and some-
times 18 kDa.
34 37a
The three-dimensional structure of
a small 166-residue lipophorin (apolipophorin-III) is
that of a four-helix bundle. It has been suggested that
it may partially unfold into an extended form, whose
amphipathic helices may bind to a phospholipid sur-
face of the lipid micelle of the lipophorin.
35
A similar
behavior may be involved in binding of mammalian
apolipoproteins. Four-helix lipid-binding proteins
have also been isolated from plants.
38
See also Box 21-
A. Specialized lipoproteins known as lipovitellins
Ch 21 IND page - E 2/14/03, 11:34 AM 1182
1183
TABLE 21-1
Classes of Lipoprotein Particles
a
Data from Havel, R. J., and Kane, J. P. (1995) in The Metabolic and Molecular Bases of Inherited Disease, 7th ed., Vol. II (Scriver, C. R., Beaudet, A.
L., Sly, W. S., and Valle, D., eds), pp. 1841 1852, McGraw-Hill, New York.
Chylomicrons 75 1200 0.93 2 7 2 3 86
VLDL 30 80 0.93 1.006 8 18 7 12 55
IDL 25 35 1.006 1.019 19 19 9 29 23
LDL 18 25 1.019 1.063 22 22 8 42 6
HDL2 912 1.063 1.125 40 33 5 17 5
HDL3 59 1.125 1.21 45 35 4 13 3
Lp (a); slow 25 30 1.04 1.09
pre-
Class Composition (weight percent)
a
Diameter
(nm)
Surface components Core lipids
Protein Phospholipid Cholesterol Cholesteryl
esters
Triacylglycerol Density
(g/ml)
These are averages and there is considerable variation.
TABLE 21-2
Properties of Major Plasma Apolipoproteins
A-I 243 29 Major HDL protein
A-II 17.4 Liver and intestine
A-IV 376 44.5
B-100 4536 513 Liver VLDL formation; ligand for LDL
receptor
B-48 2152 241 Intestine Chylomicron formation ligand
for liver chylomicron receptor
C-I 57 6.6
C-II 79 8.9 Liver Cofactor for lipoprotein lipase
C-III 79 8.8
D 31 Many tissues A lipocalin
E 299 34 Liver, VLDL Ligand for LDL receptor
(a) Variable Ligand for liver chylomicron
receptor
Designation No. residues Mass Source Function
(kDa)
A. Digestion, Synthesis, and Distribution of Triacylglycerols in the Human Body
Ch 21 IND page - E 2/14/03, 11:34 AM 1183
1184 Chapter 21. Specific Aspects of Lipid Metabolism
store phospholipid in eggs whether from nematodes,
frogs, or chickens.
39
There is some sequence similarity
to that of human apolipoprotein B-100.
2. Movement of Lipid Materials Between Cells
After the synthesis and release of chylomicrons
into the lymphatic circulation, various exchange pro-
cesses occur by which apolipoproteins, as well as
enzymes and other proteins, may be added or re-
moved. These very complex and incompletely under-
stood phenomena are presented in simplified form in
Fig. 21-1. Chylomicrons donate apolipoproteins of the
A and C families to HDL particles which, in turn,
donate apoE and may also return some apoC protein
to the chylomicrons.
Both chylomicrons and VLDL particles undergo
similar processes in the capillary blood vessels, where
their triacylglycerols are hydrolyzed to glycerol and
free fatty acids by lipoprotein lipase.
40 42a
This
enzyme requires for its activity the apolipoprotein C-II
which is present in the chylomicrons and VLDL parti-
cles. Lipoprotein lipase is also known as the clearing
Newly synthesized
triacylglycerols
Cholesterol
Phospholipids
Endocytosis
Excess
cholesterol
Bile salts
Cholesterol
Chylomicron
remnant
Cholesterol
Triacyl-
glycerols
Intestinal
mucosal cell
Triacylglycerol
Phospholipid
ApoB-48
ApoA-I,A-II
VLDL remnant
Fatty acids
LDL receptors
Cholesterol
Adipocytes and
other body cells
Excess
cholesterol
-HDL
Pre--
HDL
Glycerol
Free fatty acids and
albumin complexes
ApoE
HDL
ApoC
2 Fatty acids
Monoacylglycerol
+
Dietary
lipids
ApoB-48
ApoE
Liver cells
(hepatocytes)
Intestinal
lumen
ApoB-100
LDL
Chylomicrons
Free
phospholipids
Capillary
endothelial
cell
Lymphatic
circulation
Chylomicrons
ApoA
ApoC
Blood
VLDL
IDL
ApoB-100
ApoE
ApoC-I,II,III
ApoB-48
ApoE
ApoC-I,II,III
ApoA-I,II
ApoB-48
ApoA-I,II
ApoA-I,II
ApoC-I,II,III
ApoE
Lipoprotein
lipase
Lipoprotein
lipase
ApoB-100
ApoE
Pancreatic
lipase
Apolipoproteins
To
liver
ApoA-I
LCAT
CETP
Figure 21-1 Movement of triacylglycerols from liver and intestine to body cells and lipid carriers of blood. VLDL; very low
density lipoprotein which contains triacylglycerols, phospholipids, cholesterol, and apolipoproteins B, and C. IDL; intermedi-
ate density lipoproteins found in human plasma. LDL; low density lipoproteins which have lost most of their triacylglycerols.
ApoB-100, etc., are apolipoproteins listed in Table 21-2. LCAT, lecithin: cholesterol acyltransferase; CETP, cholesteryl ester
transfer protein (see Chapter 22).
Ch 21 IND page - E 2/14/03, 11:34 AM 1184
1185 B. The Biosynthesis of Fatty Acids and Their Esters
phorylation and moves from the cytoplasm to the
surfaces of lipid droplets in response to catechola-
mines and other lipolytic hormones. Fatty acids are a
major fuel for aerobic cells. Their conversion to acyl-
CoA derivative and oxidation to CO
2
by beta oxidation
(Fig. 17-1) and other pathways are discussed in
Chapter 17 (pp. 939 950).
B. The Biosynthesis of Fatty Acids and Their
Esters
The synthesis of fatty acids two carbon atoms at a
time from acetyl-CoA has been considered in Chapter
17 and is outlined in Fig. 17-12. In this pathway,
which resembles the oxidation sequence in reverse,
the products are saturated fatty acids with an even
number of carbon atoms as shown in Fig. 21-2. In this
section, we will consider some of the factors that lead
to variations in the chain lengths and types of fatty
acids.
1. Fatty Acid Synthases
Both bacteria and plants have separate enzymes
that catalyze the individual steps in the biosynthetic
sequence (Fig. 17-12). The fatty acyl group grows
while attached to the small acyl carrier protein
(ACP).
54 58
Control of the process is provided, in part,
by the existence of isoenzyme forms. For example, in
E. coli there are three different -oxoacyl-ACP synthas-
es. They carry out the transfer of any acyl primer from
ACP to the enzyme, decarboxylate malonyl-ACP, and
carry out the Claisen condensation (steps b, e, and f in
Eq. 17-12)
58a e
One of the isoenzymes is specialized
for the initial elongation of acetyl-ACP and also pro-
vides feedback regulation.
58c
The other two function
specifically in synthesis of unsaturated fatty acids.
In a few bacteria and protozoa and in higher
animals the fatty acid synthase consists of only one or
two multifunctional proteins. That from animal tis-
sues contains six enzymes and an acyl carrier protein
(ACP) domain as well. The human enzyme contains
2504 amino acid residues organized as a series of
functional domains.
59 59b
Pairs of the 272-kDa chains
associate to form 544-kDa dimers. The complex pro-
tein may have arisen via an evolutionary process
involving fusion of formerly separate genes.
60
The
enzyme contains an ACP-like site with a bound
4'-phosphopantetheine near the C terminus as well as
a cysteine side chain near the N terminus in the second
acylation site. Since the two SH groups can be
crosslinked by dibromopropanone,
61,62
an antiparallel
linear arrangement of the two chains was proposed.
6365
Locations of the six enzymatic activities in each chain
are indicated on page 1187. According to this picture,
factor because it clears the milky chylomicron-con-
taining lymph. It is secreted by adipocytes and other
cells and becomes attached to heparan sulfate proteg-
lycans on surfaces of capillary endothelial cells, a
major site of its action.
43
Hereditary absence of func-
tional lipoprotein lipase causes chylomicronemia, a
massive buildup of chylomicrons in plasma.
41,44
The
condition does not cause atherosclerosis but may lead
to pancreatitis if not treated. Restriction of dietary fat
to 20 g / day or less usually prevents problems. Natu-
rally occurring mutations of lipoprotein lipase involv-
ing both the aspartate of the catalytic triad (p. 635)
45
and the flexible loop that covers the active site
46
have
been discovered.
Both lipoprotein lipase and the less well under-
stood hepatic lipase are related structurally to pan-
creatic lipase.
42,42b
In addition to hydrolysis of the
triacylglycerols, the uptake of materials from lipopro-
teins probably involves shedding of intact phospholip-
ids, perhaps as liposome-like particles.
40
The free fatty acids and glycerol are taken up by
mammalian tissue cells leaving the cholesterol and
some of the phospholipids of the VLDL particles as
LDL. In humans intermediate density lipoproteins
(IDL) are formed initially, but some are converted to
LDL later. Both LDL particles and the shrunken chy-
lomicron remnants and VLDL remnants are taken
up by endocytosis in coated pits and are degraded by
body cells, principally of the liver.
47,48
The best
known of these receptors is the 839-residue LDL
receptor, which has a specific affinity for ApoB-100.
The related VLDL receptor (apoE receptor) has a
higher affinity for apoE
48 50
and may function in up-
take of both VLDL and chylomicron remnants. The
LDL receptor-related protein functions as a third
lipoprotein receptor.
51
In addition, a series of scaven-
ger receptors, found in abundance in macrophages,
take up oxidized lipoproteins and other materials.
51,52
Scavenger receptor B1 (SR-B1), which is also found in
liver cells, is involved in uptake of cholesterol from
HDL particles by hepatocytes
53
(see also Chapter 22).
Liver cells, and other cells as well, contain lipocalins
and fatty acid binding proteins (Box 21-A) that help
to carry these relatively insoluble acids to their desti-
nations within the cells. Serum albumin (Box 2-A) is
also a major carrier of free fatty acids.
53a
Within the
adipocytes the fatty acids are reconverted to triacylg-
lycerols. The low density (LDL) and high density
(HDL) lipoproteins are involved primarily in trans-
porting cholesterol to and from cells, a topic that is
discussed in Chapter 22, Section D,2.
Fatty acids are carried to tissues for use in synthe-
sis of triacylglycerols, phospholipids, and other
membrane lipids. The mobilization of fatty acids from
triacylglycerol stores and from cholesterol esters de-
pends upon hormone-sensitive lipase (p. 635).
53b, 53c
This enzyme is activated by cAMP-dependent phos-
Ch 21 IND page - E 2/14/03, 11:34 AM 1185
1186 Chapter 21. Specific Aspects of Lipid Metabolism
BOX 21-A LIPOCALINS, FATTY ACID-BINDING PROTEINS, AND LIPOPHORINS
Small hydrophobic molecules, which might
easily bind in biologically undesirable ways, are
chaperoned in animals, plants, and bacteria by
binding proteins that provide hydrophobic cavities
or crevices appropriate for holding these molecules
in readily releasable forms. The lipocalins, most of
which are extracellular proteins, have a conserved
structural motif consisting of an 8-stranded barrel
arranged as two stacked orthogonal sheets with a C-
terminal helix that blocks one end. The other end
is able to open and allow a small hydrophobic
molecule to bind in the internal cavity
a c
(see fig-
ure). Only three short amino acid sequences are
conserved within a large family of lipocalins
b,d
which includes plasma retinol-binding protein,
e
mammalian odorant-binding proteins,
f
-lactal-
bumin, apolipoprotein D,
a
and the blue biliverdin-
binding protein insecticyanin of insect
hemolymph.
g,h
Most lipocalins are soluble, but
some such as the plasma 1-microglobulin,
i
which
plays a role in the immune system (Chapter 31),
have additional functions that require them to bind
to other proteins or to cell surfaces.
j
The gelati-
nase-associated lipocalin of human neutrophils
binds bacterially derived N-formylpeptides that act
as chemotactic agents (Chapter 19) and induces
release of materials from intracellular granules.
c
A
few lipocalins have enzymatic activity. For exam-
ple, prostaglandin D synthase is both an enzyme
and a carrier of bile pigments and thyroid
hormones.
k
Most lipocalins have been found in
higher animals, but at least a few bacterial proteins
belong to the family.
d
One is the 77-residue E. coli
outer membrane lipoprotein.
a
A related family of proteins are represented by
fatty acid-binding proteins
l o
and by the intracel-
lular retinol- and retinoic acid-binding proteins
(see also Box 22-A).
p
These are 10-stranded antipar-
allel -barrels with two helices blocking an end (see
Figure).
A third group of lipid-binding proteins have a
four-helix bundle structure. They include the insect
lipophorins, which transport diacylglycerols in the
hemolymph (see main text), and nonspecific lipid
carriers of green plants.
q
An 87-residue four-helix
protein with a more open structure binds acyl-
coenzyme A molecules in liver.
r
A small 98-residue sterol-binding protein from
Structure of a crystalline fatty acid-
binding protein from liver with two
molecules of bound oleate (dark
rods). The lower molecule is more
deeply embedded in the protein and
more tightly bound than the second
molecule, which is closer to the outer
surface of the protein. Semitranspar-
ent grey marks the solvent-accessible
surface of the binding cavity. An
unknown molecule, perhaps butanoic
acid (as modeled), binds also at top of
the protein. See Thompson et al.
n
Courtesy of Leonard Banaszak.
NH
3
+
NH
3
+
COO
-
COO
-
Stereoscopic view of an -carbon
model of an insecticyanin sub-
unit with the bound biliverdin.
The N and C termini are labeled
NH
3
+
and COO

, respectively.
The positions of several amino
acid residues are indicated.
From Holden et al.
g
Courtesy of
Hazel Holden.
Ch 21 IND page - E 2/14/03, 11:34 AM 1186
1187
BOX 21-A (continued)
a
Bishop, R. E., Penfold, S. S., Frost, L. S., Hltje, J.-V., and
Weiner, J. H. (1995) J. Biol. Chem. 270, 2309723103
b
Flower, D. R., North, A. C. T., and Attwood, T. K. (1993) Protein
Sci. 2, 753761
c
Coles, M., Diercks, T., Muehlenweg, B., Bartsch, S., Zlzer, V.,
Tschesche, H., and Kessler, H. (1999) J. Mol. Biol. 289, 139157
d
Flower, D. R., Sansom, C.

You might also like